Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 217
Filtrer
1.
Methods Mol Biol ; 2845: 177-189, 2024.
Article de Anglais | MEDLINE | ID: mdl-39115666

RÉSUMÉ

Ferritinophagy is a selective form of autophagy in which ferritin, the primary intracellular iron storage protein complex, is targeted by NCOA4 (Nuclear receptor coactivator 4) to the lysosome for degradation. NCOA4-mediated ferritinophagy plays a crucial role in cellular iron metabolism, influencing iron homeostasis, heme synthesis, mitochondrial respiratory function, and ferroptosis, an iron-dependent form of cell death. Targeting ferritinophagy has emerged as a potential anticancer therapeutic strategy. In this context, we provide a flowchart of the procedures and accompanying protocols for monitoring ferritinophagic flux.


Sujet(s)
Autophagie , Ferritines , Coactivateurs de récepteurs nucléaires , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Ferritines/métabolisme , Humains , Fer/métabolisme , Lysosomes/métabolisme , Animaux
2.
Fish Shellfish Immunol ; 151: 109745, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38960105

RÉSUMÉ

Iron homeostasis is vital for the host's defense against pathogenic invasion and the ferritinophagy is a crucial mechanism in maintaining intracellular iron homeostasis by facilitating the degradation and recycling of stored iron. The nuclear receptor coactivator 4 (NCOA4) serves as a ferritinophagy receptor, facilitating the binding and delivery of ferritin to the autophagosome and lysosome. However, NCOA4 of the sea cucumber Apostichopus japonicus (AjNCOA4) has not been reported until now. In this study, we identified and characterized AjNCOA4 in A. japonicus. This gene encodes a polypeptide containing 597 amino acids with an open reading frame of 1794 bp. The inferred amino acid sequence of AjNCOA4 comprises an ARA70 domain. Furthermore, a multiple sequence alignment demonstrated varying degrees of sequence homology between AjNCOA4 from A. japonicus and other NCOA4 orthologs. The phylogenetic tree of NCOA4 correlates with the established timeline of metazoan evolution. Expression analysis revealed that AjNCOA4 is expressed in all tested tissues, including the body wall, muscle, intestine, respiratory tree, and coelomocytes. Following challenge with Vibrio splendidus, the coelomocytes exhibited a significant increase in AjNCOA4 mRNA levels, peaking at 24 h. We successfully obtained recombinant AjNCOA4 protein through prokaryotic expression and prepared a specific polyclonal antibody. Immunofluorescence and co-immunoprecipitation experiments demonstrated an interaction between AjNCOA4 and AjFerritin in coelomocytes. RNA interference-mediated knockdown of AjNCOA4 expression resulted in elevated iron ion levels in coelomocytes. Bacterial stimulation enhanced ferritinophagy in coelomocytes, while knockdown of AjNCOA4 reduced the occurrence of ferritinophagy. These findings suggest that AjNCOA4 modulates ferritinophagy induced by V. splendidus in coelomocytes of A. japonicus.


Sujet(s)
Séquence d'acides aminés , Ferritines , Coactivateurs de récepteurs nucléaires , Phylogenèse , Alignement de séquences , Stichopus , Vibrio , Animaux , Vibrio/physiologie , Stichopus/immunologie , Stichopus/génétique , Stichopus/microbiologie , Coactivateurs de récepteurs nucléaires/génétique , Coactivateurs de récepteurs nucléaires/immunologie , Ferritines/génétique , Ferritines/immunologie , Ferritines/métabolisme , Immunité innée/génétique , Régulation de l'expression des gènes/immunologie , Analyse de profil d'expression de gènes , Autophagie , Séquence nucléotidique
3.
Int Immunopharmacol ; 138: 112605, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38963979

RÉSUMÉ

Ferroptosis, characterized by the accumulation of reactive oxygen species and lipid peroxidation, is involved in various cardiovascular diseases. (Pro)renin receptor (PRR) in performs as ligands in the autophagic process, and its function in diabetic cardiomyopathy (DCM) is not fully understood. We investigated whether PRR promotes ferroptosis through the nuclear receptor coactivator 4 (NCOA 4)-mediated ferritinophagy pathway and thus contributes to DCM. We first established a mouse model of DCM with downregulated and upregulated PRR expression and used a ferroptosis inhibitor. Myocardial inflammation and fibrosis levels were then measured, cardiac function and ferroptosis-related indices were assessed. In vitro, neonatal rat ventricular primary cardiomyocytes were cultured with high glucose and transfected with recombinant adenoviruses knocking down or overexpressing the PRR, along with a ferroptosis inhibitor and small interfering RNA for the ferritinophagy receptor, NCOA4. Ferroptosis levels were measured in vitro. The results showed that the knockdown of PRR not only alleviated cardiomyocyte ferroptosis in vivo but also mitigated the HG-induced ferroptosis in vitro. Moreover, administration of Fer-1 can inhibit HG-induced ferroptosis. NCOA4 knockdown blocked the effect of PRR on ferroptosis and improved cell survival. Our result indicated that inhibition of PRR and NCOA4 expression provides a new therapeutic strategy for the treatment of DCM. The effect of PRR on the pathological process of DCM in mice may be in promoting cardiomyocyte ferroptosis through the NCOA 4-mediated ferritinophagy pathway.


Sujet(s)
Cardiomyopathies diabétiques , Ferroptose , Myocytes cardiaques , Coactivateurs de récepteurs nucléaires , Récepteur de la prorénine , Animaux , Souris , Rats , Autophagie , Cellules cultivées , Cardiomyopathies diabétiques/anatomopathologie , Cardiomyopathies diabétiques/métabolisme , Cardiomyopathies diabétiques/génétique , Modèles animaux de maladie humaine , Régulation négative , Ferritines/métabolisme , Souris de lignée C57BL , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Récepteur de la prorénine/génétique , Récepteur de la prorénine/métabolisme , Transduction du signal
4.
Cell Commun Signal ; 22(1): 376, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39061070

RÉSUMÉ

Acute kidney injury (AKI) is closely related to lysosomal dysfunction and ferroptosis in renal tubular epithelial cells (TECs), for which effective treatments are urgently needed. Although selenium nanoparticles (SeNPs) have emerged as promising candidates for AKI therapy, their underlying mechanisms have not been fully elucidated. Here, we investigated the effect of SeNPs on hypoxia/reoxygenation (H/R)-induced ferroptosis and lysosomal dysfunction in TECs in vitro and evaluated their efficacy in a murine model of ischemia/reperfusion (I/R)-AKI. We observed that H/R-induced ferroptosis was accompanied by lysosomal Fe2+ accumulation and dysfunction in TECs, which was ameliorated by SeNPs administration. Furthermore, SeNPs protected C57BL/6 mice against I/R-induced inflammation and ferroptosis. Mechanistically, we found that lysosomal Fe2+ accumulation and ferroptosis were associated with the excessive activation of NCOA4-mediated ferritinophagy, a process mitigated by SeNPs through the upregulation of X-box binding protein 1 (XBP1). Downregulation of XBP1 promoted ferritinophagy and partially counteracted the protective effects of SeNPs on ferroptosis inhibition in TECs. Overall, our findings revealed a novel role for SeNPs in modulating ferritinophagy, thereby improving lysosomal function and attenuating ferroptosis of TECs in I/R-AKI. These results provide evidence for the potential application of SeNPs as therapeutic agents for the prevention and treatment of AKI.


Sujet(s)
Ferroptose , Nanoparticules , Lésion d'ischémie-reperfusion , Sélénium , Protéine-1 liant la boite X , Animaux , Humains , Mâle , Souris , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/traitement médicamenteux , Autophagie/effets des médicaments et des substances chimiques , Ferritines/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Lysosomes/métabolisme , Lysosomes/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Nanoparticules/composition chimique , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/anatomopathologie , Sélénium/pharmacologie , Sélénium/administration et posologie , Transduction du signal/effets des médicaments et des substances chimiques , Protéine-1 liant la boite X/métabolisme , Protéine-1 liant la boite X/génétique
5.
Commun Biol ; 7(1): 732, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38886572

RÉSUMÉ

Ferroptosis is a unique iron-dependent form of non-apoptotic cell death characterized by devastating lipid peroxidation. Whilst growing evidence suggests that ferroptosis is a type of autophagy-dependent cell death, the underlying molecular mechanisms regulating ferroptosis are largely unknown. In this study, through an unbiased RNA-sequencing screening, we demonstrate the activation of a multi-faceted tumor-suppressor protein Par-4/PAWR during ferroptosis. Functional studies reveal that genetic depletion of Par-4 effectively blocks ferroptosis, whereas Par-4 overexpression sensitizes cells to undergo ferroptosis. More importantly, we have determined that Par-4-triggered ferroptosis is mechanistically driven by the autophagic machinery. Upregulation of Par-4 promotes activation of ferritinophagy (autophagic degradation of ferritin) via the nuclear receptor co-activator 4 (NCOA4), resulting in excessive release of free labile iron and, hence, enhanced lipid peroxidation and ferroptosis. Inhibition of Par-4 dramatically suppresses the NCOA4-mediated ferritinophagy signaling axis. Our results also establish that Par-4 activation positively correlates with reactive oxygen species (ROS) production, which is critical for ferritinophagy-mediated ferroptosis. Furthermore, Par-4 knockdown effectively blocked ferroptosis-mediated tumor suppression in the mouse xenograft models. Collectively, these findings reveal that Par-4 has a crucial role in ferroptosis, which could be further exploited for cancer therapy.


Sujet(s)
Autophagie , Ferroptose , Coactivateurs de récepteurs nucléaires , Espèces réactives de l'oxygène , Ferroptose/génétique , Humains , Animaux , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Souris , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire tumorale , Peroxydation lipidique , Fer/métabolisme , Protéines régulatrices de l'apoptose/métabolisme , Protéines régulatrices de l'apoptose/génétique , Transduction du signal
6.
Oncogene ; 43(26): 2000-2014, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38744953

RÉSUMÉ

Esophageal squamous cell carcinoma (ESCC) is a prevalent malignancy of the digestive system. Hypoxia is a crucial player in tumor ferroptosis resistance. However, the molecular mechanism of hypoxia-mediated ferroptosis resistance in ESCC remains unclear. Here, USP2 expression was decreased in ESCC cell lines subjected to hypoxia treatment and was lowly expressed in clinical ESCC specimens. Ubiquitin-specific protease 2 (USP2) depletion facilitated cell growth, which was blocked in USP2-overexpressing cells. Moreover, USP2 silencing enhanced the iron ion concentration and lipid peroxidation accumulation as well as suppressed ferroptosis, while upregulating USP2 promoted ferroptotic cell death in ESCC cells. Furthermore, knockout of USP2 in ESCC models discloses the essential role of USP2 in promoting ESCC tumorigenesis and inhibiting ferroptosis. In contrast, overexpression of USP2 contributes to antitumor effect and ferroptosis events in vivo. Specifically, USP2 stably bound to and suppressed the degradation of nuclear receptor coactivator 4 (NCOA4) by eliminating the Lys48-linked chain, which in turn triggered ferritinophagy and ferroptosis in ESCC cells. Our findings suggest that USP2 plays a crucial role in iron metabolism and ferroptosis and that the USP2/NCOA4 axis is a promising therapeutic target for the management of ESCC.


Sujet(s)
Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Ferroptose , Ubiquitin thiolesterase , Humains , Ferroptose/génétique , Carcinome épidermoïde de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/métabolisme , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/métabolisme , Tumeurs de l'oesophage/génétique , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Animaux , Souris , Lignée cellulaire tumorale , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Régulation de l'expression des gènes tumoraux , Ferritines/métabolisme , Ferritines/génétique , Souris nude , Autophagie/génétique , Hypoxie/métabolisme , Prolifération cellulaire/génétique , Mâle
7.
Redox Biol ; 73: 103190, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38744191

RÉSUMÉ

Parkinson's disease (PD) poses a significant challenge in neurodegenerative disorders, characterized by the progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc). The intricate mechanisms orchestrating DA neurodegeneration in PD are not fully understood, necessitating the exploration of innovative therapeutic approaches. Recent studies have implicated ferroptosis as a major contributor to the loss of DA neurons, revealing a complex interplay between iron accumulation and neurodegeneration. However, the sophisticated nature of this process challenges the conventional belief that mere iron removal could effectively prevent DA neuronal ferroptosis. Here, we report JWA, alternatively referred to as ARL6IP5, as a negative regulator of ferroptosis, capable of ameliorating DA neuronal loss in the context of PD. In this study, synchronized expression patterns of JWA and tyrosine hydroxylase (TH) in PD patients and mice were observed, underscoring the importance of JWA for DA neuronal survival. Screening of ferroptosis-related genes unraveled the engagement of iron metabolism in the JWA-dependent inhibition of DA neuronal ferroptosis. Genetic manipulation of JWA provided compelling evidence linking its neuroprotective effects to the attenuation of NCOA4-mediated ferritinophagy. Molecular docking, co-immunoprecipitation, and immunofluorescence studies confirmed that JWA mitigated DA neuronal ferroptosis by occupying the ferritin binding site of NCOA4. Moreover, the JWA-activating compound, JAC4, demonstrated promising neuroprotective effects in cellular and animal PD models by elevating JWA expression, offering a potential avenue for neuroprotection in PD. Collectively, our work establishes JWA as a novel regulator of ferritinophagy, presenting a promising therapeutic target for addressing DA neuronal ferroptosis in PD.


Sujet(s)
Neurones dopaminergiques , Ferritines , Ferroptose , Coactivateurs de récepteurs nucléaires , Maladie de Parkinson , Maladie de Parkinson/métabolisme , Maladie de Parkinson/anatomopathologie , Maladie de Parkinson/génétique , Neurones dopaminergiques/métabolisme , Neurones dopaminergiques/anatomopathologie , Animaux , Souris , Humains , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Ferritines/métabolisme , Ferritines/génétique , Fer/métabolisme , Modèles animaux de maladie humaine , Liaison aux protéines , Autophagie , Mâle
8.
Toxicology ; 505: 153831, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38768701

RÉSUMÉ

Cadmium (Cd) is a common pollutant with reproductive toxicity. Our previous study revealed that Cd triggered spermatogonia ferroptosis. However, the underlying mechanisms remain unclear. Nuclear receptor coactivator 4 (NCOA4) mediates ferritinophagy and specific degradation of ferritin through lysosomes, resulting in the release of ferrous ions. Excessive autophagy can lead to ferroptosis. This study investigated the role of autophagy in Cd-triggered ferroptosis using GC-1 spermatogonial (spg) cells which exposed to CdCl2 (5 µM, 10 µM, or 20 µM) for 24 without/with CQ. The cells which transfected with Ncoa4-siRNA were used to explore the role of NCOA4-mediated ferritinophagy in Cd-triggered ferroptosis. The results revealed that Cd caused mitochondrial swelling, rupture of cristae, and vacuolar-like changes. The Cd-treated cells exhibited more autophagosomes. Simultaneously, Cd increased intracellular iron, reactive oxygen species, and malondialdehyde concentrations while decreasing glutathione content and Superoxide Dismutase-2 activity. Moreover, Cd upregulated mRNA levels of ferritinophagy-associated genes (Ncoa4, Lc3b and Fth1), as well as enhanced protein expression of NCOA4, LC3B, and FTH1. While Cd decreased the mRNA and protein expression of p62/SQSTM1. These results showed that Cd caused ferritinophagy and ferroptosis. The use of chloroquine to inhibit autophagy ameliorated Cd-induced iron overload and ferroptosis. Moreover, Ncoa4 knockdown in spermatogonia significantly reduced intracellular iron concentration and alleviated Cd-triggered ferroptosis. In conclusion, our findings demonstrate that Cd activates the ferritinophagy pathway mediated by NCOA4, resulting in iron accumulation through ferritin degradation. This causes oxidative stress, ultimately initiating ferroptosis in spermatogonia. Our results may provide new perspectives and potential strategies for preventing and treating Cd-induced reproductive toxicity.


Sujet(s)
Autophagie , Cadmium , Ferritines , Ferroptose , Coactivateurs de récepteurs nucléaires , Spermatogonies , Ferroptose/effets des médicaments et des substances chimiques , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Mâle , Spermatogonies/effets des médicaments et des substances chimiques , Spermatogonies/métabolisme , Ferritines/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Cadmium/toxicité , Animaux , Lignée cellulaire , Souris , Espèces réactives de l'oxygène/métabolisme
9.
Nat Commun ; 15(1): 4195, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38760351

RÉSUMÉ

Osimertinib (Osi) is a widely used epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI). However, the emergence of resistance is inevitable, partly due to the gradual evolution of adaptive resistant cells during initial treatment. Here, we find that Osi treatment rapidly triggers adaptive resistance in tumor cells. Metabolomics analysis reveals a significant enhancement of oxidative phosphorylation (OXPHOS) in Osi adaptive-resistant cells. Mechanically, Osi treatment induces an elevation of NCOA4, a key protein of ferritinophagy, which maintains the synthesis of iron-sulfur cluster (ISC) proteins of electron transport chain and OXPHOS. Additionally, active ISC protein synthesis in adaptive-resistant cells significantly increases the sensitivity to copper ions. Combining Osi with elesclomol, a copper ion ionophore, significantly increases the efficacy of Osi, with no additional toxicity. Altogether, this study reveals the mechanisms of NCOA4-mediated ferritinophagy in Osi adaptive resistance and introduces a promising new therapy of combining copper ionophores to improve its initial efficacy.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Récepteurs ErbB , Ferritines , Tumeurs du poumon , Animaux , Humains , Souris , Acrylamides/pharmacologie , Dérivés de l'aniline/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Cuivre/métabolisme , Récepteurs ErbB/antagonistes et inhibiteurs , Ferritines/métabolisme , Indoles/pharmacologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Souris nude , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Phosphorylation oxydative/effets des médicaments et des substances chimiques , Pyrimidines/pharmacologie , /pharmacologie
10.
Nat Commun ; 15(1): 3802, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38714719

RÉSUMÉ

The interaction between nuclear receptor coactivator 4 (NCOA4) and the iron storage protein ferritin is a crucial component of cellular iron homeostasis. The binding of NCOA4 to the FTH1 subunits of ferritin initiates ferritinophagy-a ferritin-specific autophagic pathway leading to the release of the iron stored inside ferritin. The dysregulation of NCOA4 is associated with several diseases, including neurodegenerative disorders and cancer, highlighting the NCOA4-ferritin interface as a prime target for drug development. Here, we present the cryo-EM structure of the NCOA4-FTH1 interface, resolving 16 amino acids of NCOA4 that are crucial for the interaction. The characterization of mutants, designed to modulate the NCOA4-FTH1 interaction, is used to validate the significance of the different features of the binding site. Our results explain the role of the large solvent-exposed hydrophobic patch found on the surface of FTH1 and pave the way for the rational development of ferritinophagy modulators.


Sujet(s)
Cryomicroscopie électronique , Ferritines , Coactivateurs de récepteurs nucléaires , Ferritines/métabolisme , Ferritines/composition chimique , Ferritines/génétique , Humains , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/composition chimique , Coactivateurs de récepteurs nucléaires/génétique , Liaison aux protéines , Sites de fixation , Fer/métabolisme , Autophagie , Modèles moléculaires , Cellules HEK293 , Oxidoreductases/métabolisme , Oxidoreductases/composition chimique , Oxidoreductases/génétique , Protéolyse , Mutation
11.
Int Immunopharmacol ; 133: 112155, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38688134

RÉSUMÉ

BACKGROUND: Ferroptosis is an iron-dependent and cystathione-non-dependent non-apoptotic cell death characterized by elevated intracellular free iron levels and reduced antioxidant capacity, leading to the accumulation of lipid peroxides. Nuclear receptor coactivator 4 (NCOA4) mediates ferritinophagy, increasing labile iron levels, which can result in oxidative damage. However, the specific mechanism of NCOA4-mediated ferritinophagy in intestinal ischemia-reperfusion and the underlying mechanisms have not been reported in detail. OBJECT: 1. To investigate the role of NCOA4 in ferroptosis of intestinal epithelial cells induced by II/R injury in mouse. 2. To investigate the mechanism of action of NCOA4-induced ferroptosis. METHODS: 1. Construct a mouse II/R injury model and detect ferroptosis related markers such as HE staining, immunohistochemistry, ELISA, and WB methods. 2. Detect expression of NCOA4 in the intestine of mouse with II/R injury model and analyze its correlation with intestinal ferroptosis in mouse with II/R injury model. 3. Construct an ischemia-reperfusion model at the cellular level through hypoxia and reoxygenation, and overexpress/knockdown NCOA4 to detect markers related to ferroptosis. Based on animal experimental results, analyze the correlation and mechanism of action between NCOA4 and intestinal epithelial ferroptosis induced by II/R injury in mouse. RESULTS: 1. Ferroptosis occurred in the intestinal epithelial cells of II/R-injured mouse, and the expression of critical factors of ferroptosis, ACSL4, MDA and 15-LOX, was significantly increased, while the levels of GPX4 and GSH were significantly decreased. 2. The expression of NCOA4 in the intestinal epithelium of mouse with II/R injure was significantly increased, the expression of ferritin was significantly decreased, and the level of free ferrous ions was significantly increased; the expression of autophagy-related proteins LC3 and Beclin-1 protein was increased, and the expression of P62 was decreased, and these changes were reversed by autophagy inhibitors. 3. Knockdown of NCOA4 at the cellular level resulted in increased ferritin expression and decreased ferroptosis, and CO-IP experiments suggested that NCOA4 can bind to ferritin, which suggests that NCOA4 most likely mediates ferritinophagy to induce ferroptosis. CONCLUSION: This thesis explored the role of NCOA4 in II/R injury in mice and the mechanism of action. The research results suggest that NCOA4 can mediate ferritinophagy to induce ferroptosis during II/R injury. This experiment reveals the pathological mechanism of II/R injury and provides some scientific basis for the development of drugs for the treatment of II/R injury based on the purpose of alleviating ferroptosis.


Sujet(s)
Ferroptose , Muqueuse intestinale , Coactivateurs de récepteurs nucléaires , Lésion d'ischémie-reperfusion , Animaux , Humains , Mâle , Souris , Modèles animaux de maladie humaine , Ferritines/métabolisme , Ferroptose/génétique , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Fer/métabolisme , Souris de lignée C57BL , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/anatomopathologie
12.
Aging (Albany NY) ; 16(9): 7683-7703, 2024 04 26.
Article de Anglais | MEDLINE | ID: mdl-38683121

RÉSUMÉ

Ferroptosis, an iron-triggered modality of cellular death, has been reported to closely relate to human aging progression and aging-related diseases. However, the involvement of ferroptosis in the development and maintenance of senescent cells still remains elusive. Here, we established a doxorubicin-induced senescent HSkM cell model and found that both iron accumulation and lipid peroxidation increase in senescent cells. Moreover, such iron overload in senescent cells has changed the expression panel of the ferroptosis-response proteins. Interestingly, the iron accumulation and lipid peroxidation does not trigger ferroptosis-induced cell death. Oppositely, senescent cells manifest resistance to the ferroptosis inducers, compared to the proliferating cells. To further investigate the mechanism of ferroptosis-resistance for senescent cells, we traced the iron flux in cell and found iron arrested in lysosome. Moreover, disruption of lysosome functions by chloroquine and LLOMe dramatically triggered the senescent cell death. Besides, the ferroitinophagy-related proteins FTH1/FTL and NCOA4 knockdown also increases the senescent cell death. Thus, we speculated that iron retardation in lysosome of senescent cells is the key mechanism for ferroptosis resistance. And the lysosome is a promising target for senolytic drugs to selectively clear senescent cells and alleviate the aging related diseases.


Sujet(s)
Vieillissement de la cellule , Ferroptose , Fer , Lysosomes , Ferroptose/effets des médicaments et des substances chimiques , Humains , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Vieillissement de la cellule/physiologie , Lysosomes/métabolisme , Lysosomes/effets des médicaments et des substances chimiques , Fer/métabolisme , Peroxydation lipidique/effets des médicaments et des substances chimiques , Doxorubicine/pharmacologie , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Lignée cellulaire , Chloroquine/pharmacologie , Surcharge en fer/métabolisme , Ferritines , Oxidoreductases
13.
Environ Pollut ; 349: 123958, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38621452

RÉSUMÉ

Exposure to environmental cadmium (Cd) is known to cause neuronal death and cognitive decline in humans. Ferroptosis, a novel iron-dependent type of regulated cell death, is involved in various neurological disorders. In the present study, Cd exposure triggered ferroptosis in the mouse hippocampus and in the HT22 murine hippocampal neuronal cell line, as indicated by significant increases in ferroptotic marker expression, intracellular iron levels, and lipid peroxidation. Interestingly, ferroptosis of hippocampal neurons in response to Cd exposure relied on the induction of autophagy since the suppression of autophagy by 3-methyladenine (3-MA) and chloroquine (CQ) substantially ameliorated Cd-induced ferroptosis. Furthermore, nuclear receptor coactivator 4 (NCOA4)-mediated degradation of ferritin was required for the Cd-induced ferroptosis of hippocampal neurons, demonstrating that NCOA4 knockdown decreased intracellular iron levels and lipid peroxidation and increased cell survival, following Cd exposure. Moreover, Cd-induced mitochondrial reactive oxygen species (mtROS) generation was essential for the ferritinophagy-mediated ferroptosis of hippocampal neurons. Importantly, pretreatment with the ferroptosis inhibitor ferrostatin-1 (Fer-1) effectively attenuated Cd-induced hippocampal neuronal death and cognitive impairment in mice. Taken together, these findings indicate that ferroptosis is a novel mechanism underlying Cd-induced neurotoxicity and cognitive impairment and that the mtROS-ferritinophagy axis modulates Cd-induced neuronal ferroptosis.


Sujet(s)
Cadmium , Dysfonctionnement cognitif , Ferroptose , Hippocampe , Neurones , Coactivateurs de récepteurs nucléaires , Espèces réactives de l'oxygène , Ferroptose/effets des médicaments et des substances chimiques , Animaux , Souris , Cadmium/toxicité , Neurones/effets des médicaments et des substances chimiques , Dysfonctionnement cognitif/induit chimiquement , Dysfonctionnement cognitif/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Espèces réactives de l'oxygène/métabolisme , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Ferritines/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mâle , Autophagie/effets des médicaments et des substances chimiques , Fer/métabolisme , Peroxydation lipidique/effets des médicaments et des substances chimiques , Lignée cellulaire , Souris de lignée C57BL
14.
Redox Biol ; 72: 103158, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38631121

RÉSUMÉ

Exposure to PM2.5 is correlated with cardiac remodeling, of which cardiac hypertrophy is one of the main clinical manifestations. Ferroptosis plays an important role in cardiac hypertrophy. However, the potential mechanism of PM2.5-induced cardiac hypertrophy through ferroptosis remains unclear. This study aimed to explore the molecular mechanism of cardiac hypertrophy caused by PM2.5 and the intervention role of MitoQ involved in this process. The results showed that PM2.5 could induce cardiac hypertrophy and dysfunction in mice. Meanwhile, the characteristics of ferroptosis were observed, such as iron homeostasis imbalance, lipid peroxidation, mitochondrial damage and abnormal expression of key molecules. MitoQ treatment could effectively mitigate these alternations. After treating human cardiomyocyte AC16 with PM2.5, ferroptosis activator (Erastin) and inhibitor (Fer-1), it was found that PM2.5 could promote ferritinophagy and lead to lipid peroxidation, mitochondrial dysfunction as well as the accumulation of intracellular and mitochondrial labile iron. Subsequently, mitophagy was activated and provided an additional source of labile iron, enhancing the sensitivity of AC16 cells to ferroptosis. Furthermore, Fer-1 alleviated PM2.5-induced cytotoxicity and iron overload in the cytoplasm and mitochondria of AC16 cells. It was worth noting that during the process of PM2.5 caused ferroptosis, abnormal iron metabolism mediated the activation of ferritinophagy and mitophagy in a temporal order. In addition, NCOA4 knockdown reversed the iron homeostasis imbalance and lipid peroxidation caused by PM2.5, thereby alleviating ferroptosis. In summary, our study found that iron homeostasis imbalance-mediated the crosstalk of ferritinophagy and mitophagy played an important role in PM2.5-induced ferroptosis and cardiac hypertrophy.


Sujet(s)
Autophagie , Cardiomégalie , Ferroptose , Homéostasie , Fer , Myocytes cardiaques , Matière particulaire , Cardiomégalie/métabolisme , Cardiomégalie/étiologie , Cardiomégalie/anatomopathologie , Animaux , Souris , Fer/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Humains , Matière particulaire/effets indésirables , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Peroxydation lipidique/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Lignée cellulaire
15.
Neurochem Int ; 177: 105744, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38663454

RÉSUMÉ

Traumatic brain injury (TBI) often results in persistent neurological dysfunction, which is closely associated with white matter injury. The mechanisms underlying white matter injury after TBI remain unclear. Ferritinophagy is a selective autophagic process that degrades ferritin and releases free iron, which may cause ferroptosis. Although ferroptosis has been demonstrated to be involved in TBI, it is unclear whether ferritinophagy triggers ferroptosis in TBI. Integrated stress response inhibitor (ISRIB) has neuroprotective properties. However, the effect of ISRIB on white matter after TBI remains uncertain. We aimed to investigate whether ferritinophagy was involved in white matter injury following TBI and whether ISRIB can mitigate white matter injury after TBI by inhibiting ferritinophagy. In this study, controlled cortical impact (CCI) was performed on rats to establish the TBI model. Ferritinophagy was measured by assessing the levels of nuclear receptor coactivator 4 (NCOA4), which regulates ferritinophagy, ferritin heavy chain 1(FTH1), LC3, ATG5, and FTH1 colocalization with LC3 in the white matter. Increased NCOA4 and decreased FTH1 were detected in our study. FTH1 colocalization with LC3 enhanced in the white matter after TBI, indicating that ferritinophagy was activated. Immunofluorescence co-localization results also suggested that ferritinophagy occurred in neurons and oligodendrocytes after TBI. Furthermore, ferroptosis was assessed by determining free iron content, MDA content, GSH content, and Perl's staining. The results showed that ferroptosis was suppressed by NCOA4 knockdown via shNCOA4 lentivirus infection, indicating that ferroptosis in TBI is triggered by ferritinophagy. Besides, NCOA4 deletion notably improved white matter injury following TBI, implying that ferritinophagy contributed to white matter injury. ISRIB treatment reduced the occurrence of ferritinophagy in neurons and oligodendrocytes, attenuated ferritinophagy-induced ferroptosis, and alleviated white matter injury. These findings suggest that NCOA4-mediated ferritinophagy is a critical mechanism underlying white matter injury after TBI. ISRIB holds promise as a therapeutic agent for this condition.


Sujet(s)
Lésions traumatiques de l'encéphale , Ferritines , Coactivateurs de récepteurs nucléaires , Rat Sprague-Dawley , Substance blanche , Animaux , Lésions traumatiques de l'encéphale/métabolisme , Lésions traumatiques de l'encéphale/anatomopathologie , Substance blanche/métabolisme , Substance blanche/anatomopathologie , Substance blanche/effets des médicaments et des substances chimiques , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Ferritines/métabolisme , Mâle , Rats , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/physiologie , Autophagie/effets des médicaments et des substances chimiques , Autophagie/physiologie , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique
16.
FASEB J ; 38(7): e23584, 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38568836

RÉSUMÉ

Cisplatin-induced acute kidney injury (AKI) is commonly seen in the clinical practice, and ferroptosis, a type of non-apoptotic cell death, plays a pivotal role in it. Previous studies suggested that protein arginine methyltransferase 4 (PRMT4) was incorporated in various bioprocesses, but its role in renal injuries has not been investigated. Our present study showed that PRMT4 was highly expressed in renal proximal tubular cells, and it was downregulated in cisplatin-induced AKI. Besides, genetic disruption of PRMT4 exacerbated, while its overexpression attenuated, cisplatin-induced redox injuries in renal proximal epithelia. Mechanistically, our work showed that PRMT4 interacted with NCOA4 to inhibit ferritinophagy, a type of selective autophagy favoring lipid peroxidation to accelerate ferroptosis. Taken together, our study demonstrated that PRMT4 interacted with NCOA4 to attenuate ferroptosis in cisplatin-induced AKI, suggesting that PRMT4 might present as a new therapeutic target for cisplatin-related nephropathy.


Sujet(s)
Atteinte rénale aigüe , Cisplatine , Humains , Cisplatine/effets indésirables , Atteinte rénale aigüe/induit chimiquement , Atteinte rénale aigüe/génétique , Atteinte rénale aigüe/métabolisme , Rein/métabolisme , Facteurs de transcription/métabolisme , Autophagie , Coactivateurs de récepteurs nucléaires/génétique , Coactivateurs de récepteurs nucléaires/métabolisme
17.
Free Radic Biol Med ; 217: 1-14, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38522484

RÉSUMÉ

Age-associated decreases in follicle number and oocyte quality result in a decline in female fertility, which is associated with increased infertility. Granulosa cells play a major role in oocyte development and maturation both in vivo and in vitro. However, it is unclear whether a reduction in cryptochrome 1 (Cry1) expression contributes to granulosa cell senescence, and further exploration is needed to understand the underlying mechanisms. In this study, we investigated the role of Cry1, a core component of the molecular circadian clock, in the regulation of senescence in ovarian granulosa cells. Western blotting and qRT-PCR showed that Cry1 expression was downregulated in aged human ovarian granulosa cells and was correlated with age and anti-Müllerian hormone (AMH) levels. RNA-seq analysis suggested that ferritinophagy was increased after Cry1 knockdown in KGN cells. MDA, iron, and reactive oxygen species (ROS) assays were used to detect cellular ferritinophagy levels. Ferroptosis inhibitors, iron chelators, autophagy inhibitors, and nuclear receptor coactivator 4 (NCOA4) knockdown alleviated KGN cell senescence induced by Cry1 knockdown. Immunofluorescence, immunoprecipitation, and ubiquitination assays indicated that Cry1 affected NCOA4 ubiquitination and degradation through HERC2, thereby affecting NCOA4-mediated ferritinophagy and causing granulosa cell senescence. KL201, a Cry1 stabilizer, enhanced ovarian function in naturally aged mice by reducing ferritinophagy. Our study reveals the potential mechanisms of action of Cry1 during ovarian aging and provides new insights for the clinical treatment of age-related fertility decline.


Sujet(s)
Cryptochromes , Fer , Animaux , Femelle , Humains , Souris , Autophagie/génétique , Vieillissement de la cellule , Cryptochromes/génétique , Cellules de la granulosa/métabolisme , Fer/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Coactivateurs de récepteurs nucléaires/métabolisme , Follicule ovarique/métabolisme , Facteurs de transcription/métabolisme
18.
Cell Prolif ; 57(7): e13621, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38389491

RÉSUMÉ

Nuclear receptor coactive 4 (NCOA4), which functions as a selective cargo receptor, is a critical regulator of the particularly autophagic degradation of ferritin, a process known as ferritinophagy. Mechanistically, NCOA4-mediated ferritinophagy performs an increasingly vital role in the maintenance of intracellular iron homeostasis by promoting ferritin transport and iron release as needed. Ferritinophagy is not only involved in iron-dependent responses but also in the pathogenesis and progression of various human diseases, including metabolism-related, neurodegenerative, cardiovascular and infectious diseases. Therefore, ferritinophagy is of great importance in maintaining cell viability and function and represents a potential therapeutic target. Recent studies indicated that ferritinophagy regulates the signalling pathway associated with ferroptosis, a newly discovered type of cell death characterised by iron-dependent lipid peroxidation. Although accumulating evidence clearly demonstrates the importance of the interplay between dysfunction in iron metabolism and ferroptosis, a deeper understanding of the double-edged sword effect of ferritinophagy in ferroptosis has remained elusive. Details of the mechanisms underlying the ferritinophagy-ferroptosis axis in regulating relevant human diseases remain to be elucidated. In this review, we discuss the latest research findings regarding the mechanisms that regulate the biological function of NCOA4-mediated ferritinophagy and its contribution to the pathophysiology of ferroptosis. The important role of the ferritinophagy-ferroptosis axis in human diseases will be discussed in detail, highlighting the great potential of targeting ferritinophagy in the treatment of diseases.


Sujet(s)
Autophagie , Ferritines , Ferroptose , Fer , Coactivateurs de récepteurs nucléaires , Humains , Ferroptose/physiologie , Ferritines/métabolisme , Coactivateurs de récepteurs nucléaires/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Autophagie/physiologie , Fer/métabolisme , Animaux , Transduction du signal , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/anatomopathologie
19.
Inflamm Res ; 73(3): 363-379, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38189810

RÉSUMÉ

OBJECTIVE: Ferroptosis is a reactive oxygen species (ROS)- and iron-dependent form of non-apoptotic cell death process. Previous studies have demonstrated that ferroptosis participates in the development of inflammatory arthritis. However, the role of ferroptosis in rheumatoid arthritis (RA) inflammatory hypoxic joints remains unclear. This study sought to explore the underlying mechanism of ferroptosis on lipopolysaccharide (LPS)-induced RA fibroblast-like synoviocytes (FLSs). METHODS: FLSs, isolated from patients with RA, were treated with LPS and ferroptosis inducer (erastin and RSL-3), and ferroptosis inhibitor (Fer-1 and DFO), respectively. The cell viability was measured by CCK-8. The cell death was detected by flow cytometer. The proteins level were tested by Western blot. The cytosolic ROS and lipid peroxidation were determined using DCFH-DA and C11-BODIPY581/591 fluorescence probes, respectively. The small interfering RNA (siRNA) was used to knock down related proteins. The levels of malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), iron, inflammatory cytokines (IL6 and IL8), and LDH were analyzed by commercial kits. RESULTS: Ferroptosis was activated by LPS in RA FLS with increased cellular damage, ROS and lipid peroxidation, intracellular Fe and IL8, which can be further amplified by ferroptosis inducer (erastin and RSL-3) and inhibited by ferroptosis inhibitor (Fer-1 and DFO). Mechanistically, LPS triggered ferroptosis via NCOA4-mediated ferritinophagy in RA FLSs, and knockdown of NCOA4 strikingly prevent the process of ferroptosis. Intriguingly, LPS-induced RA FLSs became insensitive to ferroptosis and NCOA4-mediated ferritinophagy under hypoxia compared with normoxia. Knockdown of HIF-1α reverted ferroptosis and ferritinophagy evoking by LPS-induced RA FLSs inflammation under hypoxia. In addition, low dose of auranofin (AUR) induced re-sensitization of ferroptosis and ferritinophagy through inhibiting the expression of HIF-1α under hypoxia. CONCLUSIONS: NCOA4-mediated ferritinophagy was a key driver of ferroptosis in inflammatory RA FLSs. The suppression of NCOA4-mediated ferritinophagy protected RA FLSs from ferroptosis in LPS-induced inflammation under hypoxia. Targeting HIF-1α/NCOA4 and ferroptosis could be an effective and valuable therapeutic strategy for synovium hyperplasia in the patients with RA.


Sujet(s)
Polyarthrite rhumatoïde , Ferroptose , Cellules synoviales , Humains , Lipopolysaccharides/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Interleukine-8/métabolisme , Polyarthrite rhumatoïde/métabolisme , Inflammation/induit chimiquement , Inflammation/métabolisme , Hypoxie/métabolisme , Facteurs de transcription/métabolisme , Petit ARN interférent/génétique , Fibroblastes/métabolisme , Fer/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Coactivateurs de récepteurs nucléaires/métabolisme
20.
Cell Mol Immunol ; 21(3): 227-244, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38195836

RÉSUMÉ

Transcriptional coactivators regulate the rate of gene expression in the nucleus. Nuclear receptor coactivator 6 (NCOA6), a coactivator, has been implicated in embryonic development, metabolism, and cancer pathogenesis, but its role in innate immunity and inflammatory diseases remains unclear. Here, we demonstrated that NCOA6 was expressed in monocytes and macrophages and that its level was increased under proinflammatory conditions. Unexpectedly, nuclear NCOA6 was found to translocate to the cytoplasm in activated monocytes and then become incorporated into the inflammasome with NLRP3 and ASC, forming cytoplasmic specks. Mechanistically, NCOA6 associated with the ATP hydrolysis motifs in the NACHT domain of NLRP3, promoting the oligomerization of NLRP3 and ASC and thereby instigating the production of IL-1ß and active caspase-1. Of note, Ncoa6 deficiency markedly inhibited NLRP3 hyperactivation caused by the Nlrp3R258W gain-of-function mutation in macrophages. Genetic ablation of Ncoa6 substantially attenuated the severity of two NLRP3-dependent diseases, folic-induced acute tubular necrosis and crystal-induced arthritis, in mice. Consistent with these findings, NCOA6 was highly expressed in macrophages derived from gout patients, and NCOA6-positive macrophages were significantly enriched in gout macrophages according to the transcriptome profiling results. Conclusively, NCOA6 is a critical regulator of NLRP3 inflammasome activation and is therefore a promising target for NLRP3-dependent diseases, including gout.


Sujet(s)
Goutte articulaire , Goutte , Animaux , Humains , Souris , Inflammasomes/métabolisme , Interleukine-1 bêta/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Coactivateurs de récepteurs nucléaires/génétique , Coactivateurs de récepteurs nucléaires/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE