Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 377
Filtrer
1.
ASN Neuro ; 16(1): 2394352, 2024.
Article de Anglais | MEDLINE | ID: mdl-39249102

RÉSUMÉ

Iron is a critical transition metal required to sustain a healthy central nervous system. Iron is involved in metabolic reactions, enzymatic activity, myelinogenesis, and oxygen transport. However, in several pathological conditions such as cancer, neurodegeneration, and neurotrauma iron becomes elevated. Excessive iron can have deleterious effects leading to reactive oxygen species (ROS) via the Fenton reaction. Iron-derived ROS are known to drive several mechanisms such as cell death pathways including ferroptosis, necroptosis, and pyroptosis. Excessive iron present in the post-traumatic brain could trigger these harmful pathways potentiating the high rates of morbidity and mortality. In the present review, we will discuss how iron plays an intricate role in initiating ferroptosis, necroptosis, and pyroptosis, examine their potential link to traumatic brain injury morbidity and mortality, and suggest therapeutic targets.


Sujet(s)
Lésions traumatiques de l'encéphale , Ferroptose , Fer , Nécroptose , Pyroptose , Pyroptose/physiologie , Humains , Ferroptose/physiologie , Fer/métabolisme , Nécroptose/physiologie , Animaux , Lésions traumatiques de l'encéphale/métabolisme , Lésions traumatiques de l'encéphale/anatomopathologie , Espèces réactives de l'oxygène/métabolisme
2.
World J Gastroenterol ; 30(32): 3730-3738, 2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39221065

RÉSUMÉ

This editorial discusses a recently published paper in the World Journal of Gastroenterology. Our research focuses on p53's regulatory mechanism for controlling ferroptosis, as well as the intricate connection between ferroptosis and liver diseases. Ferroptosis is a specific form of programmed cell death that is de-pendent on iron and displays unique features in terms of morphology, biology, and genetics, distinguishing it from other forms of cell death. Ferroptosis can affect the liver, which is a crucial organ responsible for iron storage and meta-bolism. Mounting evidence indicates a robust correlation between ferroptosis and the advancement of liver disorders. P53 has a dual effect on ferroptosis through various distinct signaling pathways. However, additional investigations are required to clarify the regulatory function of p53 metabolic targets in this complex association with ferroptosis. In the future, researchers should clarify the mechanisms by which ferroptosis and other forms of programmed cell death contribute to the progression of liver diseases. Identifying and controlling important regulatory factors associated with ferroptosis present a promising therapeutic strategy for liver disorders.


Sujet(s)
Ferroptose , Fer , Maladies du foie , Foie , Transduction du signal , Protéine p53 suppresseur de tumeur , Ferroptose/physiologie , Humains , Maladies du foie/métabolisme , Maladies du foie/anatomopathologie , Fer/métabolisme , Foie/métabolisme , Foie/anatomopathologie , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Animaux , Évolution de la maladie
3.
Arch Dermatol Res ; 316(8): 604, 2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39240413

RÉSUMÉ

BACKGROUND: Abnormal biological behaviour of keratinocytes (KCs) is a critical pathophysiological manifestation of psoriasis. Ferroptosis is programmed cell death induced by the accumulation of lipid reactive oxygen species (ROS) in the presence of increased intracellular iron ions or inhibition of GPX4. OBJECTIVES: The purpose of this study was to investigate the effects of ferroptosis on the biological behaviour of Keratinocytes (KCs) in psoriasis vulgaris and its possible regulatory mechanisms in clinical samples, cells, and mouse models. METHODS: We first examined the differences in the expression of GPX4 and 4-HNE between psoriasis and normal human lesions. And detected KRT6, FLG, and inflammatory cytokines after inducing ferroptosis in animal and cell models by RT-qPCR, Western blot, immunohistochemistry, and flow cytometry. RESULTS: We found that GPX4 was decreased and that the oxidation product 4-hydroxy-2-nonenal (HNE) was increased in the skin lesions of patients with psoriasis vulgaris. The expression level of GPX4 correlates with the severity of skin lesions. Moreover, inducing ferroptosis promoted the expression of FLG and reduced the expression of KRT6 and inflammatory cytokines in vitro, and alleviated the phenotype of skin lesions in vivo. LIMITATIONS: Our study has limitations, notably small sample size. Larger clinical trials are necessary to investigate the association between ferroptosis and disease progression further. More research is necessary to explore how the ferroptosis inducer RSL3 regulates the abnormal biological behaviour of KCs at both cellular and animal levels and establish ferroptosis inhibitors as controls. CONCLUSIONS: This study confirms the existence of ferroptosis in psoriatic lesions, which may be inversely correlated with disease severity. The ferroptosis inducer RSL3 ameliorated psoriatic symptoms by improving the abnormal biological behaviour of KCs.


Sujet(s)
Modèles animaux de maladie humaine , Ferroptose , Kératinocytes , Phospholipid hydroperoxide glutathione peroxidase , Psoriasis , Psoriasis/anatomopathologie , Psoriasis/métabolisme , Psoriasis/immunologie , Ferroptose/physiologie , Kératinocytes/métabolisme , Kératinocytes/anatomopathologie , Humains , Animaux , Souris , Projets pilotes , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/génétique , Aldéhydes/métabolisme , Femelle , Mâle , Adulte , Kératine-6/métabolisme , Cytokines/métabolisme , Peau/anatomopathologie , Peau/métabolisme , Peau/immunologie , Adulte d'âge moyen , Résorcinol/pharmacologie , Espèces réactives de l'oxygène/métabolisme , Carbolines
4.
CNS Neurosci Ther ; 30(9): e70030, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39233353

RÉSUMÉ

BACKGROUND: Ischemic stroke leads a primary cause of mortality in human diseases, with a high disability rate worldwide. This study aims to investigate the function of ß-1,4-galactosyltransferase 1 (B4galt1) in mouse brain ischemia/reperfusion (I/R) injury. METHODS: Recombinant human B4galt1 (rh-B4galt1) was intranasally administered to the mice model of middle cerebral artery occlusion (MCAO)/reperfusion. In this study, the impact of rh-B4galt1 on cerebral injury assessed using multiple methods, including the neurological disability status scale, 2,3,5-triphenyltetrazolium chloride (TTC), Nissl and TUNEL staining. This study utilized laser speckle Doppler flowmeter to monitor the cerebral blood flow. Western blotting was performed to assess the protein expression levels, and fluorescence-labeled dihydroethidium method was performed to determine the superoxide anion generation. Assay kits were used for the measurement of iron, malondialdehyde (MDA) and glutathione (GSH) levels. RESULTS: We demonstrated that rh-B4galt1 markedly improved neurological function, reduced cerebral infarct volume and preserved the completeness of blood-brain barrier (BBB) for preventing damage. These findings further illustrated that rh-B4galt1 alleviated oxidative stress, lipid peroxidation, as well as iron deposition induced by I/R. The vital role of ferroptosis was proved in brain injury. Furthermore, the rh-B4galt1 could increase the levels of TAZ, Nrf2 and HO-1 after I/R. And TAZ-siRNA and ML385 reversed the neuroprotective effects of rh-B4galt1. CONCLUSIONS: The results indicated that rh-B4galt1 implements neuroprotective effects by modulating ferroptosis, primarily via upregulating TAZ/Nrf2/HO-1 pathway. Thus, B4galt1 could be seen as a promising novel objective for ischemic stroke therapy.


Sujet(s)
Encéphalopathie ischémique , Ferroptose , Galactosyltransferases , Heme oxygenase-1 , Facteur-2 apparenté à NF-E2 , Transduction du signal , Animaux , Humains , Mâle , Souris , Encéphalopathie ischémique/métabolisme , Encéphalopathie ischémique/prévention et contrôle , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/physiologie , Galactosyltransferases/métabolisme , Heme oxygenase-1/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne , Protéines membranaires , Souris de lignée C57BL , Facteur-2 apparenté à NF-E2/métabolisme , Lésion d'ischémie-reperfusion/prévention et contrôle , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/anatomopathologie , Transduction du signal/physiologie , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs de transcription/métabolisme
5.
Redox Rep ; 29(1): 2387465, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39102510

RÉSUMÉ

BACKGROUD: Bronchopulmonary dysplasia (BPD) is one of the most important complications plaguing neonates and can lead to a variety of sequelae. the ability of the HIF-1α/VEGF signaling pathway to promote angiogenesis has an important role in neonatal lung development. METHOD: Newborn rats were exposed to 85% oxygen. The effects of hyperoxia exposure on Pleomorphic Adenoma Gene like-2 (PLAGL2) and the HIF-1α/VEGF pathway in rats lung tissue were assessed through immunofluorescence and Western Blot analysis. In cell experiments, PLAGL2 was upregulated, and the effects of hyperoxia and PLAGL2 on cell viability were evaluated using scratch assays, CCK-8 assays, and EDU staining. The role of upregulated PLAGL2 in the HIF-1α/VEGF pathway was determined by Western Blot and RT-PCR. Apoptosis and ferroptosis effects were determined through flow cytometry and viability assays. RESULTS: Compared with the control group, the expression levels of PLAGL2, HIF-1α, VEGF, and SPC in lung tissues after 3, 7, and 14 days of hyperoxia exposure were all decreased. Furthermore, hyperoxia also inhibited the proliferation and motility of type II alveolar epithelial cells (AECII) and induced apoptosis in AECII. Upregulation of PLAGL2 restored the proliferation and motility of AECII and suppressed cell apoptosis and ferroptosis, while the HIF-1α/VEGF signaling pathway was also revived. CONCLUSIONS: We confirmed the positive role of PLAGL2 and HIF-1α/VEGF signaling pathway in promoting BPD in hyperoxia conditions, and provided a promising therapeutic targets.


Sujet(s)
Pneumocytes , Animaux nouveau-nés , Apoptose , Ferroptose , Hyperoxie , Sous-unité alpha du facteur-1 induit par l'hypoxie , Transduction du signal , Facteur de croissance endothéliale vasculaire de type A , Animaux , Rats , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Pneumocytes/métabolisme , Ferroptose/physiologie , Hyperoxie/métabolisme , Rat Sprague-Dawley , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Régulation négative , Humains , Prolifération cellulaire
6.
Front Endocrinol (Lausanne) ; 15: 1390013, 2024.
Article de Anglais | MEDLINE | ID: mdl-39157678

RÉSUMÉ

Polycystic ovarian syndrome (PCOS) is a common heterogeneous reproductive endocrine metabolic disorder in women of reproductive age characterized by clinical and biochemical hyperandrogenemia, ovulation disorders, and polycystic ovarian morphology. Ferroptosis is a novel type of cell death driven by iron accumulation and lipid peroxidation. Ferroptosis plays a role in maintaining redox balance, iron metabolism, lipid metabolism, amino acid metabolism, mitochondrial activity, and many other signaling pathways linked to diseases. Iron overload is closely related to insulin resistance, decreased glucose tolerance, and the occurrence of diabetes mellitus. There is limited research on the role of ferroptosis in PCOS. Patients with PCOS have elevated levels of ferritin and increased reactive oxygen species in ovarian GCs. Studying ferroptosis in PCOS patients is highly important for achieving personalized treatment. This article reviews the progress of research on ferroptosis in PCOS, introduces the potential connections between iron metabolism abnormalities and oxidative stress-mediated PCOS, and provides a theoretical basis for diagnosing and treating PCOS.


Sujet(s)
Ferroptose , Fer , Stress oxydatif , Syndrome des ovaires polykystiques , Humains , Syndrome des ovaires polykystiques/métabolisme , Syndrome des ovaires polykystiques/anatomopathologie , Ferroptose/physiologie , Femelle , Fer/métabolisme , Espèces réactives de l'oxygène/métabolisme , Insulinorésistance/physiologie , Animaux , Peroxydation lipidique
7.
Drug Dev Res ; 85(6): e22245, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39154227

RÉSUMÉ

Intracerebral hemorrhage (ICH) is a severe hemorrhagic stroke and induces severe secondary neurological injury. However, its pathogenesis remains to be explored. The present work investigates the role of glutathione S-transferase omega 2 (GSTO2) in ICH and the underlying mechanism. Human neuroblastoma cells (SK-N-SH) were stimulated using hemin to mimic ICH-like injury. Protein expression levels of GSTO2 and glutathione peroxidase 4 (GPX4) were detected by western blot analysis assay. Cell viability was assessed by cell counting kit-8 assay. Cell proliferation was investigated by 5-ethynyl-2'-deoxyuridine assay. Cell apoptosis was analyzed by flow cytometry. Interleukin-6 and tumor necrosis factor-α levels were quantified by enzyme-linked immunosorbent assays. Fe2+ colorimetric assay kit was used to detect Fe2+ level. A cellular reactive oxygen species (ROS) assay kit was used to detect ROS levels. Malondialdehyde (MDA) level was assessed using the MDA content assay kit. GSH level was quantified using the GSH assay kit. Co-immunoprecipitation assay was performed to identify the association between GSTO2 and GPX4. Hemin stimulation suppressed SK-N-SH cell proliferation and promoted cell apoptosis, cell inflammation, ferroptosis, and oxidative stress. GSTO2 expression was downregulated in hemin-treated SK-N-SH cells in comparison with the control group. In addition, ectopic GSTO2 expression counteracted hemin-induced inhibitory effect on cell proliferation and promoting effects on cell apoptosis, inflammation, ferroptosis, and oxidative stress. Moreover, GSTO2 was associated with GPX4 in SK-N-SH cells. GPX4 silencing attenuated GSTO2 overexpression-induced effects on hemin-stimulated SK-N-SH cell injury. GSTO2 ameliorated SK-N-SH cell apoptosis, inflammation, ferroptosis, and oxidative stress by upregulating GPX4 expression in ICH, providing a therapeutic strategy for ICH.


Sujet(s)
Apoptose , Hémorragie cérébrale , Ferroptose , Inflammation , Neuroblastome , Stress oxydatif , Phospholipid hydroperoxide glutathione peroxidase , Régulation positive , Humains , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/physiologie , Stress oxydatif/effets des médicaments et des substances chimiques , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Hémorragie cérébrale/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Inflammation/métabolisme , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Glutathione transferase/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Hémine/pharmacologie , Espèces réactives de l'oxygène/métabolisme
8.
Sheng Li Xue Bao ; 76(4): 507-516, 2024 Aug 25.
Article de Chinois | MEDLINE | ID: mdl-39192784

RÉSUMÉ

The present study aimed to investigate the occurrence of ferroptosis in mouse hippocampal tissue and changes in related pathways after exposure to high-altitude hypoxia. A low-pressure hypoxia model was established using a high-altitude environment at 4 010 m. HE staining was used to observe morphological changes in mouse hippocampal tissue, immunohistochemical staining was used to observe lipid peroxidation levels in hippocampal tissue, and corresponding kits were used to measure malondialdehyde (MDA), reduced glutathione (GSH), and Fe2+ levels in hippocampal tissue. Western blot was used to detect glutathione peroxidase 4 (GPX4), solute carrier family 7 member 11 (SLC7A11), ferritin heavy chain 1 (FTH1), ferroportin 1 (FPN1), transferrin receptor 1 (TfR1), ferroptosis suppressor protein 1 (FSP1), and acyl-CoA synthase long chain family member 4 (ACSL4). The results showed that, compared with the plain control group, the mice exposed to high-altitude hypoxia for 1, 3, 7, and 14 d exhibited significant pathological damage, disordered arrangement, and obvious nuclear condensation in the dentate gyrus of the hippocampus. Compared with the plain control group, high-altitude hypoxia exposure increased 4-hydroxynonenal (4-HNE) content in the dentate gyrus and hippocampal MDA content, whereas significantly decreased hippocampal GSH content. Compared with the plain control group, the Fe2+ content in the hippocampus of mice exposed to high-altitude hypoxia for 14 d significantly increased. Compared with the plain control group, the protein expression levels of GPX4, FTH1, FPN1, TfR1, and FSP1 in the hippocampus of mice exposed to high-altitude hypoxia were significantly down-regulated (SLC7A11 was significantly down-regulated only in the 7-d high-altitude hypoxia exposure group), while the protein expression level of ACSL4 was only significantly up-regulated in the 14-d high-altitude hypoxia exposure group. These results suggest that exposure to high-altitude hypoxia for 14 d can reduce GSH synthesis in mouse hippocampus, down-regulate GPX4 expression, lead to GSH metabolism disorders, inhibit iron storage and efflux, promote lipid peroxidation reaction, and inhibit CoQ10H2's anti-lipid peroxidation effect, ultimately leading to ferroptosis.


Sujet(s)
Mal de l'altitude , Ferroptose , Hippocampe , Hypoxie , Animaux , Ferroptose/physiologie , Hippocampe/métabolisme , Souris , Hypoxie/métabolisme , Hypoxie/physiopathologie , Mâle , Mal de l'altitude/métabolisme , Mal de l'altitude/physiopathologie , Peroxydation lipidique , Récepteurs à la transferrine/métabolisme , Altitude , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Glutathion/métabolisme , Malonaldéhyde/métabolisme , Fer/métabolisme , Transporteurs de cations/métabolisme , Système y+ de transport d'acides aminés/métabolisme , Système y+ de transport d'acides aminés/génétique
9.
Front Biosci (Landmark Ed) ; 29(8): 291, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39206899

RÉSUMÉ

Neurodegenerative disorders are typified by the progressive degeneration and subsequent apoptosis of neuronal cells. They encompass a spectrum of conditions, including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), epilepsy, brian ischemia, brian injury, and neurodegeneration with brain iron accumulation (NBIA). Despite the considerable heterogeneity in their clinical presentation, pathophysiological underpinning and disease trajectory, a universal feature of these disorders is the functional deterioration of the nervous system concomitant with neuronal apoptosis. Ferroptosis is an iron (Fe)-dependent form of programmed cell death that has been implicated in the pathogenesis of these conditions. It is intricately associated with intracellular Fe metabolism and lipid homeostasis. The accumulation of Fe is observed in a variety of neurodegenerative diseases and has been linked to their etiology and progression, although its precise role in these pathologies has yet to be elucidated. This review aims to elucidate the characteristics and regulatory mechanisms of ferroptosis, its association with neurodegenerative diseases, and recent advances in ferroptosis-targeted therapeutic strategies. Ferroptosis may therefore be a critical area for future research into neurodegenerative diseases.


Sujet(s)
Ferroptose , Fer , Maladies neurodégénératives , Ferroptose/physiologie , Humains , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/physiopathologie , Fer/métabolisme , Animaux , Neurones/métabolisme , Neurones/anatomopathologie
10.
CNS Neurosci Ther ; 30(8): e14911, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39145422

RÉSUMÉ

BACKGROUND: Epilepsy is a widespread central nervous system disorder with an estimated 50 million people affected globally. It is characterized by a bimodal incidence peak among infants and the elderly and is influenced by a variety of risk factors, including a significant genetic component. Despite the use of anti-epileptic drugs (AEDs), drug-refractory epilepsy develops in about one-third of patients, highlighting the need for alternative therapeutic approaches. AIMS: The primary aim of this study was to evaluate the neuroprotective effects of troglitazone (TGZ) in epilepsy and to explore the potential mechanisms underlying its action. METHODS: We employed both in vitro and in vivo models to assess TGZ's effects. The in vitro model involved glutamate-induced toxicity in HT22 mouse hippocampal neurons, while the in vivo model used kainic acid (KA) to induce epilepsy in mice. A range of methods, including Hoechst/PI staining, CCK-8 assay, flow cytometry, RT-PCR analysis, Nissl staining, scanning electron microscopy, and RNA sequencing, were utilized to assess various parameters such as cellular damage, viability, lipid-ROS levels, mitochondrial membrane potential, mRNA expression, seizure grade, and mitochondrial morphology. RESULTS: Our results indicate that TGZ, at doses of 5 or 20 mg/kg/day, significantly reduces KA-induced seizures and neuronal damage in mice by inhibiting the process of ferroptosis. Furthermore, TGZ was found to prevent changes in mitochondrial morphology. In the glutamate-induced HT22 cell damage model, 2.5 µM TGZ effectively suppressed neuronal ferroptosis, as shown by a reduction in lipid-ROS accumulation, a decrease in mitochondrial membrane potential, and an increase in PTGS2 expression. The anti-ferroptotic effect of TGZ was confirmed in an erastin-induced HT22 cell damage model as well. Additionally, TGZ reversed the upregulation of Plaur expression in HT22 cells treated with glutamate or erastin. The downregulation of Plaur expression was found to alleviate seizures and reduce neuronal damage in the mouse hippocampus. CONCLUSION: This study demonstrates that troglitazone has significant therapeutic potential in the treatment of epilepsy by reducing epileptic seizures and the associated brain damage through the inhibition of neuronal ferroptosis. The downregulation of Plaur expression plays a crucial role in TGZ's anti-ferroptotic effect, offering a promising avenue for the development of new epilepsy treatments.


Sujet(s)
Épilepsie , Ferroptose , Neuroprotecteurs , Troglitazone , Animaux , Souris , Épilepsie/traitement médicamenteux , Épilepsie/induit chimiquement , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/physiologie , Neuroprotecteurs/pharmacologie , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/anatomopathologie , Hippocampe/métabolisme , Acide glutamique/métabolisme , Mâle , Acide kaïnique/toxicité , Souris de lignée C57BL , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Anticonvulsivants/pharmacologie , Anticonvulsivants/usage thérapeutique
11.
Int Immunopharmacol ; 140: 112819, 2024 Oct 25.
Article de Anglais | MEDLINE | ID: mdl-39096870

RÉSUMÉ

Ferroptosis represents a novel mode of programmed cell death characterized by the intracellular accumulation of iron and lipid peroxidation, culminating in oxidative stress and subsequent cell demise. Mounting evidence demonstrates that ferroptosis contributes significantly to the onset and progression of diverse pathological conditions and diseases, including infections, neurodegenerative disorders, tissue ischemia-reperfusion injury, and immune dysregulation. Recent investigations have underscored the pivotal role of ferroptosis in the pathogenesis of rheumatoid arthritis, ulcerative colitis, systemic lupus erythematosus, and asthma. This review provides a comprehensive overview of the current understanding of the regulatory mechanisms governing ferroptosis, particularly its interplay with iron, lipid, and amino acid metabolism. Furthermore, we explore the implications of ferroptosis in autoimmune diseases and deliberate on its potential as a promising therapeutic target for diverse autoimmune disorders.


Sujet(s)
Maladies auto-immunes , Ferroptose , Ferroptose/physiologie , Humains , Maladies auto-immunes/immunologie , Animaux , Fer/métabolisme , Peroxydation lipidique , Stress oxydatif
12.
Neuromolecular Med ; 26(1): 33, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138706

RÉSUMÉ

The newly identified estrogen receptor, G protein-coupled receptor 30 (GPR30), is prevalent in the brain and has been shown to provide significant neuroprotection. Recent studies have linked ferroptosis, a newly characterized form of programmed cell death, closely with cerebral ischemia-reperfusion injury (CIRI), highlighting it as a major contributing factor. Consequently, our research aimed to explore the potential of GPR30 targeting in controlling neuronal ferroptosis and lessening CIRI impacts. Results indicated that GPR30 activation not only improved neurological outcomes and decreased infarct size in a mouse model but also lessened iron accumulation and malondialdehyde formation post-middle cerebral artery occlusion (MCAO). This protective effect extended to increased levels of Nrf2 and GPX4 proteins. Similar protective results were replicated in PC12 cells subjected to Oxygen Glucose Deprivation and Reoxygenation (OGD/R) using the GPR30-specific agonist G1. Importantly, inhibition of Nrf2 with ML385 curtailed the neuroprotective effects of GPR30 activation, suggesting that GPR30 mitigates CIRI primarily through inhibition of neuronal ferroptosis via upregulation of Nrf2 and GPX4.


Sujet(s)
Ferroptose , Infarctus du territoire de l'artère cérébrale moyenne , Souris de lignée C57BL , Facteur-2 apparenté à NF-E2 , Phospholipid hydroperoxide glutathione peroxidase , Récepteurs des oestrogènes , Récepteurs couplés aux protéines G , Lésion d'ischémie-reperfusion , Transduction du signal , Animaux , Facteur-2 apparenté à NF-E2/métabolisme , Lésion d'ischémie-reperfusion/prévention et contrôle , Lésion d'ischémie-reperfusion/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/physiologie , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/physiologie , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Cellules PC12 , Phospholipid hydroperoxide glutathione peroxidase/génétique , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Rats , Mâle , Neuroprotecteurs/usage thérapeutique , Neuroprotecteurs/pharmacologie , Modèles animaux de maladie humaine
13.
Vet Res ; 55(1): 103, 2024 Aug 18.
Article de Anglais | MEDLINE | ID: mdl-39155369

RÉSUMÉ

Porcine reproductive and respiratory syndrome virus (PRRSV) is a severe disease with substantial economic consequences for the swine industry. The DEAD-box helicase 3 (DDX3X) is an RNA helicase that plays a crucial role in regulating RNA metabolism, immunological response, and even RNA virus infection. However, it is unclear whether it contributes to PRRSV infection. Recent studies have found that the expression of DDX3X considerably increases in Marc-145 cells when infected with live PRRSV strains Ch-1R and SD16; however, it was observed that inactivated viruses did not lead to any changes. By using the RK-33 inhibitor or DDX3X-specific siRNAs to reduce DDX3X expression, there was a significant decrease in the production of PRRSV progenies. In contrast, the overexpression of DDX3X in host cells substantially increased the proliferation of PRRSV. A combination of transcriptomics and metabolomics investigations revealed that in PRRSV-infected cells, DDX3X gene silencing severely affected biological processes such as ferroptosis, the FoxO signalling pathway, and glutathione metabolism. The subsequent transmission electron microscopy (TEM) imaging displayed the typical ferroptosis features in PRRSV-infected cells, such as mitochondrial shrinkage, reduction or disappearance of mitochondrial cristae, and cytoplasmic membrane rupture. Conversely, the mitochondrial morphology was unchanged in DDX3X-inhibited cells. Furthermore, silencing of the DDX3X gene changed the expression of ferroptosis-related genes and inhibited the virus proliferation, while the drug-induced ferroptosis inversely promoted PRRSV replication. In summary, these results present an updated perspective of how PRRSV infection uses DDX3X for self-replication, potentially leading to ferroptosis via various mechanisms that promote PRRSV replication.


Sujet(s)
DEAD-box RNA helicases , Ferroptose , Virus du syndrome respiratoire et reproducteur porcin , Réplication virale , Virus du syndrome respiratoire et reproducteur porcin/physiologie , Animaux , DEAD-box RNA helicases/métabolisme , DEAD-box RNA helicases/génétique , Ferroptose/physiologie , Suidae , Syndrome dysgénésique et respiratoire porcin/virologie , Syndrome dysgénésique et respiratoire porcin/métabolisme , Lignée cellulaire
14.
Int J Biol Sci ; 20(9): 3515-3529, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993565

RÉSUMÉ

Impaired angiogenesis is a major factor contributing to delayed wound healing in diabetes. Dysfunctional mitochondria promote the formation of neutrophil extracellular traps (NETs), obstructing angiogenesis during wound healing. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have shown promise in promoting tissue repair and regeneration in diabetes; however, the precise pathways involved in this process remain unclear. In this study, NET-induced ferroptosis of endothelial cells (ECs) and angiogenesis were assessed in diabetic wound samples from both patients and animal models. In vitro and in vivo experiments were performed to examine the regulatory mechanisms of NETs in ECs using specific inhibitors and gene-knockout mice. MSC-EVs encapsulating dysfunctional mitochondria were used to trigger mitochondrial fusion and restore mitochondrial function in neutrophils to suppress NET formation. Angiogenesis in wound tissue was evaluated using color laser Doppler imaging and vascular density analysis. Wound healing was evaluated via macroscopic analysis and histological evaluation of the epithelial gap. NET-induced ferroptosis of ECs was validated as a crucial factor contributing to the impairment of angiogenesis in diabetic wounds. Mechanistically, NETs regulated ferroptosis by suppressing the PI3K/AKT pathway. Furthermore, MSC-EVs transferred functional mitochondria to neutrophils in wound tissue, triggered mitochondrial fusion, and restored mitochondrial function, thereby reducing NET formation. These results suggest that inhibiting NET formation and EC ferroptosis or activating the PI3K/AKT pathway can remarkably improve wound healing. In conclusion, this study reveals a novel NET-mediated pathway involved in wound healing in diabetes and suggests an effective therapeutic strategy for accelerating wound healing.


Sujet(s)
Cellules endothéliales , Pièges extracellulaires , Vésicules extracellulaires , Ferroptose , Cellules souches mésenchymateuses , Cicatrisation de plaie , Animaux , Ferroptose/physiologie , Cicatrisation de plaie/physiologie , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/transplantation , Cellules souches mésenchymateuses/métabolisme , Souris , Humains , Cellules endothéliales/métabolisme , Pièges extracellulaires/métabolisme , Mâle , Souris de lignée C57BL , Granulocytes neutrophiles/métabolisme , Mitochondries/métabolisme , Souris knockout , Phosphatidylinositol 3-kinases/métabolisme
15.
Front Endocrinol (Lausanne) ; 15: 1431652, 2024.
Article de Anglais | MEDLINE | ID: mdl-39036052

RÉSUMÉ

Non-alcoholic fatty liver disease (NAFLD) is a prevalent and significant global public health issue. Nonalcoholic steatohepatitis (NASH) represents an advanced stage of NAFLD in terms of pathology. However, the intricate mechanisms underlying the progression from NAFLD to NASH remain elusive. Ferroptosis, characterized by iron-dependent cell death and distinguished from other forms of cell death based on morphological, biochemical, and genetic criteria, has emerged as a potential participant with a pivotal role in driving NAFLD progression. Nevertheless, its precise mechanism remains poorly elucidated. In this review article, we comprehensively summarize the pathogenesis of NAFLD/NASH and ferroptosis while highlighting recent advances in understanding the mechanistic involvement of ferroptosis in NAFLD/NASH.


Sujet(s)
Évolution de la maladie , Ferroptose , Stéatose hépatique non alcoolique , Ferroptose/physiologie , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/métabolisme , Humains , Animaux , Fer/métabolisme
16.
CNS Neurosci Ther ; 30(7): e14865, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39042604

RÉSUMÉ

Ferroptosis is a newly discovered form of programmed cell death that is non-caspase-dependent and is characterized by the production of lethal levels of iron-dependent lipid reactive oxygen species (ROS). In recent years, ferroptosis has attracted great interest in the field of cerebral infarction because it differs morphologically, physiologically, and genetically from other forms of cell death such as necrosis, apoptosis, autophagy, and pyroptosis. In addition, ROS is considered to be an important prognostic factor for ischemic stroke, making it a promising target for stroke treatment. This paper summarizes the induction and defense mechanisms associated with ferroptosis, and explores potential treatment strategies for ischemic stroke in order to lay the groundwork for the development of new neuroprotective drugs.


Sujet(s)
Ferroptose , Accident vasculaire cérébral ischémique , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/physiologie , Humains , Accident vasculaire cérébral ischémique/traitement médicamenteux , Accident vasculaire cérébral ischémique/métabolisme , Accident vasculaire cérébral ischémique/anatomopathologie , Animaux , Espèces réactives de l'oxygène/métabolisme , Neuroprotecteurs/usage thérapeutique , Neuroprotecteurs/pharmacologie
17.
Braz J Med Biol Res ; 57: e13961, 2024.
Article de Anglais | MEDLINE | ID: mdl-38985083

RÉSUMÉ

Glioblastomas are known for their poor clinical prognosis, with recurrent tumors often exhibiting greater invasiveness and faster growth rates compared to primary tumors. To understand the intratumoral changes driving this phenomenon, we employed single-cell sequencing to analyze the differences between two pairs of primary and recurrent glioblastomas. Our findings revealed an upregulation of ferroptosis in endothelial cells within recurrent tumors, identified by the significant overexpression of the NOX4 gene. Further analysis indicated that knocking down NOX4 in endothelial cells reduced the activity of the ferroptosis pathway. Utilizing conditioned media from endothelial cells with lower ferroptosis activity, we observed a decrease in the growth rate of glioblastoma cells. These results highlighted the complex role of ferroptosis within tumors and suggested that targeting ferroptosis in the treatment of glioblastomas requires careful consideration of its effects on endothelial cells, as it may otherwise produce counterproductive outcomes.


Sujet(s)
Tumeurs du cerveau , Cellules endothéliales , Ferroptose , Glioblastome , Isocitrate dehydrogenases , Récidive tumorale locale , Humains , Glioblastome/anatomopathologie , Glioblastome/génétique , Ferroptose/génétique , Ferroptose/physiologie , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/génétique , Isocitrate dehydrogenases/génétique , Cellules endothéliales/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire
18.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(3): 377-384, 2024 Mar 28.
Article de Anglais, Chinois | MEDLINE | ID: mdl-38970511

RÉSUMÉ

Secondary nephrosis is a series of chronic kidney diseases secondary to other underlying diseases, mainly manifesting as structural and functional abnormalities of the kidneys and metabolic disorders. It is one of the important causes of end-stage renal disease, with high morbidity and significant harm. Iron is an essential metal element in human cells, and ferroptosis is a non-traditional form of iron-dependent cell death, and its main mechanisms include iron accumulation, lipid metabolism disorders, abnormal amino acid metabolism, and damage to the antioxidant system. Recently studies have found that ferroptosis is involved in the occurrence and progression of secondary nephrosis, and the mechanism of ferroptosis in different secondary nephrosis vary. Therefore, an in-depth and systematic understanding of the association between ferroptosis and secondary nephrosis, as well as their specific regulatory mechanisms, can provide a theoretical basis for the diagnosis, prevention, treatment, and prognosis assessment of secondary nephrosis, laying the foundation for exploring new clinical therapeutic targets for secondary nephrosis.


Sujet(s)
Ferroptose , Fer , Néphrose , Humains , Ferroptose/physiologie , Fer/métabolisme , Néphrose/métabolisme , Animaux , Défaillance rénale chronique/complications , Métabolisme lipidique
19.
Allergol Immunopathol (Madr) ; 52(4): 60-67, 2024.
Article de Anglais | MEDLINE | ID: mdl-38970266

RÉSUMÉ

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is a familiar disease, and owns high morbidity and mortality, which critically damages the health of patients. Ubiquitin-specific peptidase 8 (USP8) is a pivotal protein to join in the regulation of some diseases. In a previous report, it was determined that USP8 expression is down-regulated in LPS-treated BEAS-2B cells, and USP8 restrains inflammatory response and accelerates cell viability. However, the regulatory roles of USP8 on ferroptosis in COPD are rarely reported, and the associated molecular mechanisms keep vague. OBJECTIVE: To investigate the regulatory functions of USP8 in COPD progression. MATERIAL AND METHODS: The lung functions were measured through the Buxco Fine Pointe Series Whole Body Plethysmography (WBP). The Fe level was tested through the Fe assay kit. The protein expressions were assessed through western blot. The levels of tumor necrosis -factor-α, interleukin 6, and interleukin 8 were evaluated through enzyme-linked immunosorbent serologic assay. Cell viability was tested through CCK-8 assay. RESULTS: In this work, it was discovered that overexpression of USP8 improved lung function in COPD mice. In addition, overexpression of USP8 repressed ferroptosis by regulating glutathione peroxidase 4 and acyl-CoA synthetase long-chain family 4 expressions in COPD mice. Overexpression of USP8 suppressed inflammation in COPD mice. Furthermore, overexpression of USP8 suppressed ferroptosis in COPD cell model. At last, it was verified that overexpression of USP8 accelerated ubiquitin aldehyde-binding protein 1 (OTUB1)/solute carrier family 7 member 11 (SLC7A11) pathway. CONCLUSION: This study manifested that overexpression of USP8 restrained inflammation and ferroptosis in COPD by regulating the OTUB1/SLC7A11 signaling pathway. This discovery hinted that USP8 could be a potential target for COPD treatment.


Sujet(s)
Système y+ de transport d'acides aminés , Ferroptose , Broncho-pneumopathie chronique obstructive , Transduction du signal , Ubiquitin thiolesterase , Ferroptose/physiologie , Broncho-pneumopathie chronique obstructive/métabolisme , Broncho-pneumopathie chronique obstructive/génétique , Animaux , Humains , Souris , Transduction du signal/immunologie , Système y+ de transport d'acides aminés/métabolisme , Système y+ de transport d'acides aminés/génétique , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Mâle , Inflammation/métabolisme , Inflammation/immunologie , Modèles animaux de maladie humaine , Souris de lignée C57BL , Complexes de tri endosomique requis pour le transport/métabolisme , Complexes de tri endosomique requis pour le transport/génétique , Lignée cellulaire , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Endopeptidases
20.
CNS Neurosci Ther ; 30(7): e14887, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39073013

RÉSUMÉ

AIMS: Neuroinflammation is a recognized contributor to cognitive disorders like Alzheimer's disease, with ferroptosis emerging as a novel mechanism underlying cognitive dysfunction associated with neuroinflammation. Insulin, pivotal in the central nervous system, holds promise for cognitive function enhancement. This study aimed to establish a cognitive impairment model through intracerebroventricular injection of lipopolysaccharide (LPS) and explore the impact of intracerebroventricular insulin injection on cognitive function in mice. METHODS: We employed diverse experimental techniques, including animal behavior testing, molecular assays, targeted metabolomics, nuclear medicine, and electron microscopy, to assess neurodegenerative changes, brain insulin resistance (IR), glucose uptake and metabolism, and ferroptosis. The model of cognitive impairment was induced via intracerebroventricular injection of LPS, followed by intracerebroventricular administration of insulin to evaluate its effects. RESULTS: Insulin treatment effectively mitigated LPS-induced cognitive decline and safeguarded against neuronal degeneration. Furthermore, insulin alleviated LPS-induced insulin resistance, enhanced glucose uptake in the hippocampus, and promoted the Pentose Phosphate Pathway (PPP) and nicotinamide adenine dinucleotide phosphate (NADPH) production. Additionally, insulin activated the glutathione (GSH)-glutathione peroxidase 4 (GPX4) pathway, reducing lipid peroxidation, and mitochondrial damage characteristic of LPS-induced ferroptosis in the hippocampus. CONCLUSION: Our findings underscore the therapeutic potential of insulin in alleviating LPS-induced cognitive impairment and ferroptosis by modulating glucose metabolism. This study offers a promising avenue for future interventions targeting cognitive decline.


Sujet(s)
Dysfonctionnement cognitif , Ferroptose , Glucose , Hippocampe , Insuline , Lipopolysaccharides , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/physiologie , Dysfonctionnement cognitif/traitement médicamenteux , Dysfonctionnement cognitif/induit chimiquement , Dysfonctionnement cognitif/métabolisme , Lipopolysaccharides/toxicité , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Souris , Mâle , Glucose/métabolisme , Souris de lignée C57BL , Insulinorésistance/physiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE