Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 649
Filtrar
1.
Mol Med ; 30(1): 150, 2024 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-39272008

RESUMO

Cis-dichlorodiammineplatinum(II) (CDDP), while widely utilized in tumor therapy, results in toxic side effects that patients find intolerable. The specific mechanism by which CDDP inflicts ovarian damage remains unclear. This study aimed to explore the involvement of ferrostatin-1 (FER-1) and ferroptosis in CDDP-induced ovarian toxicity. This study established models of CDDP-induced injury in granulosa cells (GCs) and rat model of premature ovarian failure (POF). CCK-8 assessed the effects of CDDP and FER-1 on GC viability. FerroOrange and Mito-FerroGreen, DCFH-DA and MitoSox-Red, Rhodamine 123 and Transmission electron microscopy (TEM) measured Fe2+, reactive oxygen species (ROS), mitochondrial membrane potential and the mitochondrial morphology in GC cells, respectively. Serum hormone levels; organ indices; malondialdehyde, superoxide dismutase, and glutathione analyses; and western blotting were performed to examine ferroptosis's role in vitro. Molecular docking simulation was evaluated the interaction between FER-1 and GPX4 or FER-1 and NRF2. Molecular docking simulations were conducted to evaluate the interactions between FER-1 and GPX4, as well as FER-1 and NRF2. The findings revealed that CDDP-induced ovarian toxicity involved iron accumulation, increased ROS accumulation, and mitochondrial dysfunction, leading to endocrine disruption and tissue damage in rats. These changes correlated with NRF2, HO-1, and GPX4 levels. However, FER-1 decreased the extent of ferroptosis. Thus, ferroptosis appears to be a crucial mechanism of CDDP-induced ovarian injury, with GPX4 as potential protective targets.


Assuntos
Cisplatino , Cicloexilaminas , Ferroptose , Simulação de Acoplamento Molecular , Fenilenodiaminas , Espécies Reativas de Oxigênio , Animais , Feminino , Ferroptose/efeitos dos fármacos , Cicloexilaminas/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Cisplatino/efeitos adversos , Fenilenodiaminas/farmacologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Ovário/efeitos dos fármacos , Ovário/metabolismo , Ovário/patologia , Insuficiência Ovariana Primária/induzido quimicamente , Insuficiência Ovariana Primária/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Ratos Sprague-Dawley , Modelos Animais de Doenças , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo
2.
Nat Commun ; 15(1): 7611, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39218970

RESUMO

The development of functional neurons is a complex orchestration of multiple signaling pathways controlling cell proliferation and differentiation. Because the balance of antioxidants is important for neuronal survival and development, we hypothesized that ferroptosis must be suppressed to gain neurons. We find that removal of antioxidants diminishes neuronal development and laminar organization of cortical organoids, which is fully restored when ferroptosis is inhibited by ferrostatin-1 or when neuronal differentiation occurs in the presence of vitamin A. Furthermore, iron-overload-induced developmental growth defects in C. elegans are ameliorated by vitamin E and A. We determine that all-trans retinoic acid activates the Retinoic Acid Receptor, which orchestrates the expression of anti-ferroptotic genes. In contrast, retinal and retinol show radical-trapping antioxidant activity. Together, our study reveals an unexpected function of vitamin A in coordinating the expression of essential cellular gatekeepers of ferroptosis, and demonstrates that suppression of ferroptosis by radical-trapping antioxidants or by vitamin A is required to obtain mature neurons and proper laminar organization in cortical organoids.


Assuntos
Antioxidantes , Caenorhabditis elegans , Ferroptose , Neurônios , Vitamina A , Animais , Ferroptose/efeitos dos fármacos , Vitamina A/farmacologia , Vitamina A/metabolismo , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/efeitos dos fármacos , Antioxidantes/farmacologia , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/citologia , Cicloexilaminas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Vitamina E/farmacologia , Receptores do Ácido Retinoico/metabolismo , Receptores do Ácido Retinoico/genética , Tretinoína/farmacologia , Organoides/efeitos dos fármacos , Organoides/metabolismo , Neurogênese/efeitos dos fármacos , Camundongos , Humanos , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Transdução de Sinais/efeitos dos fármacos , Fenilenodiaminas
3.
Sci Rep ; 14(1): 20180, 2024 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-39215144

RESUMO

Spinal cord injury (SCI) results in irreversible neurological impairment. After SCI, Ferritinophagy-induced free iron released from ferritin can lead to extensive lipid peroxidation and aggravate neurological damage. NRF2/HO-1 pathway is to endow cells with a protective effect against oxidative stress, and it plays an important role in the transcriptional activation of a series of antioxidant and detoxification genes. UAMC-3203 is a ferrostatin-1(Fer-1) analogue with better solubility and stability, which can more effectively inhibit ferroptosis after SCI. A rat SCI model was constructed, and the recovery of motor function was observed after treatment with UAMC-3203. ELISA was employed to assess the impact of UAMC-3203 on inflammation-related factors, while immunofluorescence was utilized to investigate the influence of UAMC-3203 on neuronal count as well as the activation of astrocytes and microglia/macrophages. Malondialdehyde (MDA) were detected to reflect the level of oxidation products. Western blot analysis was used to measure the level of ferroptosis markers and the expression of NRF2/HO-1. Our findings demonstrate that UAMC-3203 inhibits the production of reactive oxygen species (ROS) and lipid peroxides, preventing ferroptosis and reducing neuronal degeneration. Additionally, UAMC-3203 suppresses astrocyte proliferation and microglia/macrophage activation, as well as the release of ferroptosis-related inflammatory factors. These combined effects contribute to the preservation of spinal cord tissue and the facilitation of motor function recovery. UAMC-3203 maybe inhibit ferroptosis after SCI to promote functional recovery.


Assuntos
Ferroptose , Fator 2 Relacionado a NF-E2 , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal , Animais , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/patologia , Ferroptose/efeitos dos fármacos , Ratos , Recuperação de Função Fisiológica/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Modelos Animais de Doenças , Masculino , Cicloexilaminas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Fenilenodiaminas/farmacologia , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Microglia/metabolismo , Microglia/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Heme Oxigenase (Desciclizante)
4.
ACS Chem Biol ; 19(9): 1883-1887, 2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39116319

RESUMO

Lipid peroxidation, the key step in the ferroptosis process, requires the oxidation of the double bonds of phospholipids in cellular membrane structures. Current research on ferroptosis mechanisms and new drug development has focused on naturally occurring phospholipids with internal double bonds. However, whether unnatural terminal double bonds can be involved in ferroptosis remains to be elucidated. In this study, we introduced terminal double bonds at the sn-2 position of phospholipids (Terminal Olefin Fatty Acids, TOFA) and discovered that these artificial phospholipids can kill cells alone, without ferroptosis inducers, and can be inhibited only by some ferroptosis inhibitors, such as ferrostatin-1, liproxstatin-1, alpha-tocopherol, but not deferoxamine mesylate. Our results reveal that phospholipids with terminal double bonds can participate in ferroptosis through an atypical mechanism. Moreover, further mechanistic studies could confirm that controlling the double bond position could be useful to maneuver ferroptosis and develop new drugs.


Assuntos
Ácidos Graxos , Ferroptose , Plasmalogênios , Ferroptose/efeitos dos fármacos , Humanos , Ácidos Graxos/química , Plasmalogênios/metabolismo , Plasmalogênios/química , Plasmalogênios/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Cicloexilaminas/farmacologia , Cicloexilaminas/química , Cicloexilaminas/síntese química , Fenilenodiaminas/farmacologia , Fenilenodiaminas/química , alfa-Tocoferol/farmacologia , alfa-Tocoferol/síntese química , alfa-Tocoferol/química , Quinoxalinas , Compostos de Espiro
5.
FASEB J ; 38(15): e23850, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-39091212

RESUMO

Atherosclerosis is a leading cause of cardiovascular diseases (CVDs), often resulting in major adverse cardiovascular events (MACEs), such as myocardial infarction and stroke due to the rupture or erosion of vulnerable plaques. Ferroptosis, an iron-dependent form of cell death, has been implicated in the development of atherosclerosis. Despite its involvement in CVDs, the specific role of ferroptosis in atherosclerotic plaque stability remains unclear. In this study, we confirmed the presence of ferroptosis in unstable atherosclerotic plaques and demonstrated that the ferroptosis inhibitor ferrostatin-1 (Fer-1) stabilizes atherosclerotic plaques in apolipoprotein E knockout (Apoe-/-) mice. Using bioinformatic analysis combining RNA sequencing (RNA-seq) with single-cell RNA sequencing (scRNA-seq), we identified Yes-associated protein 1 (YAP1) as a potential key regulator of ferroptosis in vascular smooth muscle cells (VSMCs) of unstable plaques. In vitro, we found that YAP1 protects against oxidized low-density lipoprotein (oxLDL)-induced ferroptosis in VSMCs. Mechanistically, YAP1 exerts its anti-ferroptosis effects by regulating the expression of glutaminase 1 (GLS1) to promote the synthesis of glutamate (Glu) and glutathione (GSH). These findings establish a novel mechanism where the inhibition of ferroptosis promotes the stabilization of atherosclerotic plaques through the YAP1/GLS1 axis, attenuating VSMC ferroptosis. Thus, targeting the YAP1/GLS1 axis to suppress VSMC ferroptosis may represent a novel strategy for preventing and treating unstable atherosclerotic plaques.


Assuntos
Ferroptose , Músculo Liso Vascular , Placa Aterosclerótica , Proteínas de Sinalização YAP , Animais , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Camundongos , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Proteínas de Sinalização YAP/metabolismo , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/genética , Camundongos Knockout , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Fenilenodiaminas/farmacologia , Cicloexilaminas/farmacologia , Apolipoproteínas E/metabolismo , Apolipoproteínas E/genética
6.
BMC Gastroenterol ; 24(1): 245, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39090535

RESUMO

BACKGROUND: Ferroptosis is a newly recognized form of regulatory cell death characterized by severe lipid peroxidation triggered by iron overload and the production of reactive oxygen species (ROS). However, the role of ferroptosis in severe acute pancreatitis(SAP) has not been fully elucidated. METHODS: We established four severe acute pancreatitis models of rats including the sham control group, the SAP group, the Fer -1-treated SAP (SAP + Fer-1) group, the 3-MA-treated SAP (SAP + 3-MA) group. The SAP group was induced by retrograde injection of sodium taurocholate into the pancreatic duct. The other two groups were intraperitoneally injected with ferroptosis inhibitor (Fer-1) and autophagy inhibitor (3-MA), respectively. The model of severe acute pancreatitis with amylase crest-related inflammatory factors was successfully established. Then we detected ferroptosis (GPX4, SLC7A1 etc.) and autophagy-related factors (LC3II, p62 ect.) to further clarify the relationship between ferroptosis and autophagy. RESULTS: Our study found that ferroptosis occurs during the development of SAP, such as iron and lipid peroxidation in pancreatic tissues, decreased levels of reduced glutathione peroxidase 4 (GPX 4) and glutathione (GSH), and increased malondialdehyde(MDA) and significant mitochondrial damage. In addition, ferroptosis related proteins such as GPX4, solute carrier family 7 member 11(SLC7A11) and ferritin heavy chain 1(FTH1) were significantly decreased. Next, the pathogenesis of ferroptosis in SAP was studied. First, treatment with the ferroptosis inhibitor ferrostatin-1(Fer-1) significantly alleviated ferroptosis in SAP. Interestingly, autophagy occurs during the pathogenesis of SAP, and autophagy promotes the occurrence of ferroptosis in SAP. Moreover, 3-methyladenine (3-MA) inhibition of autophagy can significantly reduce iron overload and ferroptosis in SAP. CONCLUSIONS: Our results suggest that ferroptosis is a novel pathogenesis of SAP and is dependent on autophagy. This study provides a new theoretical basis for the study of SAP.


Assuntos
Autofagia , Modelos Animais de Doenças , Ferroptose , Peroxidação de Lipídeos , Pancreatite , Ratos Sprague-Dawley , Animais , Pancreatite/metabolismo , Pancreatite/patologia , Ratos , Masculino , Adenina/análogos & derivados , Adenina/farmacologia , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Ácido Taurocólico , Cicloexilaminas/farmacologia , Pâncreas/patologia , Pâncreas/metabolismo , Fenilenodiaminas/farmacologia , Malondialdeído/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Doença Aguda , Glutationa/metabolismo , Ferro/metabolismo
7.
Sci Rep ; 14(1): 19877, 2024 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-39191890

RESUMO

Osteoarthritis (OA) is a common joint disease associated with the aging of the population, and it reduces the quality of life of patients. It is characterized by the destruction of articular cartilage and the secretion of inflammatory cytokines. Owing to the unclear pathogenesis of OA, current treatment methods have significant limitations. Oxidative stress has been revealed to play an important role in the development of OA. Our experiments indicated that the levels of GSH decreased and the level of MDA increased in chondrocytes, which induced ferroptosis in chondrocytes in OA. We also revealed that ferroptosis was the main mechanism of cartilage destruction caused by the addition of the ferroptosis activator erastin and the ferroptosis inhibitor ferrostatin-1. NOX1 is the main modulator of oxidative stress by increasing the generation of reactive oxidative species (ROS). We suppressed the expression of NOX1 in chondrocytes through cell transfection. The expression of collagen II and MMP13, and the secretion of IL-1ß and TNF-α were reversed. An increase in the mitochondrial membrane potential and a decrease in the level of intracellular ROS indicate an improvement in oxidative damage. Additionally, we determined the effect of the Nrf2/HO-1 pathway on NOX1-mediated chondrocyte injury. We found that NOX1 inhibited the expression of Nrf2/HO-1, but the activation of Nrf2 improved the oxidative damage to chondrocytes in vivo and vitro. This study revealed that NOX1-mediated oxidative stress induces chondrocyte ferroptosis by inhibiting the Nrf2/HO-1 pathway. Our findings contribute to revealing the pathogenesis of OA, providing targets for drug design and optimizing the clinical treatment of OA.


Assuntos
Condrócitos , Ferroptose , Heme Oxigenase-1 , NADPH Oxidase 1 , Fator 2 Relacionado a NF-E2 , Osteoartrite , Estresse Oxidativo , Espécies Reativas de Oxigênio , Transdução de Sinais , Condrócitos/metabolismo , Ferroptose/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Animais , NADPH Oxidase 1/metabolismo , NADPH Oxidase 1/genética , Heme Oxigenase-1/metabolismo , Camundongos , Osteoartrite/metabolismo , Osteoartrite/patologia , Espécies Reativas de Oxigênio/metabolismo , Cicloexilaminas/farmacologia , Masculino , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Humanos , Proteínas de Membrana , Fenilenodiaminas
8.
Int J Nanomedicine ; 19: 8253-8270, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39157734

RESUMO

Background: Myocardial infarction (MI) is characterized by irreversible cardiomyocyte death resulting from an inadequate supply of oxygenated blood to the myocardium. Recent studies have indicated that ferroptosis, a form of regulated cell death, exacerbates myocardial injury during MI. Concurrently, the upregulation of CD47 on the surface of damaged myocardium following MI impairs the clearance of dead cells by macrophages, thereby hindering efferocytosis. In this context, simultaneously inhibiting ferroptosis and enhancing efferocytosis may represent a promising strategy to mitigate myocardial damage post-MI. Methods: In this study, we engineered platelet membrane-coated hollow mesoporous silicon nanoparticles (HMSN) to serve as a drug delivery system, encapsulating ferroptosis inhibitor, Ferrostatin-1, along with an anti-CD47 antibody. We aimed to assess the potential of these nanoparticles (designated as Fer-aCD47@PHMSN) to specifically target the site of MI and evaluate their efficacy in reducing cardiomyocyte death and inflammation. Results: The platelet membrane coating on the nanoparticles significantly enhanced their ability to successfully target the site of myocardial infarction (MI). Our findings demonstrate that treatment with Fer-aCD47@PHMSN resulted in a 38.5% reduction in cardiomyocyte ferroptosis under hypoxia, indicated by decreased lipid peroxidation and increased in vitro. Additionally, Fer-aCD47@PHMSN improved cardiomyocyte efferocytosis by approximately 15% in vitro. In MI mice treated with Fer-aCD47@PHMSN, we observed a substantial reduction in cardiomyocyte death (nearly 30%), decreased inflammation, and significant improvement in cardiac function. Conclusion: Our results demonstrated that the cooperation between the two agents induced anti-ferroptosis effects and enhanced dead cardiomyocyte clearance by macrophage as well as anti-inflammation effects. Thus, our nanoparticle Fer-aCD47@PHMSN provides a new therapeutic strategy for targeted therapy of MI.


Assuntos
Antígeno CD47 , Ferroptose , Infarto do Miocárdio , Miócitos Cardíacos , Nanopartículas , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Ferroptose/efeitos dos fármacos , Animais , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Nanopartículas/química , Camundongos , Antígeno CD47/metabolismo , Fagocitose/efeitos dos fármacos , Cicloexilaminas/farmacologia , Cicloexilaminas/química , Masculino , Fenilenodiaminas/farmacologia , Fenilenodiaminas/química , Macrófagos/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Portadores de Fármacos/química , Humanos , Eferocitose
9.
Otol Neurotol ; 45(8): 947-953, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39072683

RESUMO

BACKGROUND: Gentamicin is a commonly used aminoglycoside antibiotic, with ototoxicity as a significant side effect. Ferroptosis, an iron-dependent form of cell death, has been implicated in a variety of disorders. Whether ferroptosis impacts gentamicin ototoxicity is not yet known. The current work used an in-vitro model to examine the influence of gentamicin-induced ferroptosis on cochlear hair cell damage and probable molecular biological pathways. METHODS: House Ear Institute-Organ of Corti 1 (HEI-OC1) cells were treated with different concentrations of gentamicin for 24 hours, with or without ferrostatin-1 pretreatment, to observe gentamicin-induced ferroptosis. The role of p53/solute carrier family 7 member 11 (SLC7A11)/glutathione peroxidase 4 (GPX4) signaling in gentamicin-induced ferroptosis was explored by pretreating cells with the p53 inhibitor pifithrin-α (PFT-α). We investigated the effect of gentamicin on cells by assessing cell viability. Cellular proteins were isolated and Western blots were performed to detect changes in the expression of p53, SLC7A11, and GPX4. Fluorescence staining was used to assess levels of reactive oxygen species. An enzymatic detection kit was used to detect glutathione, Fe, and malondialdehyde markers. RESULTS: Gentamicin reduced cell viability, glutathione content, and SLC7A11 and GPX4 protein levels, and increased levels of p53 protein, reactive oxygen species, malondialdehyde, and Fe. These effects were largely blocked by pretreatment with ferrostatin-1. Pretreatment with the p53 inhibitor PFT-α prevented the gentamicin-induced reduction in SLC7A11 and GPX4, which alleviated several features of ferroptosis including glutathione depletion, iron overload, and lipid peroxidation build-up. CONCLUSION: Gentamicin induces ferroptosis in the HEI-OC1 cell line, and the mechanism may be related to the p53/SLC7A11/GPX4 signaling pathway.


Assuntos
Sistema y+ de Transporte de Aminoácidos , Antibacterianos , Ferroptose , Gentamicinas , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Transdução de Sinais , Proteína Supressora de Tumor p53 , Ferroptose/efeitos dos fármacos , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Gentamicinas/toxicidade , Gentamicinas/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Sistema y+ de Transporte de Aminoácidos/metabolismo , Camundongos , Transdução de Sinais/efeitos dos fármacos , Antibacterianos/farmacologia , Antibacterianos/toxicidade , Linhagem Celular , Células Ciliadas Auditivas/efeitos dos fármacos , Células Ciliadas Auditivas/metabolismo , Órgão Espiral/efeitos dos fármacos , Órgão Espiral/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Cicloexilaminas/farmacologia , Glutationa Peroxidase/metabolismo , Fenilenodiaminas
10.
Ecotoxicol Environ Saf ; 281: 116680, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38964057

RESUMO

Inhaling polyhexamethylene guanidine (PHMG) aerosol, a broad-spectrum disinfectant, can lead to severe pulmonary fibrosis. Ferroptosis, a form of programmed cell death triggered by iron-dependent lipid peroxidation, is believed to play a role in the chemical-induced pulmonary injury. This study aimed to investigate the mechanism of ferroptosis in the progression of PHMG-induced pulmonary fibrosis. C57BL/6 J mice and the alveolar type II cell line MLE-12 were used to evaluate the toxicity of PHMG in vivo and in vitro, respectively. The findings indicated that iron deposition was observed in PHMG induced pulmonary fibrosis mouse model and ferroptosis related genes have changed after 8 weeks PHMG exposure. Additionally, there were disturbances in the antioxidant system and mitochondrial damage in MLE-12 cells following a 12-hour treatment with PHMG. Furthermore, the study observed an increase in lipid peroxidation and a decrease in GPX4 activity in MLE-12 cells after exposure to PHMG. Moreover, pretreatment with the ferroptosis inhibitors Ferrostatin-1 (Fer-1) and Liproxstatin-1 (Lip-1) not only restored the antioxidant system and GPX4 activity but also mitigated lipid peroxidation. Current data exhibit the role of ferroptosis pathway in PHMG-induced pulmonary fibrosis and provide a potential target for future treatment.


Assuntos
Ferroptose , Guanidinas , Peroxidação de Lipídeos , Camundongos Endogâmicos C57BL , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Fibrose Pulmonar , Animais , Ferroptose/efeitos dos fármacos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Camundongos , Peroxidação de Lipídeos/efeitos dos fármacos , Linhagem Celular , Guanidinas/toxicidade , Guanidinas/farmacologia , Masculino , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/patologia , Cicloexilaminas/farmacologia , Fenilenodiaminas , Quinoxalinas , Compostos de Espiro
11.
Biomed Environ Sci ; 37(6): 617-627, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38988112

RESUMO

Objective: The aim of this study was to explore the role and mechanism of ferroptosis in SiO 2-induced cardiac injury using a mouse model. Methods: Male C57BL/6 mice were intratracheally instilled with SiO 2 to create a silicosis model. Ferrostatin-1 (Fer-1) and deferoxamine (DFO) were used to suppress ferroptosis. Serum biomarkers, oxidative stress markers, histopathology, iron content, and the expression of ferroptosis-related proteins were assessed. Results: SiO 2 altered serum cardiac injury biomarkers, oxidative stress, iron accumulation, and ferroptosis markers in myocardial tissue. Fer-1 and DFO reduced lipid peroxidation and iron overload, and alleviated SiO 2-induced mitochondrial damage and myocardial injury. SiO 2 inhibited Nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream antioxidant genes, while Fer-1 more potently reactivated Nrf2 compared to DFO. Conclusion: Iron overload-induced ferroptosis contributes to SiO 2-induced cardiac injury. Targeting ferroptosis by reducing iron accumulation or inhibiting lipid peroxidation protects against SiO 2 cardiotoxicity, potentially via modulation of the Nrf2 pathway.


Assuntos
Modelos Animais de Doenças , Ferroptose , Sobrecarga de Ferro , Camundongos Endogâmicos C57BL , Miócitos Cardíacos , Dióxido de Silício , Silicose , Animais , Ferroptose/efeitos dos fármacos , Masculino , Camundongos , Sobrecarga de Ferro/metabolismo , Dióxido de Silício/toxicidade , Silicose/metabolismo , Silicose/tratamento farmacológico , Silicose/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Desferroxamina/farmacologia , Fenilenodiaminas/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Estresse Oxidativo/efeitos dos fármacos , Ferro/metabolismo , Cicloexilaminas/farmacologia
12.
J Biochem Mol Toxicol ; 38(8): e23791, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39082238

RESUMO

Liver injury caused by acetaminophen (APAP) overdose is the leading cause of acute liver failure in western countries. The mode of APAP-induced cell death has been controversially discussed with ferroptosis emerging as a more recent hypothesis. Ferroptosis is characterized by ferrous iron-catalyzed lipid peroxidation (LPO) causing cell death, which can be prevented by the lipophilic antioxidants ferrostatin-1 and UAMC-3203. To assess the efficacy of these ferroptosis inhibitors, we used two murine models of APAP hepatotoxicity, APAP overdose alone or in combination with FeSO4 in fasted male C57BL/6J mice. APAP triggered severe liver injury in the absence of LPO measured as hepatic malondialdehyde (MDA) levels. In contrast, ferrous iron co-treatment aggravated APAP-induced liver injury and caused extensive LPO. Standard doses of ferrostatin-1 did not affect MDA levels or the injury in both models. In contrast, UAMC-3203 partially protected in both models and reduced LPO in the presence of ferrous iron. However, UAMC-3203 attenuated the translocation of phospho-JNK through downregulation of the mitochondrial anchor protein Sab resulting in reduced mitochondrial dysfunction and liver injury. Thus, APAP toxicity does not involve ferroptosis under normal conditions. The lack of effects of ferroptosis inhibitors in the pathophysiology indicates that ferroptosis signaling pathways are not relevant therapeutic targets.


Assuntos
Acetaminofen , Doença Hepática Induzida por Substâncias e Drogas , Modelos Animais de Doenças , Ferroptose , Camundongos Endogâmicos C57BL , Animais , Acetaminofen/efeitos adversos , Acetaminofen/toxicidade , Ferroptose/efeitos dos fármacos , Masculino , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Camundongos , Cicloexilaminas/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Fenilenodiaminas/farmacologia
13.
Exp Neurol ; 379: 114849, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38857748

RESUMO

Cerebral ischemic stroke is a serious disease with high mortality and disability rates. However, few neuroprotective drugs have been used for ischemic stroke in the clinic. Two main reasons may be responsible for this failure: difficulty in penetrating the blood-brain barrier (BBB) and easily inactivated in the blood circulation. Ferroptosis, a lipid oxidation-related cell death, plays significant roles in cerebral ischemia-reperfusion injury. We utilized RVG29, a peptide derived from Rabies virus glycoprotein, to obtain BBB-targeted lipid nanoparticles (T-LNPs) in order to investigate whether T-LNPs improved the neuroprotective effects of Ferrostatin-1 (Fer1, an inhibitor of ferroptosis) against cerebral ischemic damage. T-LNPs significantly increased BBB penetration following oxygen/glucose deprivation exposure in an in vitro BBB model and enhanced the fluorescence distribution in brain tissues at 6 h post-administration in a cerebral ischemic murine model. Moreover, T-LNPs encapsulated Fer1 (T-LNPs-Fer1) significantly enhanced the inhibitory effects of Fer1 on ferroptosis by maintaining the homeostasis of NADPH oxidase 4 (NOX4) and glutathione peroxidase 4 (GPX4) signals in neuronal cells after cerebral ischemia. T-LNPs-Fer1 significantly suppressed oxidative stress [heme oxygenase-1 expression and malondialdehyde (the product of lipid ROS reaction)] in neurons and alleviated ischemia-induced neuronal cell death, compared to Fer1 alone without encapsulation. Furthermore, T-LNPs-Fer1 significantly reduced cerebral infarction and improved behavior functions compared to Fer1-treated cerebral ischemic mice after 45-min ischemia/24-h reperfusion. These findings showed that the T-LNPs helped Fer1 penetrate the BBB and improved the neuroprotection of Fer1 against cerebral ischemic damage in experimental stroke, providing a feasible translational strategy for the development of clinical drugs for the treatment of ischemic stroke.


Assuntos
Barreira Hematoencefálica , Cicloexilaminas , Nanopartículas , Fármacos Neuroprotetores , Fenilenodiaminas , Animais , Camundongos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Fármacos Neuroprotetores/farmacologia , Nanopartículas/administração & dosagem , Masculino , Cicloexilaminas/farmacologia , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/patologia , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Isquemia Encefálica/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Ferroptose/efeitos dos fármacos , Glicoproteínas , Lipossomos , Fragmentos de Peptídeos , Proteínas Virais
14.
Chem Biol Interact ; 399: 111121, 2024 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-38944326

RESUMO

The toxicity of silica nanoparticles (SiNPs) to lung is known. We previously demonstrated that exposure to SiNPs promoted pulmonary impairments, but the precise pathogenesis remains elucidated. Ferroptosis has now been identified as a unique form of oxidative cell death, but whether it participated in SiNPs-induced lung injury remains unclear. In this work, we established a rat model with sub-chronic inhalation exposure of SiNPs via intratracheal instillation, and conducted histopathological examination, iron detection, and ferroptosis-related lipid peroxidation and protein assays. Moreover, we evaluated the effect of SiNPs on epithelial ferroptosis, possible mechanisms using in vitro-cultured human bronchial epithelial cells (16HBE), and also assessed the ensuing impact on fibroblast activation for fibrogenesis. Consequently, fibrotic lesions occurred in the rat lungs, concomitantly by enhanced lipid peroxidation, iron overload, and ferroptosis. Consistently, the in vitro data showed SiNPs triggered oxidative stress and caused the accumulation of lipid peroxides, resulting in ferroptosis. Importantly, the mechanistic investigation revealed miR-21-5p as a key player in the epithelial ferroptotic process induced by SiNPs via targeting GCLM for GSH depletion. Of note, ferrostatin-1 could greatly suppress ferroptosis and alleviate epithelial injury and ensuing fibroblast activation by SiNPs. In conclusion, our findings first revealed SiNPs triggered epithelial ferroptosis through miR-21-5p/GCLM signaling and thereby promoted fibroblast activation for fibrotic lesions, and highlighted the therapeutic potential of inhibiting ferroptosis against lung impairments upon SiNPs exposure.


Assuntos
Células Epiteliais , Ferroptose , Pulmão , MicroRNAs , Nanopartículas , Transdução de Sinais , Dióxido de Silício , Ferroptose/efeitos dos fármacos , Animais , Humanos , MicroRNAs/metabolismo , MicroRNAs/genética , Ratos , Nanopartículas/química , Transdução de Sinais/efeitos dos fármacos , Pulmão/patologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Masculino , Glutamato-Cisteína Ligase/metabolismo , Glutamato-Cisteína Ligase/genética , Linhagem Celular , Ratos Sprague-Dawley , Estresse Oxidativo/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Glutationa/metabolismo , Cicloexilaminas/farmacologia , Fenilenodiaminas
15.
Kaohsiung J Med Sci ; 40(8): 710-721, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38837857

RESUMO

The proinflammatory properties of high-mobility group box protein 1 (HMGB1) in sepsis have been extensively studied. This study aimed to investigate the impact of HMGB1 on ferroptosis and its molecular mechanism in sepsis-induced acute lung injury (ALI). A septic mouse model was established using the cecal ligation and puncture method. Blocking HMGB1 resulted in improved survival rates, reduced lung injury, decreased levels of ferroptosis markers (reactive oxygen species, malondialdehyde, and Fe2+), and enhanced antioxidant enzyme activities (superoxide dismutase and catalase) in septic mice. In addition, knockdown of HMGB1 reduced cellular permeability, ferroptosis markers, and raised antioxidant enzyme levels in lipopolysaccharide (LPS)-stimulated MLE-12 cells. Silencing of HMGB1 led to elevations in the expressions of ferroptosis core-regulators in LPS-treated MLE-12 cells, such as solute carrier family 7 member 11 (SLC7A11), solute carrier family 3 member A2 (SLC3A2), and glutathione peroxidase 4. Furthermore, blocking HMGB1 did not alter ferroptosis, oxidative stress-related changes, and permeability in LPS-treated MLE-12 cells that were pretreated with ferrostatin-1 (a ferroptosis inhibitor). HMGB1 inhibition also led to elevated expressions of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream targets, heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase 1 (NQO1) in LPS-treated MLE-12 cells and lung tissues from septic mice. The Nrf2-specific inhibitor ML385 reversed the effects of HMGB1 silencing on ferroptosis and cell permeability in LPS-treated MLE-12 cells. Our findings indicated that the inhibition of HMGB1 restrains ferroptosis and oxidative stress, thereby alleviating sepsis-induced ALI through the activation of Nrf2 signaling.


Assuntos
Lesão Pulmonar Aguda , Ferroptose , Proteína HMGB1 , Lipopolissacarídeos , Fator 2 Relacionado a NF-E2 , Estresse Oxidativo , Sepse , Transdução de Sinais , Animais , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Proteína HMGB1/metabolismo , Proteína HMGB1/antagonistas & inibidores , Proteína HMGB1/genética , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/tratamento farmacológico , Ferroptose/efeitos dos fármacos , Sepse/complicações , Sepse/metabolismo , Sepse/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Camundongos , Lipopolissacarídeos/toxicidade , Transdução de Sinais/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Cicloexilaminas/farmacologia , Linhagem Celular , Modelos Animais de Doenças , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/antagonistas & inibidores , NAD(P)H Desidrogenase (Quinona)/metabolismo , NAD(P)H Desidrogenase (Quinona)/genética , Sistema y+ de Transporte de Aminoácidos
16.
Int Immunopharmacol ; 138: 112579, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-38944951

RESUMO

Obstructive sleep apnea, typically characterized by chronic intermittent hypoxia (CIH), is linked to cognitive dysfunction in children. Ferroptosis, a novel form of cell death characterized by lethal iron accumulation and lipid peroxidation, is implicated in neurodegenerative diseases and ischemia-reperfusion injuries. Nevertheless, its contribution to CIH-induced cognitive dysfunction and its interaction with endoplasmic reticulum stress (ERS) remain uncertain. In this study, utilizing a CIH model in 4-week-old male mice, we investigated ferroptosis and its potential involvement in ERS regulation during cognitive dysfunction. Our findings indicate ferroptosis activation in prefrontal cortex neurons, leading to neuron loss, mitochondrial damage, decreased levels of GPX4, SLC7A11, FTL, and FTH, increased levels of reactive oxygen species (ROS), malondialdehyde (MDA), Fe2+, ACSL4, TFRC, along with the activation of ERS-related PERK-ATF4-CHOP pathway. Treatment with the ferroptosis inhibitor liproxstatin-1 (Lip-1) and the iron chelator deferoxamine (DFO) effectively mitigated the neuron injury and cognitive dysfunction induced by CIH, significantly reducing Fe2+ and partly restoring expression levels of ferroptosis-related proteins. Furhermore, the use of Lip-1 and DFO downregulated p-PERK, ATF4 and CHOP, and upregulated Nrf2 expression, suggesting that inhibiting ferroptosis reduce ERS and that the transcription factor Nrf2 is involved in the process. In summary, our findings indicate that cognitive impairment in CIH mice correlates with the induction of neuronal ferroptosis, facilitated by the System xc - GPX4 functional axis, lipid peroxidation, and the iron metabolism pathway, along with ferroptosis-mediated ERS in the prefrontal cortex. Nrf2 has been identified as a potential regulator of ferroptosis and ERS involved in the context of CIH.


Assuntos
Disfunção Cognitiva , Estresse do Retículo Endoplasmático , Ferroptose , Hipóxia , Neurônios , Animais , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Masculino , Hipóxia/metabolismo , Hipóxia/complicações , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Camundongos , Camundongos Endogâmicos C57BL , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Desferroxamina/farmacologia , Desferroxamina/uso terapêutico , Cicloexilaminas/farmacologia , Modelos Animais de Doenças , Espécies Reativas de Oxigênio/metabolismo , Apneia Obstrutiva do Sono/complicações , Apneia Obstrutiva do Sono/metabolismo , Humanos , Quinoxalinas , Compostos de Espiro , Sistema y+ de Transporte de Aminoácidos
17.
PeerJ ; 12: e17551, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38887622

RESUMO

Background: Keloid is a chronic proliferative fibrotic disease caused by abnormal fibroblasts proliferation and excessive extracellular matrix (ECM) production. Numerous fibrotic disorders are significantly influenced by ferroptosis, and targeting ferroptosis can effectively mitigate fibrosis development. This study aimed to investigate the role and mechanism of ferroptosis in keloid development. Methods: Keloid tissues from keloid patients and normal skin tissues from healthy controls were collected. Iron content, lipid peroxidation (LPO) level, and the mRNA and protein expression of ferroptosis-related genes including solute carrier family 7 member 11 (SLC7A11), glutathione peroxidase 4 (GPX4), transferrin receptor (TFRC), and nuclear factor erythroid 2-related factor 2 (Nrf2) were determined. Mitochondrial morphology was observed using transmission electron microscopy (TEM). Keloid fibroblasts (KFs) were isolated from keloid tissues, and treated with ferroptosis inhibitor ferrostatin-1 (fer-1) or ferroptosis activator erastin. Iron content, ferroptosis-related marker levels, LPO level, mitochondrial membrane potential, ATP content, and mitochondrial morphology in KFs were detected. Furthermore, the protein levels of α-smooth muscle actin (α-SMA), collagen I, and collagen III were measured to investigate whether ferroptosis affect fibrosis in KFs. Results: We found that iron content and LPO level were substantially elevated in keloid tissues and KFs. SLC7A11, GPX4, and Nrf2 were downregulated and TFRC was upregulated in keloid tissues and KFs. Mitochondria in keloid tissues and KFs exhibited ferroptosis-related pathology. Fer-1 treatment reduced iron content, restrained ferroptosis and mitochondrial dysfunction in KFs, Moreover, ferrostatin-1 restrained the protein expression of α-SMA, collagen I, and collagen III in KFs. Whereas erastin treatment showed the opposite results. Conclusion: Ferroptosis exists in keloid. Ferrostatin-1 restrained ECM deposition and fibrosis in keloid through inhibiting ferroptosis, and erastin induced ECM deposition and fibrosis through intensifying ferroptosis.


Assuntos
Cicloexilaminas , Ferroptose , Fibroblastos , Fibrose , Queloide , Fator 2 Relacionado a NF-E2 , Fenilenodiaminas , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Humanos , Ferroptose/efeitos dos fármacos , Queloide/patologia , Queloide/metabolismo , Queloide/tratamento farmacológico , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Cicloexilaminas/farmacologia , Fibrose/metabolismo , Fibrose/patologia , Fenilenodiaminas/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Masculino , Peroxidação de Lipídeos/efeitos dos fármacos , Feminino , Adulto , Ferro/metabolismo , Sistema y+ de Transporte de Aminoácidos/metabolismo , Sistema y+ de Transporte de Aminoácidos/genética , Receptores da Transferrina/metabolismo , Receptores da Transferrina/genética , Piperazinas/farmacologia , Actinas/metabolismo , Actinas/genética , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos
18.
J Oleo Sci ; 73(7): 991-999, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38945927

RESUMO

In this study, we evaluated the cancer cell killing activity of koji mold-derived extracts using several solvents. The koji mold lipid extract (KML) exhibited potent cytotoxicity against a human leukemia cell line. Fractionation of the KML via silica gel chromatography revealed the presence of active components in fraction (Fr.) 6. Cytotoxic effects of Fr. 6 were inhibited by the ferroptosis inhibitors, ferrostatin-1 and SRS11-92, and the iron chelator, deferoxamine. Interestingly, ferroptosis inhibitors failed to prevent the KML-induced cell death. Fr. 6 decreased the expression of glutathione peroxidase 4 (GPx4) and increased the level of peroxidized plasma membrane lipids. Furthermore, Fr. 6 decreased the intracellular glutathione levels. Overall, our results suggest that Fr. 6 included in KML induces ferroptosis in HL-60 cells.


Assuntos
Ferroptose , Glutationa , Peroxidação de Lipídeos , Oxirredução , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Humanos , Células HL-60 , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Ferroptose/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Glutationa/metabolismo , Oxirredução/efeitos dos fármacos , Desferroxamina/farmacologia , Cicloexilaminas/farmacologia , Lipídeos , Fenilenodiaminas/farmacologia , Lipídeos de Membrana/metabolismo , Quelantes de Ferro/farmacologia
19.
Neurotox Res ; 42(3): 27, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38819761

RESUMO

Early and prolonged exposure to anesthetic agents could cause neurodevelopmental disorders in children. Astrocytes, heavily outnumber neurons in the brain, are crucial regulators of synaptic formation and function during development. However, how general anesthetics act on astrocytes and the impact on cognition are still unclear. In this study, we investigated the role of ferroptosis and GPX4, a major hydroperoxide scavenger playing a pivotal role in suppressing the process of ferroptosis, and their underlying mechanism in isoflurane-induced cytotoxicity in astrocytes and cognitive impairment. Our results showed that early 6 h isoflurane anesthesia induced cognitive impairment in mice. Ferroptosis-relative genes and metabolic changes were involved in the pathological process of isoflurane-induced cytotoxicity in astrocytes. The level of GPX4 was decreased while the expression of 4-HNE and generation of ROS were elevated after isoflurane exposure. Selectively blocking ferroptosis with Fer-1 attenuated the abovementioned cytotoxicity in astrocytes, paralleling with the reverse of the changes in GPX4, ROS and 4-HNE secondary to isoflurane anesthesia. Fer-1 attenuated the cognitive impairment induced by prolonged isoflurane exposure. Thus, ferroptosis conduced towards isoflurane-induced cytotoxicity in astrocytes via suppressing GPX4 and promoting lipid peroxidation. Fer-1 was expected to be an underlying intervention for the neurotoxicity induced by isoflurane in the developing brain, and to alleviate cognitive impairment in neonates.


Assuntos
Animais Recém-Nascidos , Astrócitos , Disfunção Cognitiva , Cicloexilaminas , Ferroptose , Isoflurano , Fenilenodiaminas , Animais , Camundongos , Anestésicos Inalatórios/toxicidade , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/prevenção & controle , Disfunção Cognitiva/metabolismo , Ferroptose/efeitos dos fármacos , Ferroptose/fisiologia , Isoflurano/toxicidade , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Cicloexilaminas/farmacologia , Fenilenodiaminas/farmacologia
20.
J Reprod Immunol ; 164: 104260, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38761507

RESUMO

INTRODUCTION: Recent studies have suggested the involvement of ferroptosis in preterm birth. Despite compelling evidence, the underlying mechanism remains unknown. This investigation aimed to determine the therapeutic effects of Ferrostatin-1 (Fer-1), an inhibitor of ferroptosis, in preterm birth and fetal brain injury. METHODS: Human placenta samples and clinical data of participants were collected to ascertain whether placental ferroptosis was associated with preterm birth. Lipopolysaccharide (LPS)-induced preterm birth mouse model was used to examine the protective effects of Fer-1 on preterm birth. Fetal brain tissues and offspring mice at 5 and 8 weeks were studied to determine the effects of Fer-1 on the cognitive function of offspring. RESULTS: We examined the mechanism of spontaneous preterm birth and discovered that placental ferroptosis was associated with preterm birth. Fer-1 inhibited preterm birth by ameliorating placental ferroptosis and maternal inflammation, thus improving LPS-induced intrauterine inflammation to maintain pregnancy. Antenatal administration of Fer-1 prevented LPS-induced fetal brain damage in the acute phase and improved long-term neurodevelopmental impairments by improving placental neuroendocrine signaling and maintaining placental function. CONCLUSION: Fer-1 inhibited preterm birth and fetal brain injury by inhibiting maternal inflammation and improving placental function. Our findings provide a novel therapeutic strategy for preterm birth.


Assuntos
Lesões Encefálicas , Cicloexilaminas , Ferroptose , Lipopolissacarídeos , Fenilenodiaminas , Placenta , Nascimento Prematuro , Animais , Feminino , Gravidez , Camundongos , Nascimento Prematuro/imunologia , Nascimento Prematuro/prevenção & controle , Humanos , Cicloexilaminas/farmacologia , Cicloexilaminas/administração & dosagem , Placenta/patologia , Placenta/imunologia , Placenta/efeitos dos fármacos , Lesões Encefálicas/prevenção & controle , Lesões Encefálicas/imunologia , Lesões Encefálicas/etiologia , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/patologia , Fenilenodiaminas/farmacologia , Fenilenodiaminas/administração & dosagem , Lipopolissacarídeos/imunologia , Ferroptose/efeitos dos fármacos , Inflamação/imunologia , Inflamação/tratamento farmacológico , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...