Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 295
1.
Exp Dermatol ; 33(6): e15114, 2024 Jun.
Article En | MEDLINE | ID: mdl-38853773

Ferroptosis is a novel type of cell death that is dependent on lipid peroxidation and iron accumulation, which distinguishes it from other types of programmed cell death. Current research indicates a significant association between ferroptosis and various pathological conditions, including cancer, neurological disorders, and cardiovascular diseases, albeit with a relatively unexplored role in dermatological afflictions. This paper elaborates on the mechanisms and signalling pathways of ferroptosis, summarizing the recent studies on ferroptosis and its related factors in dermatosis. Our objective is to shed light on novel perspectives and therapeutic strategies for dermatosis, enhancing the understanding of this under-researched area through this comprehensive review.


Ferroptosis , Signal Transduction , Skin Diseases , Ferroptosis/physiology , Humans , Skin Diseases/metabolism , Iron/metabolism , Lipid Peroxidation , Animals
2.
Brain Res Bull ; 213: 110991, 2024 Jul.
Article En | MEDLINE | ID: mdl-38823725

Neurodegenerative diseases such as Parkinson's disease (PD) have complex pathogenetic mechanisms. Genetic, age, and environmental factors are all related to PD. Due to the unclear pathogenesis of PD and the lack of effective cure methods, it is urgent to find new targets for treating PD patients. Ferroptosis is a form of cell death that is reliant on iron and exhibits distinct morphological and mechanistic characteristics compared to other types of cell death. It encompasses a range of biological processes, including iron/lipid metabolism and oxidative stress. In recent years, research has found that ferroptosis plays a crucial role in the pathophysiological processes of neurodegenerative diseases and stroke. Therefore, ferroptosis is also closely related to PD, This article reviews the core mechanisms of ferroptosis and elucidates the correlation between PD and ferroptosis. In addition, new compounds that have emerged in recent years to exert anti PD effects by inhibiting the ferroptosis signaling pathway were summarized. I hope to further elaborate the relationship between ferroptosis and PD through the review of this article, and provide new strategies for developing PD treatments targeting ferroptosis.


Ferroptosis , Parkinson Disease , Ferroptosis/physiology , Humans , Parkinson Disease/metabolism , Parkinson Disease/pathology , Animals , Iron/metabolism , Oxidative Stress/physiology , Signal Transduction/physiology , Lipid Metabolism/physiology
3.
BMC Biol ; 22(1): 134, 2024 Jun 10.
Article En | MEDLINE | ID: mdl-38858683

BACKGROUND: Inherited retinal dystrophies (IRDs) are a group of debilitating visual disorders characterized by the progressive degeneration of photoreceptors, which ultimately lead to blindness. Among the causes of this condition, mutations in the PCYT1A gene, which encodes the rate-limiting enzyme responsible for phosphatidylcholine (PC) de novo synthesis via the Kennedy pathway, have been identified. However, the precise mechanisms underlying the association between PCYT1A mutations and IRDs remain unclear. To address this knowledge gap, we focused on elucidating the functions of PCYT1A in the retina. RESULTS: We found that PCYT1A is highly expressed in Müller glial (MG) cells in the inner nuclear layer (INL) of the retina. Subsequently, we generated a retina-specific knockout mouse model in which the Pcyt1a gene was targeted (Pcyt1a-RKO or RKO mice) to investigate the molecular mechanisms underlying IRDs caused by PCYT1A mutations. Our findings revealed that the deletion of Pcyt1a resulted in retinal degenerative phenotypes, including reduced scotopic electroretinogram (ERG) responses and progressive degeneration of photoreceptor cells, accompanied by loss of cells in the INL. Furthermore, through proteomic and bioinformatic analyses, we identified dysregulated retinal fatty acid metabolism and activation of the ferroptosis signalling pathway in RKO mice. Importantly, we found that PCYT1A deficiency did not lead to an overall reduction in PC synthesis within the retina. Instead, this deficiency appeared to disrupt free fatty acid metabolism and ultimately trigger ferroptosis. CONCLUSIONS: This study reveals a novel mechanism by which mutations in PCYT1A contribute to the development of IRDs, shedding light on the interplay between fatty acid metabolism and retinal degenerative diseases, and provides new insights into the treatment of IRDs.


Fatty Acids , Ferroptosis , Mice, Knockout , Retina , Animals , Mice , Retina/metabolism , Ferroptosis/physiology , Ferroptosis/genetics , Fatty Acids/metabolism , Choline-Phosphate Cytidylyltransferase/genetics , Choline-Phosphate Cytidylyltransferase/metabolism , Retinal Dystrophies/genetics , Retinal Dystrophies/metabolism
4.
Clinics (Sao Paulo) ; 79: 100372, 2024.
Article En | MEDLINE | ID: mdl-38733688

OBJECTIVE: This study aims to analyze the relationship between the Kelch-like ECH-associated protein 1 (Keap1)/Nuclear factor-erythroid 2-related factor 2 (Nrf2) and Epilepsy (EP), as well as its mechanism of action. METHODS: Thirty Wistar rats were divided into a control group (without treatment), a model group (EP modeling), and an inhibition group (EP modeling + intervention by Keap1/Nrf2 signaling pathway inhibitor ATRA) and subject to Morris water maze experiment. Then, the expression of Oxidative Stress (OS) markers, ferroptosis-associated proteins and Keap1/Nrf2 pathway in rat hippocampus was measured. In addition, rat hippocampal neuronal cell HT22 was purchased and treated accordingly based on the results of grouping, and cell proliferation and apoptosis in the three groups were determined. RESULTS: Compared with rats in the model group, those in the inhibition group showed shorter escape latency and an increased number of platform crossings (p < 0.05). Significant OS and neuron ferroptosis, increased apoptosis rate, elevated Keap1 expression, and decreased Nrf2 expression were observed in the model group compared to the control group (p < 0.05). The inhibition group exhibited notably improved OS and ferroptosis, as well as enhanced neuronal viability (p < 0.05). CONCLUSION: Inhibition of the Keap1/Nrf2 pathway can reverse the OS and neuron viability in EP rats.


Epilepsy , Ferroptosis , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Neurons , Oxidative Stress , Rats, Wistar , Signal Transduction , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/physiology , Kelch-Like ECH-Associated Protein 1/metabolism , Oxidative Stress/physiology , Signal Transduction/physiology , Ferroptosis/physiology , Ferroptosis/drug effects , Neurons/metabolism , Epilepsy/metabolism , Epilepsy/physiopathology , Male , Hippocampus/metabolism , Apoptosis/physiology , Rats , Disease Progression , Disease Models, Animal
5.
Acta Physiol (Oxf) ; 240(7): e14159, 2024 Jul.
Article En | MEDLINE | ID: mdl-38767438

AIM: Ferroptosis is a novel type of programmed cell death that performs a critical function in diabetic nephropathy (DN). Augmenter of liver regeneration (ALR) exists in the inner membrane of mitochondria, and inhibits inflammation, apoptosis, and oxidative stress in acute kidney injury; however, its role in DN remains unexplored. Here, we aimed to identify the role of ALR in ferroptosis induction and macrophage activation in DN. METHODS: The expression of ALR was examined in DN patients, db/db DN mice, and HK-2 cells treated with high glucose (HG). The effects of ALR on ferroptosis and macrophage activation were investigated with ALR conditional knockout, lentivirus transfection, transmission electron microscopy, qRT-PCR and western blotting assay. Mass spectrometry and rescue experiments were conducted to determine the mechanism of ALR. RESULTS: ALR expression was reduced in the kidney tissues of DN patients and mice, serum of DN patients, and HG-HK-2 cells. Moreover, the inhibition of ALR promoted ferroptosis, macrophage activation, and DN progression. Mechanistically, ALR can directly bind to carnitine palmitoyltransferase-1A (CPT1A), the key rate-limiting enzyme of fatty acid oxidation (FAO), and inhibit the expression of CPT1A to regulate lipid metabolism involving FAO and lipid droplet-mitochondrial coupling in DN. CONCLUSION: Taken together, our findings revealed a crucial protective role of ALR in ferroptosis induction and macrophage activation in DN and identified it as an alternative diagnostic marker and therapeutic target for DN.


Carnitine O-Palmitoyltransferase , Diabetic Nephropathies , Ferroptosis , Lipid Metabolism , Macrophage Activation , Animals , Carnitine O-Palmitoyltransferase/metabolism , Carnitine O-Palmitoyltransferase/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/genetics , Mice , Lipid Metabolism/physiology , Ferroptosis/physiology , Humans , Male , Mice, Knockout , Mice, Inbred C57BL , Oxidoreductases Acting on Sulfur Group Donors
6.
Neurotox Res ; 42(3): 27, 2024 May 31.
Article En | MEDLINE | ID: mdl-38819761

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.


Animals, Newborn , Astrocytes , Cognitive Dysfunction , Ferroptosis , Isoflurane , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Isoflurane/toxicity , Ferroptosis/drug effects , Ferroptosis/physiology , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/metabolism , Mice , Anesthetics, Inhalation/toxicity , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Reactive Oxygen Species/metabolism
7.
Exp Neurol ; 377: 114804, 2024 Jul.
Article En | MEDLINE | ID: mdl-38704083

BACKGROUND: Sevoflurane (SEV) has been found to induce neurotoxicity and cognitive impairment, leading to the development of degenerative diseases. Protein kinase C delta (PRKCD) is upregulated in the hippocampus of SEV-treated mice and may be related to SEV-related neurotoxicity. However, the underlying molecular mechanisms by which SEV mediates neurotoxicity via PRKCD remain unclear. METHODS: Normal mice and PRKCD knockout (KO) mice were exposed to SEV. Hippocampal neurons were isolated from mice hippocampal tissues. H&E staining was used for pathological morphology of hippocampal tissues, and NISSL staining was used to analyze the number of hippocampal neurons. The mRNA and protein levels were determined using quantitative real-time PCR, western blot, immunofluorescence staining and immunohistochemical staining. The mitochondrial microstructure was observed by transmission electron microscopy. Cell viability was detected by cell counting kit 8 assay, and ferroptosis was assessed by detecting related marker levels. The cognitive ability of mice was assessed by morris water maze test. And the protein levels of PRKCD, ferroptosis-related markers and Hippo pathway-related markers were examined by western bolt. RESULTS: SEV increased PRKCD expression and ferroptosis in hippocampal tissues of mice. Also, SEV promoted mouse hippocampal neuron injury by inducing ferroptosis via upregulating PRKCD expression. Knockout of PRKCD alleviated SEV-induced neurotoxicity and cognitive impairment in mice, and relieved SEV-induced ferroptosis in hippocampal neurons. PRKCD could inhibit the activity of Hippo pathway, and its knockdown also overturned SEV-mediated ferroptosis by activating Hippo pathway. CONCLUSION: SEV could induce neurotoxicity and cognitive impairment by promoting ferroptosis via inactivating Hippo pathway through increasing PRKCD expression.


Cognitive Dysfunction , Ferroptosis , Hippo Signaling Pathway , Hippocampus , Mice, Knockout , Protein Kinase C-delta , Protein Serine-Threonine Kinases , Sevoflurane , Signal Transduction , Up-Regulation , Animals , Sevoflurane/toxicity , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cognitive Dysfunction/genetics , Mice , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Up-Regulation/drug effects , Protein Kinase C-delta/metabolism , Protein Kinase C-delta/genetics , Ferroptosis/drug effects , Ferroptosis/physiology , Mice, Inbred C57BL , Male , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Anesthetics, Inhalation/toxicity , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/metabolism
8.
Neurochem Int ; 177: 105759, 2024 Jul.
Article En | MEDLINE | ID: mdl-38735393

BACKGROUND: Exosomes generated from adipose-derived mesenchymal stem cells (Exos), and in particular hypoxia-pretreated ADSCs (HExos), possess therapeutic properties that promote spinal cord repair following spinal cord injury (SCI). Nevertheless, the regulatory mechanisms through which HExos exert their effects remain unclear. METHODS: Here, next-generation sequencing (NGS) was utilized to examine abnormal circRNA expression comparing HExos to Exos. Bioinformatics analysis and RNA pulldown assays together with luciferase reporter assays were applied to determine interactions among miRNAs, mRNAs and circRNAs. ELISA and immunofluorescence staining were used to examine inflammatory cytokine levels, apoptosis and ROS deposition in LPS-treated HT-22 cells, respectively. The therapeutic effects of Exos and HExos on a mouse model of SCI were analyzed by immunohistochemistry and immunofluorescence staining. RESULTS: Our findings confirmed that HExos have more significant therapeutic influences on decreasing ROS and inflammatory cytokine levels post-SCI than Exos. NGS revealed that circ-Wdfy3 expression levels were significantly higher in HExos than Exos. Downregulation of circ-Wdfy3 led to a decrease in HExo-induced therapeutic effects on spinal cord repair post-SCI, indicating that circ-Wdfy3 has a critical role in the regulation of HExo-mediated protection against SCI. Our bioinformatics, RNA pulldown and luciferase reporter data demonstrated that GPX4 and miR-423-3p were downstream targets of circ-Wdfy3. GPX4 downregulation or miR-423-3p overexpression reversed the protective effects of circ-Wdfy3 on LPS-treated HT-22 cells. Furthermore, overexpression of circ-Wdfy3 led to an in increase in the Exo-induced therapeutic effects on spinal cord repair post-SCI through the inhibition of ferroptosis. CONCLUSIONS: circ-WDfy3-overexpressing Exos promote spinal cord repair post-SCI through mediation of ferroptosis via the miR-138-5p/GPX4 pathway.


Exosomes , Ferroptosis , RNA, Circular , Spinal Cord Injuries , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Spinal Cord Injuries/therapy , Exosomes/metabolism , Animals , Ferroptosis/physiology , RNA, Circular/genetics , RNA, Circular/metabolism , RNA, Circular/biosynthesis , Mice , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Neurons/metabolism , Male , MicroRNAs/genetics , MicroRNAs/metabolism
9.
Biomed Pharmacother ; 175: 116722, 2024 Jun.
Article En | MEDLINE | ID: mdl-38729051

Ulcerative colitis (UC) is a complex immune-mediated chronic inflammatory bowel disease. It is mainly characterized by diffuse inflammation of the colonic and rectal mucosa with barrier function impairment. Identifying new biomarkers for the development of more effective UC therapies remains a pressing task for current research. Ferroptosis is a newly identified form of regulated cell death characterized by iron-dependent lipid peroxidation. As research deepens, ferroptosis has been demonstrated to be involved in the pathological processes of numerous diseases. A growing body of evidence suggests that the pathogenesis of UC is associated with ferroptosis, and the regulation of ferroptosis provides new opportunities for UC treatment. However, the specific mechanisms by which ferroptosis participates in the development of UC remain to be more fully and thoroughly investigated. Therefore, in this review, we focus on the research advances in the mechanism of ferroptosis in recent years and describe the potential role of ferroptosis in the pathogenesis of UC. In addition, we explore the underlying role of the crosslinked pathway between ferroptosis and other mechanisms such as macrophages, neutrophils, autophagy, endoplasmic reticulum stress, and gut microbiota in UC. Finally, we also summarize the potential compounds that may act as ferroptosis inhibitors in UC in the future.


Colitis, Ulcerative , Ferroptosis , Ferroptosis/drug effects , Ferroptosis/physiology , Humans , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Animals , Gastrointestinal Microbiome , Endoplasmic Reticulum Stress/drug effects , Signal Transduction , Lipid Peroxidation/drug effects , Molecular Targeted Therapy
10.
Biomed Pharmacother ; 175: 116753, 2024 Jun.
Article En | MEDLINE | ID: mdl-38761423

Ferroptosis is a form of cell death mediated by iron and lipid peroxidation (LPO). Recent studies have provided compelling evidence to support the involvement of ferroptosis in the pathogenesis of various neurodegenerative diseases (NDDs), such as Alzheimer's disease (AD), Parkinson's disease (PD). Therefore, understanding the mechanisms that regulate ferroptosis in NDDs may improve disease management. Ferroptosis is regulated by multiple mechanisms, and different degradation pathways, including autophagy and the ubiquitinproteasome system (UPS), orchestrate the complex ferroptosis response by directly or indirectly regulating iron accumulation or lipid peroxidation. Ubiquitination plays a crucial role as a protein posttranslational modification in driving ferroptosis. Notably, E3 ubiquitin ligases (E3s) and deubiquitinating enzymes (DUBs) are key enzymes in the ubiquitin system, and their dysregulation is closely linked to the progression of NDDs. A growing body of evidence highlights the role of ubiquitin system enzymes in regulating ferroptosis sensitivity. However, reports on the interaction between ferroptosis and ubiquitin signaling in NDDs are scarce. In this review, we first provide a brief overview of the biological processes and roles of the UPS, summarize the core molecular mechanisms and potential biological functions of ferroptosis, and explore the pathophysiological relevance and therapeutic implications of ferroptosis in NDDs. In addition, reviewing the roles of E3s and DUBs in regulating ferroptosis in NDDs aims to provide new insights and strategies for the treatment of NDDs. These include E3- and DUB-targeted drugs and ferroptosis inhibitors, which can be used to prevent and ameliorate the progression of NDDs.


Ferroptosis , Neurodegenerative Diseases , Ubiquitin-Protein Ligases , Ferroptosis/drug effects , Ferroptosis/physiology , Humans , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/enzymology , Animals , Ubiquitin-Protein Ligases/metabolism , Deubiquitinating Enzymes/metabolism , Ubiquitination , Signal Transduction/drug effects , Molecular Targeted Therapy
11.
Neurochem Res ; 49(7): 1643-1654, 2024 Jul.
Article En | MEDLINE | ID: mdl-38782838

Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra. Ferroptosis, an iron-dependent form of regulated cell death, may contribute to the progression of PD owing to an unbalanced brain redox status. Physical exercise is a complementary therapy that can modulate ferroptosis in PD by regulating the redox system through the activation of nuclear factor (erythroid-derived 2)-like 2 (NRF2) and brain-derived neurotrophic factor (BDNF) signaling. However, the precise effects of physical exercise on ferroptosis in PD remain unclear. In this review, we explored how physical exercise influences NRF2 and BDNF signaling and affects ferroptosis in PD. We further investigated relevant publications over the past two decades by searching the PubMed, Web of Science, and Google Scholar databases using keywords related to physical exercise, PD, ferroptosis, and neurotrophic factor antioxidant signaling. This review provides insights into current research gaps and demonstrates the necessity for future research to elucidate the specific mechanisms by which exercise regulates ferroptosis in PD, including the assessment of different exercise protocols and their long-term effects. Ultimately, exploring these aspects may lead to the development of improved exercise interventions for the better management of patients with PD.


Brain-Derived Neurotrophic Factor , Exercise , Ferroptosis , NF-E2-Related Factor 2 , Parkinson Disease , NF-E2-Related Factor 2/metabolism , Humans , Brain-Derived Neurotrophic Factor/metabolism , Ferroptosis/physiology , Parkinson Disease/metabolism , Parkinson Disease/therapy , Animals , Exercise/physiology , Signal Transduction/physiology
12.
Exp Gerontol ; 192: 112443, 2024 Jul.
Article En | MEDLINE | ID: mdl-38697556

OBJECTIVE: Ferroptosis has been recognized as being closely associated with cognitive impairment. Research has established that Alzheimer's disease (AD)-associated proteins, such as amyloid precursor protein (APP) and phosphorylated tau, are involved in brain iron metabolism. These proteins are found in high concentrations within senile plaques and neurofibrillary tangles. Repetitive transcranial magnetic stimulation (rTMS) offers a non-pharmacological approach to AD treatment. This study aims to explore the potential therapeutic effects of rTMS on cognitive impairment through the modulation of the ferroptosis pathway, thereby laying both a theoretical and experimental groundwork for the application of rTMS in treating Alzheimer's disease. METHODS: The study utilized senescence-accelerated mouse prone 8 (SAMP8) mice to model brain aging-related cognitive impairment, with senescence-accelerated-mouse resistant 1 (SAMR1) mice acting as controls. The SAMP8 mice were subjected to high-frequency rTMS at 25 Hz for durations of 14 and 28 days. Cognitive function was evaluated using behavioral tests. Resting-state functional magnetic resonance imaging (rs-fMRI) assessed alterations in cerebral activity by measuring the fractional amplitude of low-frequency fluctuations (fALFF) of the blood oxygen level-dependent signal. Neuronal recovery post-rTMS in the SAMP8 model was examined via HE and Nissl staining. Immunohistochemistry was employed to detect the expression of APP and Phospho-Tau (Thr231). Oxidative stress markers were quantified using biochemical assay kits. ELISA methods were utilized to measure hippocampal levels of Fe2+ and Aß1-42. Finally, the expression of proteins related to the ferroptosis pathway was determined through western blot analysis. RESULTS: The findings indicate that 25 Hz rTMS enhances cognitive function and augments cerebral activity in SAMP8 model mice. Treatment with rTMS in these mice resulted in diminished oxidative stress and safeguarded neurons against damage. Additionally, iron accumulation was mitigated, and the expression of ferroptosis pathway proteins Gpx4, system Xc-, and Nrf2 was elevated. CONCLUSIONS: The Tau/APP-Fe-GPX4/system Xc-/Nrf2 pathway is implicated in the remedial effects of rTMS on cognitive dysfunction, offering a theoretical and experimental basis for employing rTMS in AD treatment.


Aging , Cognitive Dysfunction , Disease Models, Animal , Ferroptosis , Transcranial Magnetic Stimulation , Animals , Transcranial Magnetic Stimulation/methods , Ferroptosis/physiology , Cognitive Dysfunction/therapy , Mice , Aging/physiology , Male , Magnetic Resonance Imaging , tau Proteins/metabolism , Alzheimer Disease/therapy , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism
13.
J Cancer Res Clin Oncol ; 150(5): 228, 2024 May 03.
Article En | MEDLINE | ID: mdl-38700533

BACKGROUND: Lung cancer is a serious threat to human health and is the first leading cause of cancer death. Ferroptosis, a newly discovered form of programmed cell death associated with redox homeostasis, is of particular interest in the lung cancer, given the high oxygen environment of lung cancer. NADPH has reducing properties and therefore holds the potential to resist ferroptosis. Resistance to ferroptosis exists in lung cancer, but the role of NADK in regulating ferroptosis in lung cancer has not been reported yet. METHODS: Immunohistochemistry (IHC) was used to analyse the expression of NADK in 86 cases of lung adenocarcinoma(LUAD) and adjacent tissues, and a IHC score was assigned to each sample. Chi-square and kaplan-meier curve was performed to analyse the differences in metastasis and five-year survival between the two groups with NADK high or low scores. Proliferation of NADK-knockdown LUAD cell lines was detected in vivo and vitro. Furthermore, leves of ROS, MDA and Fe2+ were measured to validate the effect and mechanism of NADK on ferroptosis in LUAD. RESULTS: The expression of NADK was significantly evaluated in LUAD tissues as compared to adjacent non-cancerous tissues. The proliferation of NADK-knockdown cells was inhibited both in vivo and vitro, and increasing levels of intracellular ROS, Fe2+ and lipid peroxide products (MDA) were observed. Furthermore, NADK-knockdown promoted the ferroptosis of LUAD cells induced by Erastin/RSL3 by regulating the level of NADPH and the expression of FSP1. Knockdown of NADK enhanced the sensitivities of LUAD cells to Erastin/RSL3-induced ferroptosis by regulating NADPH level and FSP1 expression. CONCLUSIONS: NADK is over-expressed in LUAD patients. Knockdown of NADK inhibited the proliferation of LUAD cells both in vitro and in vivo and promotes the Erastin/RSL3-induced ferroptosis of LUAD cells by down-regulating the NADPH/FSP1 axis.


Adenocarcinoma of Lung , Ferroptosis , Lung Neoplasms , NADP , Animals , Female , Humans , Male , Mice , Middle Aged , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Cell Line, Tumor , Cell Proliferation , Ferroptosis/genetics , Ferroptosis/physiology , Gene Knockdown Techniques , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice, Nude , NADP/metabolism
14.
J Neuroinflammation ; 21(1): 116, 2024 May 03.
Article En | MEDLINE | ID: mdl-38702778

BACKGROUND: Subarachnoid hemorrhage (SAH), a severe subtype of stroke, is characterized by notably high mortality and morbidity, largely due to the lack of effective therapeutic options. Although the neuroprotective potential of PPARg and Nrf2 has been recognized, investigative efforts into oroxin A (OA), remain limited in preclinical studies. METHODS: SAH was modeled in vivo through filament perforation in male C57BL/6 mice and in vitro by exposing HT22 cells to hemin to induce neuronal damage. Following the administration of OA, a series of methods were employed to assess neurological behaviors, brain water content, neuronal damage, cell ferroptosis, and the extent of neuroinflammation. RESULTS: The findings indicated that OA treatment markedly improved survival rates, enhanced neurological functions, mitigated neuronal death and brain edema, and attenuated the inflammatory response. These effects of OA were linked to the suppression of microglial activation. Moreover, OA administration was found to diminish ferroptosis in neuronal cells, a critical factor in early brain injury (EBI) following SAH. Further mechanistic investigations uncovered that OA facilitated the translocation of nuclear factor erythroid 2-related factor 2 (Nrf-2) from the cytoplasm to the nucleus, thereby activating the Nrf2/GPX4 pathway. Importantly, OA also upregulated the expression of FSP1, suggesting a significant and parallel protective effect against ferroptosis in EBI following SAH in synergy with GPX4. CONCLUSION: In summary, this research indicated that the PPARg activator OA augmented the neurological results in rodent models and diminished neuronal death. This neuroprotection was achieved primarily by suppressing neuronal ferroptosis. The underlying mechanism was associated with the alleviation of cellular death through the Nrf2/GPX4 and FSP1/CoQ10 pathways.


Ferroptosis , Mice, Inbred C57BL , Neuroinflammatory Diseases , Subarachnoid Hemorrhage , Animals , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/pathology , Subarachnoid Hemorrhage/complications , Ferroptosis/drug effects , Ferroptosis/physiology , Mice , Male , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurons/metabolism , Neurons/drug effects , Neurons/pathology
15.
J Cardiothorac Surg ; 19(1): 265, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38664788

BACKGROUND: Hypoxia/reoxygenation (H/R) induces cardiomyocyte ferroptosis, a core remodeling event in myocardial ischemia/reperfusion injury. Methyltransferase-like 14 (METTL14) emerges as a writer of N6-methyladenosine (m6A) modification. This study was conducted to decipher the role of METTL14 in H/R-induced cardiomyocyte ferroptosis. METHODS: Mouse cardiomyocytes HL-1 were cultured and underwent H/R treatment. The degree of ferroptosis after H/R treatment was appraised by the cell counting kit-8 assay, assay kits (ROS/GSH/Fe2+), and Western blotting (GPX4/ACSL4). The intracellular expressions of METTL14, pri-miR-146a-5p, miR-146a-5p, or adaptor protein phosphotyrosine interacting with PH domain and leucine zipper 1 (APPL1) were examined by real-time quantitative polymerase chain reaction or Western blotting, with m6A quantification analysis and RNA immunoprecipitation to determine the total m6A level and the expression of pri-miR-146a-5p bound to DiGeorge critical region 8 (DGCR8) and m6A-modified pri-miR-146a-5p. The binding of miR-146a-5p to APPL1 was testified by the dual-luciferase assay. RESULTS: H/R treatment induced cardiomyocyte ferroptosis (increased ROS, Fe2+, and ACSL4 and decreased GSH and GPX4) and upregulated METTL14 expression. METTL14 knockdown attenuated H/R-induced cardiomyocyte ferroptosis. METTL14 induced the recognition of pri-miR-146a-5p by DGCR8 by increasing m6A modification on pri-miR-146a-5p, which promoted the conversion of pri-miR-146a-5p into miR-146a-5p and further repressed APPL1 transcription. miR-146a-5p upregulation or APPL1 downregulation limited the inhibitory effect of METTL14 downregulation on H/R-induced cardiomyocyte ferroptosis. CONCLUSION: METTL14 promoted miR-146a-5p expression through the recognition and processing of pri-miR-146a-5p by DGCR8, which repressed APPL1 transcription and triggered H/R-induced cardiomyocyte ferroptosis.


Adenosine , Adenosine/analogs & derivatives , Ferroptosis , Methyltransferases , Myocardial Reperfusion Injury , Myocytes, Cardiac , Ferroptosis/physiology , Ferroptosis/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Animals , Adenosine/metabolism , Mice , Methyltransferases/metabolism , Methyltransferases/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/genetics , MicroRNAs/genetics , MicroRNAs/metabolism
16.
Brain Res Bull ; 211: 110948, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38614406

BACKGROUND: The treatment for cerebral ischemia remains limited, and new therapeutic strategies are urgently needed. Exosome has shown great promise for the treatment of cerebral ischemia. Steroid receptor coactivator-3 (SRC-3) was reported to be involved in neurological performances. In this study, we aimed to investigate the protective effects of mesenchymal stem cell (MSC)-derived exosomes overexpressing SRC-3 on cerebral ischemia in mice. METHODS: The mice were treated with an intracerebroventricular injection of GFP-overexpressed exosomes (GFP-exo) and SRC-3-overexpressed exosomes (SRC3-exo) in a middle cerebral artery occlusion (MCAO) model of cerebral ischemia. RESULTS: The results showed that SRC3-exo treatment significantly inhibited lipid peroxidation and ferroptosis of the neurons subjected to oxygen-glucose deprivation. It further suppressed the activation of microglia and astrocytes, and decreased the production of pro-inflammatory cytokines in the brains of MCAO mice. Furthermore, SRC3-exo treatment reduced the water content of brain tissue and infarct size, which alleviated the neurological damage and improved neurological performances in the MCAO mice. CONCLUSIONS: Our results suggest that MSC-derived exosomes expressing SRC3 can be a therapeutic strategy for cerebral ischemia by inhibiting ferroptosis.


Brain Ischemia , Exosomes , Ferroptosis , Infarction, Middle Cerebral Artery , Mesenchymal Stem Cells , Nuclear Receptor Coactivator 3 , Animals , Exosomes/metabolism , Exosomes/transplantation , Mice , Ferroptosis/physiology , Mesenchymal Stem Cells/metabolism , Male , Brain Ischemia/metabolism , Brain Ischemia/therapy , Nuclear Receptor Coactivator 3/metabolism , Nuclear Receptor Coactivator 3/genetics , Infarction, Middle Cerebral Artery/metabolism , Mice, Inbred C57BL , Neurons/metabolism , Disease Models, Animal , Astrocytes/metabolism , Brain/metabolism
18.
Toxicol Appl Pharmacol ; 486: 116936, 2024 May.
Article En | MEDLINE | ID: mdl-38641223

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that is pivotal in development, metabolic homeostasis, and immune responses. While recent research has highlighted AhR's significant role in modulating oxidative stress responses, its mechanistic relationship with ferroptosis-an iron-dependent, non-apoptotic cell death-remains to be fully elucidated. In our study, we discovered that AhR plays a crucial role in ferroptosis, in part by transcriptionally regulating the expression of the solute carrier family 7 member 11 (SLC7A11). Our findings indicate that both pharmacological inactivation and genetic ablation of AhR markedly enhance erastin-induced ferroptosis. This enhancement is achieved by suppressing SLC7A11, leading to increased lipid peroxidation. We also obtained evidence of post-translational modifications of SLC7A11 during ferroptosis. Additionally, we observed that indole 3-pyruvate (I3P), an endogenous ligand of AhR, protects cells from ferroptosis through an AhR-dependent mechanism. Based on these insights, we propose that AhR transcriptionally regulates the expression of SLC family genes, which in turn play a pivotal role in mediating ferroptosis. This underscores AhR's essential role in suppressing lipid oxidation and ensuring cell survival under oxidative stress.


Amino Acid Transport System y+ , Ferroptosis , Receptors, Aryl Hydrocarbon , Signal Transduction , Ferroptosis/drug effects , Ferroptosis/physiology , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Humans , Animals , Mice , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Lipid Peroxidation/drug effects , Mice, Inbred C57BL , Mice, Knockout , Gene Expression Regulation , Piperazines/pharmacology
19.
Sci Total Environ ; 934: 172618, 2024 Jul 15.
Article En | MEDLINE | ID: mdl-38663589

Neurotoxicity, stemming from exposure to various chemical, biological, and physical agents, poses a substantial threat to the intricate network of the human nervous system. This article explores the implications of ferroptosis, a regulated form of programmed cell death characterized by iron-dependent lipid peroxidation, in environmental-induced neurotoxicity. While apoptosis has historically been recognized as a primary mechanism in neurotoxic events, recent evidence suggests the involvement of additional pathways, including ferroptosis. The study aims to conduct a comprehensive review of the existing literature on ferroptosis induced by environmental neurotoxicity across diverse agents such as natural toxins, insecticides, particulate matter, acrylamide, nanoparticles, plastic materials, metal overload, viral infections, anesthetics, chemotherapy, and radiation. The primary objective is to elucidate the diverse mechanisms through which these agents trigger ferroptosis, leading to neuronal cell death. Furthermore, the article explores potential preventive or therapeutic strategies that could mitigate ferroptosis, offering insights into protective measures against neurological damage induced by environmental stressors. This comprehensive review contributes to our evolving understanding of neurotoxicological processes, highlighting ferroptosis as a significant contributor to neuronal cell demise induced by environmental exposures. The insights gained from this study may pave the way for the development of targeted interventions to protect against ferroptosis-mediated neurotoxicity and ultimately safeguard public health.


Ferroptosis , Neurotoxicity Syndromes , Ferroptosis/drug effects , Ferroptosis/physiology , Humans , Neurotoxicity Syndromes/etiology , Environmental Exposure/adverse effects , Environmental Pollutants/toxicity , Neurons/drug effects , Lipid Peroxidation/drug effects
20.
Exp Neurol ; 377: 114797, 2024 Jul.
Article En | MEDLINE | ID: mdl-38670252

Diabetic is a major contributor to the unfavorable prognosis of ischemic stroke. However, intensive hypoglycemic strategies do not improve stroke outcomes, implying that diabetes may affect stroke outcomes through other ways. Ferroptosis is a novel programmed cell death pathway associated with the development of diabetes and ischemic stroke. This study aimed to investigate the effect of streptozotocin (STZ)-induced diabetes on ferroptosis after stroke from the immune cell perspective, and to provide a theoretical foundation for the clinical management of ischemic stroke in patients with diabetes. The results revealed that STZ-induced diabetes not only facilitates the infiltration of neutrophils into the brain after stroke, but also upregulates the expression of lipocalin 2 (LCN2) in neutrophils. LCN2 promotes lipid peroxide accumulation by increasing intracellular ferrous ions, which intensify ferroptosis in major brain cell populations, especially neurons. Our findings suggest that STZ-induced diabetes aggravates ischemic stroke partially by mediating ferroptosis through neutrophil-derived LCN2. These data contribute to improved understanding of post-stroke immune regulation in diabetes, and offer a potentially novel therapeutic target for the management of acute-stage ischemic stroke complicated with diabetes.


Diabetes Mellitus, Experimental , Ferroptosis , Ischemic Stroke , Lipocalin-2 , Mice, Inbred C57BL , Neurons , Neutrophils , Up-Regulation , Lipocalin-2/metabolism , Animals , Ferroptosis/physiology , Ferroptosis/drug effects , Neutrophils/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Male , Neurons/metabolism , Neurons/pathology , Neurons/drug effects , Mice
...