Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 14(1): 5078, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38429394

ABSTRACT

Ferroptosis is a recently identified form of programmed cell death that plays an important role in the pathophysiological process of osteoarthritis (OA). Herein, we investigated the protective effect of moderate mechanical stress on chondrocyte ferroptosis and further revealed the internal molecular mechanism. Intra-articular injection of sodium iodoacetate (MIA) was conducted to induce the rat model of OA in vivo, meanwhile, interleukin-1 beta (IL-1ß) was treated to chondrocytes to induce the OA cell model in vitro. The OA phenotype was analyzed by histology and microcomputed tomography, the ferroptosis was analyzed by transmission electron microscope and immunofluorescence. The expression of ferroptosis and cartilage metabolism-related factors was analyzed by immunohistochemical and Western blot. Animal experiments revealed that moderate-intensity treadmill exercise could effectively reduce chondrocyte ferroptosis and cartilage matrix degradation in MIA-induced OA rats. Cell experiments showed that 4-h cyclic tensile strain intervention could activate Nrf2 and inhibit the NF-κB signaling pathway, increase the expression of Col2a1, GPX4, and SLC7A11, decrease the expression of MMP13 and P53, thereby restraining IL-1ß-induced chondrocyte ferroptosis and degeneration. Inhibition of NF-κB signaling pathway relieved the chondrocyte ferroptosis and degeneration. Meanwhile, overexpression of NF-κB by recombinant lentivirus reversed the positive effect of CTS on chondrocytes. Moderate mechanical stress could activate the Nrf2 antioxidant system, inhibit the NF-κB p65 signaling pathway, and inhibit chondrocyte ferroptosis and cartilage matrix degradation by regulating P53, SLC7A11, and GPX4.


Subject(s)
Ferroptosis , Osteoarthritis , Stress, Mechanical , Animals , Rats , Chondrocytes/metabolism , Interleukin-1beta/metabolism , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , NF-kappa B/physiology , Osteoarthritis/metabolism , Osteoarthritis/pathology , Signal Transduction , Tumor Suppressor Protein p53/metabolism , X-Ray Microtomography , Transcription Factor RelA/metabolism , Transcription Factor RelA/physiology , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/physiology
2.
Biochemistry ; 60(37): 2761-2772, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34492183

ABSTRACT

Glutathione peroxidase 4 (GPx4) serves as the only enzyme that protects membranes through the reduction of lipid hydroperoxides, preventing membrane oxidative damage and cell death through ferroptosis. Recently, GPx4 has gained attention as a therapeutic target for cancer through inhibition and as a target for inflammatory diseases through activation. In addition, GPx4 isoforms perform several distinct moonlighting functions including cysteine cross-linking of protamines during sperm cell chromatin remodeling, a function for which molecular and structural details are undefined. Despite the importance in biology, disease, and potential for drug development, little is known about GPx4 functional interactions at high resolution. This study presents the first NMR assignments of GPx4, and the electrostatic interaction of GPx4 with the membrane is characterized. Mutagenesis reveals the cationic patch residues that are key to membrane binding and stabilization. The cationic patch is observed to be important in binding headgroups of highly anionic cardiolipin. A novel lipid binding site is observed adjacent to the catalytic site and may enable protection of lipid-headgroups from oxidative damage. Arachidonic acid is also found to engage with GPx4, while cholesterol did not display any interaction. The cationic patch residues were also found to enable DNA binding, the first observation of this interaction. Electrostatic DNA binding explains a mechanism for the nuclear isoform of GPx4 to target DNA-bound protamines and to potentially reduce oxidatively damaged DNA. Together, these results highlight the importance of electrostatics in the function of GPx4 and illuminate how the multifunctional enzyme is able to fill multiple biological roles.


Subject(s)
Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/ultrastructure , Binding Sites , Catalytic Domain , Cell Death , Cell Membrane/metabolism , Cell Membrane/physiology , DNA/metabolism , DNA/physiology , Ferroptosis , Glutathione Peroxidase/metabolism , Humans , Lipid Peroxides/metabolism , Magnetic Resonance Imaging/methods , Membrane Lipids/metabolism , Membrane Lipids/physiology , Oxidative Stress , Phospholipid Hydroperoxide Glutathione Peroxidase/physiology , Protein Binding , Protein Isoforms/metabolism , Static Electricity
3.
Sci Rep ; 10(1): 17934, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087733

ABSTRACT

PRDX4, a member of peroxiredoxin family, is largely concentrated in the endoplasmic reticulum (ER) and plays a pivotal role in the redox relay during oxidative protein folding as well as in peroxidase reactions. A testis-specific PRDX4 variant transcript (PRDX4t) lacks the conventional exon 1, which encodes the signal peptide that is required for entry into the ER lumen, but instead carries alternative exon 1, which is transcribed from the upstream promoter in a testis-specific manner and results in the PRDX4t protein being localized in the cytosol. However, the potential roles of PRDX4t in male genital action remain unknown. Using a CRISPR/Cas9 system, we first disrupted the testis-specific promoter/exon 1 and generated mice that were specifically deficient in PRDX4t. The resulting PRDX4t knockout (KO) mice underwent normal spermatogenesis and showed no overt abnormalities in the testis. Mating PRDX4t KO male mice with wild-type (WT) female mice produced normal numbers of offspring, indicating that a PRDX4t deficiency alone had no effect on fertility in the male mice. We then generated mice lacking both PRDX4 and PRDX4t by disrupting exon 2, which is communal to these variants. The resulting double knockout (DKO) mice were again fertile, and mature sperm isolated from the epididymis of DKO mice exhibited a normal fertilizing ability in vitro. In the meantime, the protein levels of glutathione peroxidase 4 (GPX4), which plays an essential role in the disulfide bond formation during spermatogenesis, were significantly increased in the testis and caput epididymis of the DKO mice compared with the WT mice. Based on these results, we conclude that the disruption of the function of PRDX4t in the spermatogenic process appears to be compensated by other factors including GPX4.


Subject(s)
Fertility/genetics , Genetic Variation/genetics , Peroxiredoxins/genetics , Peroxiredoxins/physiology , Spermatogenesis/genetics , Animals , Exons , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity/genetics , Peroxiredoxins/deficiency , Peroxiredoxins/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/physiology , Pregnancy , Testis/metabolism
4.
Life Sci ; 260: 118077, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32810509

ABSTRACT

AIMS: Multiple myeloma (MM) is the second hematological plasma cell malignany and sensitive to fingolimod (FTY720), a novel immunosuppressant. Previous study shows FTY720-induced apoptosis and autophagy can cause cell death in MM cells, however, the high death rate cannot fully be explained. The study aims to investigate further mechanism of how FTY720 kills MM cells. MATERIALS AND METHODS: Experiments are performed on 25 human primary cell samples and two MM cell lines by flow cytometry, fluorescence microscopy, and transmission electron microscopy. Expressions of relative factors are tested by qRT-PCR or western blot. KEY FINDINGS: Ferroptosis-specific inhibitors, deferoxamine mesylate (DFOM) and ferropstatin-1 (Fer-1), reverse FTY720-induced cell death in MM cells. Glutathione peroxidase 4 (GPX4) and soluble carrier family 7 member 11 (SLC7A11), key regulators of ferroptosis, are highly expressed in primary MM cells and can be decreased by FTY720 at the mRNA and protein level in MM cells. In addition, FTY720 induces other characteristic changes of ferroptosis. Furthermore, FTY720 can dephosphorylate AMP-activated protein kinase subunit ɑ (AMPKɑ) at the Thr172 site by activating protein phosphatase 2A (PP2A) and reduce the expression of phosphorylated eukaryotic elongation factor 2 (eEF2), finally cause MM cell death. Using LB-100, a PP2A inhibitor, AICAR, an agonist of AMPK, and bafilomycin A1 (Baf-A1), an autophagy inhibitor, we discover that FTY720 induces ferroptosis and autophagy through the PP2A/AMPK pathway, and ferroptosis and autophagy can reinforce each other. SIGNIFICANCE: These results provide a new perspective on the treatment of MM.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/drug effects , Ferroptosis/drug effects , Fingolimod Hydrochloride/pharmacology , Multiple Myeloma/pathology , Protein Phosphatase 2/metabolism , Amino Acid Transport System y+/drug effects , Amino Acid Transport System y+/physiology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cyclohexylamines/pharmacology , Deferoxamine/pharmacology , Humans , Multiple Myeloma/drug therapy , Phenylenediamines/pharmacology , Phospholipid Hydroperoxide Glutathione Peroxidase/drug effects , Phospholipid Hydroperoxide Glutathione Peroxidase/physiology , Signal Transduction/drug effects
5.
Int J Mol Sci ; 21(14)2020 Jul 11.
Article in English | MEDLINE | ID: mdl-32664576

ABSTRACT

Ferroptosis is an iron-dependent form of cell death characterized by intracellular lipid peroxide accumulation and redox imbalance. Ferroptosis shows specific biological and morphological features when compared to the other cell death patterns. The loss of lipid peroxide repair activity by glutathione peroxidase 4 (GPX4), the presence of redox-active iron and the oxidation of polyunsaturated fatty acid (PUFA)-containing phospholipids are considered as distinct fingerprints of ferroptosis. Several pathways, including amino acid and iron metabolism, ferritinophagy, cell adhesion, p53, Keap1/Nrf2 and phospholipid biosynthesis, can modify susceptibility to ferroptosis. Through the decades, various diseases, including acute kidney injury; cancer; ischemia-reperfusion injury; and cardiovascular, neurodegenerative and hepatic disorders, have been associated with ferroptosis. In this review, we provide a comprehensive analysis of the main biological and biochemical mechanisms of ferroptosis and an overview of chemicals used as inducers and inhibitors. Then, we report the contribution of ferroptosis to the spectrum of liver diseases, acute or chronic. Finally, we discuss the use of ferroptosis as a therapeutic approach against hepatocellular carcinoma, the most common form of primary liver cancer.


Subject(s)
Ferroptosis , Liver Diseases/pathology , Animals , Autophagy/physiology , Chemical and Drug Induced Liver Injury/pathology , Cyclohexylamines/pharmacology , Cysteine/metabolism , Ferroptosis/drug effects , Glutathione/biosynthesis , Heme/metabolism , Humans , Iron/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Lipid Peroxidation/drug effects , Lipoxygenase/physiology , Liver Neoplasms/pathology , Oxidative Stress , Phenylenediamines/pharmacology , Phospholipid Hydroperoxide Glutathione Peroxidase/physiology , Piperazines/pharmacology , Quinoxalines/pharmacology , Reactive Oxygen Species/metabolism , Reperfusion Injury/pathology , Signal Transduction , Sorafenib/pharmacology , Spiro Compounds/pharmacology , Sulfasalazine/pharmacology , Tumor Suppressor Protein p53/metabolism , alpha-Tocopherol/pharmacology
6.
Blood ; 136(11): 1303-1316, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32458004

ABSTRACT

Metabolic alterations in cancer represent convergent effects of oncogenic mutations. We hypothesized that a metabolism-restricted genetic screen, comparing normal primary mouse hematopoietic cells and their malignant counterparts in an ex vivo system mimicking the bone marrow microenvironment, would define distinctive vulnerabilities in acute myeloid leukemia (AML). Leukemic cells, but not their normal myeloid counterparts, depended on the aldehyde dehydrogenase 3a2 (Aldh3a2) enzyme that oxidizes long-chain aliphatic aldehydes to prevent cellular oxidative damage. Aldehydes are by-products of increased oxidative phosphorylation and nucleotide synthesis in cancer and are generated from lipid peroxides underlying the non-caspase-dependent form of cell death, ferroptosis. Leukemic cell dependence on Aldh3a2 was seen across multiple mouse and human myeloid leukemias. Aldh3a2 inhibition was synthetically lethal with glutathione peroxidase-4 (GPX4) inhibition; GPX4 inhibition is a known trigger of ferroptosis that by itself minimally affects AML cells. Inhibiting Aldh3a2 provides a therapeutic opportunity and a unique synthetic lethality to exploit the distinctive metabolic state of malignant cells.


Subject(s)
Aldehyde Oxidoreductases/physiology , Carbolines/pharmacology , Cyclohexylamines/pharmacology , Ferroptosis/drug effects , Hematopoiesis/physiology , Leukemia, Myeloid, Acute/enzymology , Neoplasm Proteins/physiology , Phenylenediamines/pharmacology , Aldehyde Oxidoreductases/genetics , Aldehydes/pharmacology , Animals , Cell Line, Tumor , Cytarabine/administration & dosage , Doxorubicin/administration & dosage , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Lipid Peroxidation , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/physiology , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Oleic Acid/pharmacology , Oncogene Proteins, Fusion/physiology , Oxidation-Reduction , Oxidative Stress , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase/physiology
7.
Neuromolecular Med ; 21(2): 110-119, 2019 06.
Article in English | MEDLINE | ID: mdl-30600476

ABSTRACT

Huntington's disease (HD) is an autosomal dominant and fatal neurodegenerative disorder, which is caused by an abnormal CAG repeat in the huntingtin gene. Despite its well-defined genetic origin, the molecular mechanisms of neuronal death are unclear yet, thus there are no effective strategies to block or postpone the process of HD. Ferroptosis, a recently identified iron-dependent cell death, attracts considerable attention due to its putative involvement in neurodegenerative diseases. Accumulative data suggest that ferroptosis is very likely to participate in HD, and inhibition of the molecules and signaling pathways involved in ferroptosis can significantly eliminate the symptoms and pathology of HD. This review first describes evidence for the close relevance of ferroptosis and HD in patients and mouse models, then summarizes advances for the mechanisms of ferroptosis involved in HD, finally outlines some therapeutic strategies targeted ferroptosis. Comprehensive understanding of the emerging roles of ferroptosis in the occurrence of HD will help us to explore effective therapies for slowing the progression of this disease.


Subject(s)
Ferroptosis , Huntington Disease/physiopathology , Animals , Antioxidants/pharmacology , Disease Models, Animal , Ferroptosis/drug effects , Ferroptosis/physiology , Humans , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Huntington Disease/therapy , Iron/metabolism , Iron Chelating Agents/pharmacology , Lipid Peroxidation , Mice , Mice, Transgenic , NF-E2-Related Factor 2/physiology , Neuroglia/metabolism , Neurons/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/deficiency , Phospholipid Hydroperoxide Glutathione Peroxidase/physiology
8.
Oncol Rep ; 41(2): 1264-1274, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30535490

ABSTRACT

Reactive oxygen species (ROS) have been widely accepted as critical molecules playing regulatory roles in various biological processes, including proliferation, differentiation and apoptotic/ferroptotic/necrotic cell death. Emerging evidence suggests that ROS may be involved in the induction of epithelial­to­mesenchymal transition (EMT), which has been reported to promote cancer stem­like cell (CSC) generation. Recent data indicate that altered accumulation of ROS is associated with CSC generation, EMT and hypoxia exposure, but the underlying mechanisms are poorly understood. In the present study, we derived CSCs from Panc­1 human pancreatic cancer cells and characterized them using serial replating assays and western blot analysis. Functional identification of viable cells was performed using the CCK­8 assay and colony formation assays. The expression of various antioxidant enzymes, including superoxide dismutase (SOD) and glutathione peroxidase (GPX), was measured by western blot analysis in Panc­1 CSCs. The role of GPX4 in regulating biological processes of Panc­1 CSCs was assessed by proliferation, sphere formation and invasion assays with or without oxidative stress. Manipulation of GPX4 expression by siRNA knockdown or an overexpression vector was performed to assess functions including proliferation, colony formation and invasion. EMT hallmark genes were detected after GPX4 alteration by RT­qPCR and western blot analysis. Panc­1 CSCs displayed more resistance to hypoxia exposure. Compared with the parental Panc­1 cells, Panc­1 CSCs expressed an obviously higher endogenous GPX4 level, indicating their role in maintaining homeostasis. During GPX4 knockdown, ROS accumulation was promoted following oxidative stress exposure to either H2O2 or erastin. Additionally, overexpression of GPX4 eliminated ROS induction by oxidative stress exposure and thus, exerted protective effects on physiological processes in the Panc­1 CSCs. Knockdown of GPX4 arrested cell cycle progression at the G1/G0 phase; inhibited cell proliferation, colony formation, invasion and the stemness phenotype in the Panc­1 CSCs; and decreased the EMT phenotype. Collectively, GPX4 plays a critical role in maintaining oxidative homeostasis and regulates several biological processes, including stemness and EMT, in Panc­1 CSCs.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/pathology , Phospholipid Hydroperoxide Glutathione Peroxidase/physiology , Reactive Oxygen Species/metabolism , Gene Knockdown Techniques , Homeostasis , Humans , Oxidative Stress , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...