Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 628
1.
PeerJ ; 12: e17551, 2024.
Article En | MEDLINE | ID: mdl-38887622

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.


Cyclohexylamines , Ferroptosis , Fibroblasts , Fibrosis , Keloid , NF-E2-Related Factor 2 , Phenylenediamines , Phospholipid Hydroperoxide Glutathione Peroxidase , Humans , Ferroptosis/drug effects , Keloid/pathology , Keloid/metabolism , Keloid/drug therapy , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Cyclohexylamines/pharmacology , Fibrosis/metabolism , Fibrosis/pathology , Phenylenediamines/pharmacology , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Male , Lipid Peroxidation/drug effects , Female , Adult , Iron/metabolism , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Receptors, Transferrin/metabolism , Receptors, Transferrin/genetics , Piperazines/pharmacology , Actins/metabolism , Actins/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Membrane Potential, Mitochondrial/drug effects
2.
Ann Clin Lab Sci ; 54(2): 190-200, 2024 Mar.
Article En | MEDLINE | ID: mdl-38802151

OBJECTIVE: Polydopamine nanoparticles (PDA NPs) are a promising topic in the fields of drug delivery, tissue engineering, bioimaging, etc. The present study aims to explore the impact of PDA NPs carrying ferroptosis inhibitor ferstatin-1 (Fer-1) on myocardial ischemia-reperfusion injury (MIRI). METHODS: After establishment of a rat model of MIRI and PDA NPs, the rats were divided into 4 groups: model group, sham operation group, Fer-1 group, and nano+Fer-1 group (n=8). To detect the effect of PDA NPs encapsulating Fer-1 on ferroptosis in MIRI rats, we further set up NOX4 overexpression group (pc-NOX4 group), NOX4 inhibitor group (Fulvene-5 group), nano+Fer-1+pc-NOX4 group, and nano+Fer-1+Fulvene-5 group (n=8). A CCK-8 assay was conducted to assess cell viability and staining to detect cardiomyocyte apoptosis and observe myocardial infraction. RESULTS: PDA NPs loaded with Fer-1 were successfully prepared with good safety and biocompatibility. Administration of PDA NPs carrying Fer-1 notably alleviated myocardial injury and hindered the process of ferroptosis in MIRI rats when inducing downregulation of NOX4 expression. Additionally, overexpression of GPX4 significantly attenuated myocardial injury in MIRI rats. While Fer-1 was shown to inhibit the expression of NOX4, the NOX4 inhibitor Fulvene-5 greatly elevated GPX4 and FTH1 expression in cardiomyocytes, and down-regulated the content of Fe2+, especially in the nanometer+Fer-1+Fulvene-5 group. CONCLUSION: With promising safety and biocompatibility, PDA NPs encapsulated Fer-1 decrease GPX4 and FTH1 expression by inhibiting the level of NOX4 in myocardial cells of MIRI rats, thereby suppressing ferroptosis of cardiomyocytes and alleviating myocardial injury.


Ferroptosis , Indoles , Myocardial Reperfusion Injury , NADPH Oxidase 4 , Nanoparticles , Phospholipid Hydroperoxide Glutathione Peroxidase , Polymers , Animals , NADPH Oxidase 4/metabolism , Myocardial Reperfusion Injury/drug therapy , Indoles/pharmacology , Ferroptosis/drug effects , Rats , Polymers/chemistry , Nanoparticles/chemistry , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Male , Rats, Sprague-Dawley , Cyclohexylamines/pharmacology , Down-Regulation/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Apoptosis/drug effects , Phenylenediamines
3.
Ecotoxicol Environ Saf ; 279: 116481, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38788562

Manganese (Mn) overexposure has been associated with the development of neurological damage reminiscent of Parkinson's disease, while the underlying mechanisms have yet to be fully characterized. This study aimed to investigate the mechanisms leading to injury in dopaminergic neurons induced by Mn and identify novel treatment approaches. In the in vivo and in vitro models, ICR mice and dopaminergic neuron-like PC12 cells were exposed to Mn, respectively. We treated them with anti-ferroptotic agents ferrostatin-1 (Fer-1), deferoxamine (DFO), HIF-1α activator dimethyloxalylglycine (DMOG) and inhibitor LW6. We also used p53-siRNA to verify the mechanism underlying Mn-induced neurotoxicity. Fe and Mn concentrations increased in ICR mice brains overexposed to Mn. Additionally, Mn-exposed mice exhibited movement impairment and encephalic pathological changes, with decreased HIF-1α, SLC7A11, and GPX4 proteins and increased p53 protein levels. Fer-1 exhibited protective effects against Mn-induced both behavioral and biochemical changes. Consistently, in vitro, Mn exposure caused ferroptosis-related changes and decreased HIF-1α levels, all ameliorated by Fer-1. Upregulation of HIF-1α by DMOG alleviated the Mn-associated ferroptosis, while LW6 exacerbated Mn-induced neurotoxicity through downregulating HIF-1α. p53 knock-down also rescued Mn-induced ferroptosis without altering HIF-1α protein expression. Mn overexposure resulted in ferroptosis in dopaminergic neurons, mediated through the HIF-1α/p53/SLC7A11 pathway.


Amino Acid Transport System y+ , Brain , Ferroptosis , Hypoxia-Inducible Factor 1, alpha Subunit , Manganese , Mice, Inbred ICR , Tumor Suppressor Protein p53 , Animals , Ferroptosis/drug effects , PC12 Cells , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Manganese/toxicity , Brain/drug effects , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Rats , Male , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Cyclohexylamines/pharmacology , Phenylenediamines/toxicity , Phenylenediamines/pharmacology , Deferoxamine/pharmacology , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Amino Acids, Dicarboxylic
4.
Free Radic Biol Med ; 220: 271-287, 2024 Aug 01.
Article En | MEDLINE | ID: mdl-38734267

Bilirubin-induced brain damage is a serious clinical consequence of hyperbilirubinemia, yet the underlying molecular mechanisms remain largely unknown. Ferroptosis, an iron-dependent cell death, is characterized by iron overload and lipid peroxidation. Here, we report a novel regulatory mechanism of demethylase AlkB homolog 5 (ALKBH5) in acyl-coenzyme A synthetase long-chain family member 4 (ACSL4)-mediated ferroptosis in hyperbilirubinemia. Hyperdifferential PC12 cells and newborn Sprague-Dawley rats were used to establish in vitro and in vivo hyperbilirubinemia models, respectively. Proteomics, coupled with bioinformatics analysis, first suggested the important role of ferroptosis in hyperbilirubinemia-induced brain damage. In vitro experiments showed that ferroptosis is activated in hyperbilirubinemia, and ferroptosis inhibitors (desferrioxamine and ferrostatin-1) treatment effectively alleviates hyperbilirubinemia-induced oxidative damage. Notably, we observed that the ferroptosis in hyperbilirubinemia was regulated by m6A modification through the downregulation of ALKBH5 expression. MeRIP-seq and RIP-seq showed that ALKBH5 may trigger hyperbilirubinemia ferroptosis by stabilizing ACSL4 mRNA via m6A modification. Further, hyperbilirubinemia-induced oxidative damage was alleviated through ACSL4 genetic knockdown or rosiglitazone-mediated chemical repression but was exacerbated by ACSL4 overexpression. Mechanistically, ALKBH5 promotes ACSL4 mRNA stability and ferroptosis by combining the 669 and 2015 m6A modified sites within 3' UTR of ACSL4 mRNA. Our findings unveil a novel molecular mechanism of ferroptosis and suggest that m6A-dependent ferroptosis could be an underlying clinical target for the therapy of hyperbilirubinemia.


AlkB Homolog 5, RNA Demethylase , Coenzyme A Ligases , Ferroptosis , RNA Stability , Rats, Sprague-Dawley , Animals , Ferroptosis/genetics , Rats , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , AlkB Homolog 5, RNA Demethylase/metabolism , AlkB Homolog 5, RNA Demethylase/genetics , PC12 Cells , Cyclohexylamines/pharmacology , Humans , Deferoxamine/pharmacology , Oxidative Stress , Brain Injuries/metabolism , Brain Injuries/genetics , Brain Injuries/pathology , Brain Injuries/etiology , Phenylenediamines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Male , Disease Models, Animal , Lipid Peroxidation
5.
J Dent Res ; 103(7): 712-722, 2024 Jul.
Article En | MEDLINE | ID: mdl-38766865

Inflammation and loss of articular cartilage are considered the major cause of temporomandibular joint osteoarthritis (TMJOA), a painful condition of the temporomandibular joint (TMJ). To determine the cause of TMJ osteoarthritis in these patients, synovial fluid of TMJOA patients was compared prior to and after hyaluronic lavage, revealing substantially elevated levels of interleukin (IL) 1ß, reactive oxidative stress (ROS), and an overload of Fe3+ and Fe2+ prior to lavage, indicative of ferroptosis as a mode of chondrocyte cell death. To ask whether prolonged inflammatory conditions resulted in ferroptosis-like transformation in vitro, we subjected TMJ chondrocytes to IL-1ß treatment, resulting in a shift in messenger RNA sequencing gene ontologies related to iron homeostasis and oxidative stress-related cell death. Exposure to rat unilateral anterior crossbite conditions resulted in reduced COL2A1 expression, fewer chondrocytes, glutathione peroxidase 4 (GPX4) downregulation, and 4-hydroxynonenal (4-HNE) upregulation, an effect that was reversed after intra-articular injections of the ferroptosis inhibitor ferrostatin 1 (Fer-1). Our study demonstrated that ferroptosis conditions affected mitochondrial structure and function, while the inhibitor Fer-1 restored mitochondrial structure and the inhibition of hypoxia-inducible factor 1α (HIF-1α) or the transferrin receptor 1 (TFRC) rescued IL-1ß-induced loss of mitochondrial membrane potential. Inhibition of HIF-1α downregulated IL-1ß-induced TFRC expression, while inhibition of TFRC did not downregulate IL-1ß-induced HIF-1α expression in chondrocytes. Moreover, inhibition of HIF-1α or TFRC downregulated the IL-1ß-induced MMP13 expression in chondrocytes, while inhibition of HIF-1α or TFRC rescued IL-1ß-inhibited COL2A1 expression in chondrocytes. Furthermore, upregulation of TFRC promoted Fe2+ entry into chondrocytes, inducing the Fenton reaction and lipid peroxidation, which in turn caused ferroptosis, a disruption in chondrocyte functions, and an exacerbation of condylar cartilage degeneration. Together, these findings illustrate the far-reaching effects of chondrocyte ferroptosis in TMJOA as a mechanism causing chondrocyte death through iron overload, oxidative stress, and articular cartilage degeneration and a potential major cause of TMJOA.


Chondrocytes , Ferroptosis , Hypoxia-Inducible Factor 1, alpha Subunit , Interleukin-1beta , Osteoarthritis , Oxidative Stress , Receptors, Transferrin , Temporomandibular Joint Disorders , Chondrocytes/metabolism , Chondrocytes/drug effects , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Rats , Receptors, Transferrin/metabolism , Osteoarthritis/metabolism , Temporomandibular Joint Disorders/metabolism , Male , Humans , Rats, Sprague-Dawley , Inflammation , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Temporomandibular Joint/metabolism , Temporomandibular Joint/pathology , Cyclohexylamines/pharmacology , Cartilage, Articular/metabolism , Collagen Type II , Reactive Oxygen Species/metabolism , Female , Aldehydes , Phenylenediamines
6.
Acta Biomater ; 181: 362-374, 2024 06.
Article En | MEDLINE | ID: mdl-38663684

Ferroptosis induced by lipid peroxide (LPO) accumulation is an effective cell death pathway for cancer therapy. However, how to effectively induce ferroptosis at tumor sites and improve its therapeutic effectiveness remains challenging. Here, MnFe2O4@NaGdF4@NLG919@HA (MGNH) nanocomplex with tumor-specific targeting and TME response is constructed to overcome immunosuppressive tumor microenvironment (TME) to potentiate the curative effect of ferroptosis by coupling the immune checkpoint indoleamine 2,3-dioxygenase (IDO) inhibitor, NLG919, and hyaluronic acid (HA) to novel ultra-small MnFe2O4@NaGdF4 (MG) nanoparticles with a Janus structure. Firstly, tumor site-precise delivery of MG and NLG919 is achieved with HA targeting. Secondly, MG acts as a magnetic resonance imaging contrast agent, which not only has a good photothermal effect to realize tumor photothermal therapy, but also depletes glutathione and catalyzes the production of reactive oxygen species from endogenous H2O2, which effectively promotes the accumulation of LPO and inhibits the expression of glutathione peroxidase 4, achieving enhanced ferroptosis. Thirdly, NLG919 inhibits the differentiation of Tregs by blocking the tryptophan/kynurenine immune escape pathway, thereby reversing immunosuppressive TME together with the Mn2+-activated cGAS-STING pathway. This work contributes new perspectives for the development of novel ultra-small Janus nanoparticles to reshape immunosuppressive TME and ferroptosis activation. STATEMENT OF SIGNIFICANCE: The Janus structured MnFe2O4@NaGdF4@NLG919@HA (MGNH) nanocomplex was synthesized, which can realize the precise delivery of T1/T2 contrast agents MnFe2O4@NaGdF4 (MG) and NLG919 at the tumor site under the ultra-small Janus structural characteristics and targeted molecule HA. The production of ROS, consumption of GSH, and photothermal properties of MGNH make it possible for CDT/PTT activated ferroptosis, and synergistically disrupt and reprogram tumor growth and immunosuppressive tumor microenvironment with NLG919 and Mn2+-mediated activation of cGAS-STING pathway, achieving CDT/PTT/immunotherapy activated by ferroptosis. Meanwhile, ultra-small structural properties of MGNH facilitate subsequent metabolic clearance by the body, allowing for the minimization of potential biotoxicity associated with its prolonged retention.


Ferroptosis , Immunotherapy , Nanoparticles , Tumor Microenvironment , Ferroptosis/drug effects , Immunotherapy/methods , Animals , Nanoparticles/chemistry , Mice , Tumor Microenvironment/drug effects , Humans , Cell Line, Tumor , Neoplasms/pathology , Neoplasms/therapy , Neoplasms/drug therapy , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Cyclohexylamines/pharmacology , Cyclohexylamines/chemistry , Imidazoles , Isoindoles
7.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167182, 2024 Jun.
Article En | MEDLINE | ID: mdl-38653359

OBJECTIVE: This work aimed to investigate the role of rhythm gene PER1 in mediating granulosa cell ferroptosis and lipid metabolism of polycystic ovary syndrome (PCOS). METHODS: We injected dehydroepiandrosterone and Ferrostatin-1 (Fer-1) into mice to explore the mechanism of ferroptosis in PCOS. The effect of PER1 on ferroptosis-like changes in granulosa cells was explored by overexpression of PER1 plasmid transfection and Fer-1 treatment. RESULTS: We found that Fer-1 ameliorated the characteristic polycystic ovary morphology, suppressed ferroptosis in the PCOS mice. PER1 and ALOX15 were highly expressed in PCOS, whereas SREBF2 was lowly expressed. Overexpression of PER1 decreased granulosa cell viability and inhibited proliferation. Meanwhile, overexpression of PER1 increased lipid reactive oxygen species, 4-Hydroxynonenal (4-HNE), Malondialdehyde (MDA), total Fe, and Fe2+ levels in granulosa cells and decreased Glutathione (GSH) content. Fer-1, SREBF2 overexpression, or ALOX15 silencing treatment reversed the effects of PER1 overexpression on granulosa cells. PER1 binds to the SREBF2 promoter and represses SREBF2 transcription. SREBF2 binds to the ALOX15 promoter and represses ALOX15 transcription. Correlation analysis of clinical trials showed that PER1 was positively correlated with total cholesterol, low-density lipoprotein cholesterol, luteinizing hormone, testosterone, 4-HNE, MDA, total Fe, Fe2+, and ALOX15. In contrast, PER1 was negatively correlated with SREBF2, high-density lipoprotein cholesterol, follicle-stimulating hormone, progesterone, and GSH. CONCLUSION: This study demonstrates that the rhythm gene PER1 promotes ferroptosis and dysfunctional lipid metabolism in granulosa cells in PCOS by inhibiting SREBF2/ALOX15 signaling.


Ferroptosis , Granulosa Cells , Lipid Metabolism , Polycystic Ovary Syndrome , Animals , Female , Humans , Mice , Arachidonate 12-Lipoxygenase , Arachidonate 15-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/genetics , Cyclohexylamines/pharmacology , Dehydroepiandrosterone/metabolism , Ferroptosis/genetics , Granulosa Cells/metabolism , Granulosa Cells/pathology , Lipid Metabolism/genetics , Phenylenediamines/pharmacology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/pathology , Reactive Oxygen Species/metabolism
8.
Free Radic Biol Med ; 218: 26-40, 2024 Jun.
Article En | MEDLINE | ID: mdl-38570172

Nuclear factor erythroid 2-related factor 2 (Nrf2) plays a crucial role in ferroptosis by regulating the cellular antioxidant response and maintaining redox balance. However, compounds that induce ferroptosis through dual antioxidant pathways based on Nrf2 have not been fully explored. In our study, we investigated the impact of Gambogic acid (GA) on MCF-7 cells and HepG2 cells in vitro. The cytotoxicity, colony formation assay and cell cycle assay demonstrated potent tumor-killing ability of GA, while its effect was rescued by ferroptosis inhibitors. Furthermore, RNA sequencing revealed the enrichment of ferroptosis pathway mediated by GA. In terms of ferroptosis indicators detection, evidences for GA were provided including reactive oxygen species (ROS) accumulation, alteration in mitochondrial membrane potential (MMP), disappearance of mitochondrial cristae, lipid peroxidation induction, malondialdehyde (MDA) accumulation promotion, iron ion accumulation as well as glutathione (GSH)/thioredoxin (Trx) depletion. Notably, Ferrostatin-1 (Fer-1) and Liproxstatin-1 (Lip-1) successfully rescued GA-induced MDA accumulation. In terms of mechanism, Nrf2 was found to play a pivotal role in GA-induced ferroptosis by inducing protein alterations through the iron metabolism pathway and GSH/Trx dual antioxidant pathway. Furthermore, GA exerted good antitumor activity in vivo through GSH/Trx dual antioxidant pathway, and Fer-1 significantly attenuated its efficacy. In conclusion, our findings first provided new evidence for GA as an inducer of ferroptosis, and Nrf2-mediated GSH/Trx dual antioxidant system played an important role in GA-induced ferroptosis.


Antioxidants , Ferroptosis , Glutathione , NF-E2-Related Factor 2 , Quinoxalines , Reactive Oxygen Species , Spiro Compounds , Xanthones , Ferroptosis/drug effects , Xanthones/pharmacology , Humans , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Glutathione/metabolism , Animals , Antioxidants/pharmacology , Reactive Oxygen Species/metabolism , Mice , MCF-7 Cells , Hep G2 Cells , Xenograft Model Antitumor Assays , Membrane Potential, Mitochondrial/drug effects , Antineoplastic Agents/pharmacology , Lipid Peroxidation/drug effects , Cyclohexylamines/pharmacology , Phenylenediamines/pharmacology , Cell Proliferation/drug effects
9.
Sci Rep ; 14(1): 9548, 2024 04 25.
Article En | MEDLINE | ID: mdl-38664508

Ferroptosis is closely associated with inflammatory diseases, including acute pancreatitis (AP); however, the involvement of ferroptosis in hypertriglyceridemic pancreatitis (HTGP) remains unclear. In the present study, we aimed to explore the relationship between lipid metabolism and ferroptosis in HTGP and the alleviating effect of liproxstatin-1 (Lip-1) in vivo. This study represents the first exploration of lipid metabolism and endoplasmic reticulum stress (ERS) in HTGP, targeting ferroptosis as a key factor in HTGP. Hypertriglyceridemia (HTG) was induced under high-fat diet conditions. Cerulein was then injected to establish AP and HTGP models. Lip-1, a specific ferroptosis inhibitor, was administered before the induction of AP and HTGP in rats, respectively. Serum triglyceride, amylase, inflammatory factors, pathological and ultrastructural structures, lipid peroxidation, and iron overload indicators related to ferroptosis were tested. Moreover, the interaction between ferroptosis and ERS was assessed. We found HTG can exacerbate the development of AP, with an increased inflammatory response and intensified ferroptosis process. Lip-1 treatment can attenuate pancreatic injury by inhibiting ferroptosis through lipid metabolism and further resisting activations of ERS-related proteins. Totally, our results proved lipid metabolism can promote ferroptosis in HTGP by regulating ACSL4/LPCAT3 protein levels. Additionally, ERS may participate in ferroptosis via the Bip/p-EIF2α/CHOP pathway, followed by the alleviating effect of Lip-1 in the rat model.


Endoplasmic Reticulum Stress , Ferroptosis , Hypertriglyceridemia , Lipid Metabolism , Pancreatitis , Quinoxalines , Spiro Compounds , Animals , Ferroptosis/drug effects , Pancreatitis/drug therapy , Pancreatitis/metabolism , Pancreatitis/pathology , Hypertriglyceridemia/drug therapy , Hypertriglyceridemia/metabolism , Rats , Endoplasmic Reticulum Stress/drug effects , Male , Lipid Metabolism/drug effects , Cyclohexylamines/pharmacology , Disease Models, Animal , Rats, Sprague-Dawley , Lipid Peroxidation/drug effects , Diet, High-Fat/adverse effects , Pancreas/drug effects , Pancreas/pathology , Pancreas/metabolism , Triglycerides/blood , Triglycerides/metabolism
10.
J Pharmacol Sci ; 155(2): 44-51, 2024 Jun.
Article En | MEDLINE | ID: mdl-38677785

Subretinal hemorrhages result in poor vision and visual field defects. During hemorrhage, several potentially toxic substances are released from iron-based hemoglobin and hemin, inducing cellular damage, the detailed mechanisms of which remain unknown. We examined the effects of excess intracellular iron on retinal pigment epithelial (RPE) cells. A Fe2+ probe, SiRhoNox-1 was used to investigate Fe2+ accumulation after treatment with hemoglobin or hemin in the human RPE cell line ARPE-19. We also evaluated the production of reactive oxygen species (ROS) and lipid peroxidation. Furthermore, the protective effect of-an iron chelator, 2,2'-bipyridyl (BP), and ferrostatin-1 (Fer-1) on the cell damage, was evaluated. Fe2+ accumulation increased in the hemoglobin- or hemin-treated groups, as well as intracellular ROS production and lipid peroxidation. In contrast, BP treatment suppressed RPE cell death, ROS production, and lipid peroxidation. Pretreatment with Fer-1 ameliorated cell death in a concentration-dependent manner and suppressed ROS production and lipid peroxidation. Taken together, these findings indicate that hemoglobin and hemin, as well as subretinal hemorrhage, may induce RPE cell damage and visual dysfunction via intracellular iron accumulation.


Hemin , Hemoglobins , Iron , Retinal Pigment Epithelium , Humans , Cell Death/drug effects , Cell Line , Cyclohexylamines/pharmacology , Hemin/pharmacology , Hemoglobins/metabolism , Iron/metabolism , Iron Chelating Agents/pharmacology , Lipid Peroxidation/drug effects , Phenylenediamines/pharmacology , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology
11.
Bioorg Med Chem ; 105: 117716, 2024 May 01.
Article En | MEDLINE | ID: mdl-38608329

In this study, a series of new formylpiperazine-derived ferroptosis inhibitors were designed and synthesized based on the structure of a known ferroptosis inhibitor, ferrostatin-1 (Fer-1). The anti-ferroptosis activity of these synthetic compounds in human umbilical vein endothelial cells (HUVECs) induced by Erastin was evaluated. It was found that some of the new compounds, especially compound 26, showed potent anti-ferroptosis activity, as evidenced by its ability to restore cell viability, reduce iron accumulation, scavenge reactive oxygen species, maintain mitochondrial membrane potential, increase GSH levels, decrease LPO and MDA content, and upregulate GPX4 expression. Moreover, compound 26 exhibited superior microsomal stability than Fer-1. The present results suggest that compound 26 is a promising lead compound for the development of new ferroptosis inhibitors for the treatment of vascular diseases.


Cell Survival , Cyclohexylamines , Drug Design , Ferroptosis , Human Umbilical Vein Endothelial Cells , Piperazines , Humans , Ferroptosis/drug effects , Piperazines/pharmacology , Piperazines/chemical synthesis , Piperazines/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Structure-Activity Relationship , Cyclohexylamines/pharmacology , Cyclohexylamines/chemistry , Cyclohexylamines/chemical synthesis , Cell Survival/drug effects , Molecular Structure , Phenylenediamines/pharmacology , Phenylenediamines/chemistry , Phenylenediamines/chemical synthesis , Dose-Response Relationship, Drug , Reactive Oxygen Species/metabolism , Ferrous Compounds/pharmacology , Ferrous Compounds/chemistry , Ferrous Compounds/chemical synthesis , Membrane Potential, Mitochondrial/drug effects
12.
J Integr Med ; 22(3): 286-294, 2024 May.
Article En | MEDLINE | ID: mdl-38565435

OBJECTIVE: Research has shown that celastrol can effectively treat a variety of diseases, yet when passing a certain dosage threshold, celastrol becomes toxic, causing complications such as liver and kidney damage and erythrocytopenia, among others. With this dichotomy in mind, it is extremely important to find ways to preserve celastrol's efficacy while reducing or preventing its toxicity. METHODS: In this study, insulin-resistant HepG2 (IR-HepG2) cells were prepared using palmitic acid and used for in vitro experiments. IR-HepG2 cells were treated with celastrol alone or in combination with N-acetylcysteine (NAC) or ferrostatin-1 (Fer-1) for 12, 24 or 48 h, at a range of doses. Cell counting kit-8 assay, Western blotting, quantitative reverse transcription-polymerase chain reaction, glucose consumption assessment, and flow cytometry were performed to measure celastrol's cytotoxicity and whether the cell death was linked to ferroptosis. RESULTS: Celastrol treatment increased lipid oxidation and decreased expression of anti-ferroptosis proteins in IR-HepG2 cells. Celastrol downregulated glutathione peroxidase 4 (GPX4) mRNA. Molecular docking models predicted that solute carrier family 7 member 11 (SLC7A11) and GPX4 were covalently bound by celastrol. Importantly, we found for the first time that the application of ferroptosis inhibitors (especially NAC) was able to reduce celastrol's toxicity while preserving its ability to improve insulin sensitivity in IR-HepG2 cells. CONCLUSION: One potential mechanism of celastrol's cytotoxicity is the induction of ferroptosis, which can be alleviated by treatment with ferroptosis inhibitors. These findings provide a new strategy to block celastrol's toxicity while preserving its therapeutic effects. Please cite this article as: Liu JJ, Zhang X, Qi MM, Chi YB, Cai BL, Peng B, Zhang DH. Ferroptosis inhibitors reduce celastrol toxicity and preserve its insulin sensitizing effects in insulin resistant HepG2 cells. J Integr Med. 2024; 22(3): 286-294.


Ferroptosis , Insulin Resistance , Pentacyclic Triterpenes , Humans , Hep G2 Cells , Pentacyclic Triterpenes/pharmacology , Ferroptosis/drug effects , Triterpenes/pharmacology , Cyclohexylamines/pharmacology , Acetylcysteine/pharmacology , Phenylenediamines/pharmacology , Molecular Docking Simulation , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
13.
Exp Cell Res ; 438(1): 114029, 2024 May 01.
Article En | MEDLINE | ID: mdl-38608805

Aberrant expression of airway epithelial E-cadherin is a key feature of asthma, yet the underlying mechanisms are largely unknown. Ferroptosis is a novel form of regulated cell death involved in asthma pathogenesis. This study was aimed to evaluate the role of ferroptosis and to investigate whether ferroptosis mediates E-cadherin disruption in mixed granulocyte asthma (MGA). Two murine models of MGA were established using toluene diisocyanate (TDI) or ovalbumin with Complete Freund's Adjuvant (OVA/CFA). Specific antagonists of ferroptosis, including Liproxstatin-1 (Lip-1) and Ferrostatin-1 (Fer-1) were given to the mice. The allergen-exposed mice displayed markedly shrunk mitochondria in the airway epithelia, with decreased volume and denser staining accompanied by down-regulated GPX4 as well as up-regulated FTH1 and malondialdehyde, which are markers of ferroptosis. Decreased pulmonary expression of E-cadherin was also observed, with profound loss of membrane E-cadherin in the airway epithelia, as well as increased secretion of sE-cadherin. Treatment with Lip-1 not only showed potent protective effects against the allergen-induced airway hyperresponsiveness and inflammatory responses, but also rescued airway epithelial E-cadherin expression and inhibited the release of sE-cadherin. Taken together, our data demonstrated that ferroptosis mediates airway epithelial E-cadherin dysfunction in MGA.


Asthma , Cadherins , Disease Models, Animal , Ferroptosis , Granulocytes , Animals , Female , Mice , Asthma/metabolism , Asthma/pathology , Asthma/chemically induced , Cadherins/metabolism , Cyclohexylamines/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/drug effects , Ferroptosis/drug effects , Granulocytes/metabolism , Granulocytes/pathology , Mice, Inbred BALB C , Ovalbumin , Phenylenediamines/pharmacology , Quinoxalines , Spiro Compounds
14.
Angew Chem Int Ed Engl ; 63(21): e202402537, 2024 May 21.
Article En | MEDLINE | ID: mdl-38509827

Research on ferroptosis in myocardial ischemia/reperfusion injury (MIRI) using mitochondrial viscosity as a nexus holds great promise for MIRI therapy. However, high-precision visualisation of mitochondrial viscosity remains a formidable task owing to the debilitating electrostatic interactions caused by damaged mitochondrial membrane potential. Herein, we propose a dual-locking mitochondria-targeting strategy that incorporates electrostatic forces and probe-protein molecular docking. Even in damaged mitochondria, stable and precise visualisation of mitochondrial viscosity in triggered and medicated MIRI was achieved owing to the sustained driving forces (e.g., pi-cation, pi-alkyl interactions, etc.) between the developed probe, CBS, and the mitochondrial membrane protein. Moreover, complemented by a western blot, we confirmed that ferrostatin-1 exerts its therapeutic effect on MIRI by improving the system xc-/GSH/GPX4 antioxidant system, confirming the therapeutic value of ferroptosis in MIRI. This study presents a novel strategy for developing robust mitochondrial probes, thereby advancing MIRI treatment.


Ferroptosis , Myocardial Reperfusion Injury , Ferroptosis/drug effects , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Molecular Docking Simulation , Animals , Mitochondria/metabolism , Mitochondria/drug effects , Humans , Cyclohexylamines/chemistry , Cyclohexylamines/pharmacology , Phenylenediamines/chemistry , Phenylenediamines/pharmacology
15.
Aging (Albany NY) ; 16(7): 5987-6007, 2024 Mar 25.
Article En | MEDLINE | ID: mdl-38536018

Ferroptosis is a specific type of programmed cell death characterized by iron-dependent lipid peroxidation. Understanding the involvement of ferroptosis in calcium oxalate (CaOx) stone formation may reveal potential targets for this condition. The publicly available dataset GSE73680 was used to identify 61 differentially expressed ferroptosis-related genes (DEFERGs) between normal kidney tissues and Randall's plaques (RPs) from patients with nephrolithiasis through employing weighted gene co-expression network analysis (WGCNA). The findings were validated through in vitro and in vivo experiments using CaOx nephrolithiasis rat models induced by 1% ethylene glycol administration and HK-2 cell models treated with 1 mM oxalate. Through WGCNA and the machine learning algorithm, we identified LAMP2 and MDM4 as the hub DEFERGs. Subsequently, nephrolithiasis samples were classified into cluster 1 and cluster 2 based on the expression of the hub DEFERGs. Validation experiments demonstrated decreased expression of LAMP2 and MDM4 in CaOx nephrolithiasis animal models and cells. Treatment with ferrostatin-1 (Fer-1), a ferroptosis inhibitor, partially reversed oxidative stress and lipid peroxidation in CaOx nephrolithiasis models. Moreover, Fer-1 also reversed the expression changes of LAMP2 and MDM4 in CaOx nephrolithiasis models. Our findings suggest that ferroptosis may be involved in the formation of CaOx kidney stones through the regulation of LAMP2 and MDM4.


Biomarkers , Ferroptosis , Nephrolithiasis , Ferroptosis/drug effects , Animals , Nephrolithiasis/metabolism , Nephrolithiasis/genetics , Nephrolithiasis/pathology , Rats , Biomarkers/metabolism , Humans , Male , Calcium Oxalate/metabolism , Lipid Peroxidation/drug effects , Oxidative Stress/drug effects , Kidney Calculi/metabolism , Kidney Calculi/genetics , Kidney Calculi/pathology , Cyclohexylamines/pharmacology , Phenylenediamines/pharmacology , Disease Models, Animal , Rats, Sprague-Dawley , Cell Line
16.
Biol Reprod ; 110(5): 1012-1024, 2024 May 09.
Article En | MEDLINE | ID: mdl-38320204

Cyclophosphamide (CP) is a widely used chemotherapeutic drug and immunosuppressant in the clinic, and the hypoandrogenism caused by CP is receiving more attention. Some studies found that ferroptosis is a new mechanism of cell death closely related to chemotherapeutic drugs and plays a key role in regulating reproductive injuries. The purpose of this study is to explore ferroptosis' role in testicular Leydig cell dysfunction and molecular mechanisms relating to it. In this study, the level of ferroptosis in the mouse model of testicular Leydig cell dysfunction induced by CP was significantly increased and further affected testosterone synthesis. The ferroptosis inhibitors ferrostatin-1 (Fer-1) and iron chelator deferoxamine (DFO) can improve injury induced by CP. The results of immunohistochemistry showed that Fer-1 and DFO could improve the structural disorder of seminiferous tubules and the decrease of the number of Leydig cells in testicular tissue induced by CP. Immunofluorescence and western blot confirmed that Fer-1 and DFO could improve the expression of key enzymes in testosterone synthesis. The activation of SMAD family member 2 (Smad2)/cyclin-dependent kinase inhibitor 1A (Cdkn1a) pathway can improve the ferroptosis of Leydig cells induced by CP and protect the function of Leydig cells. By inhibiting the Smad2/Cdkn1a signal pathway, CP can regulate ferroptosis, resulting in testicular Leydig cell dysfunction. In this study, CP-induced hypoandrogenism is explained theoretically and a potential therapeutic strategy is provided.


Cyclophosphamide , Ferroptosis , Leydig Cells , Smad2 Protein , Animals , Male , Ferroptosis/drug effects , Leydig Cells/drug effects , Leydig Cells/metabolism , Mice , Cyclophosphamide/toxicity , Smad2 Protein/metabolism , Signal Transduction/drug effects , Cyclohexylamines/pharmacology , Phenylenediamines/pharmacology , Testis/drug effects , Testis/metabolism , Testis/pathology
17.
FASEB J ; 38(2): e23401, 2024 01 31.
Article En | MEDLINE | ID: mdl-38236196

Ferroptosis, a type of iron-catalyzed necrosis, is responsible for vascular smooth muscle cell (VSMC) death and serves as a potential therapeutic target for alleviating aortic aneurysm. Here, our study explored the underlying mechanism of ferroptosis affecting VSMC functions and the resultant formation of AAA using its inhibitor Ferrostatin-1 (Fer-1). Microarray-based gene expression profiling was employed to identify differentially expressed genes related to AAA and ferroptosis. An AAA model was established by angiotensin II (Ang II) induction in apolipoprotein E-knockout (ApoE-/- ) mice, followed by injection of Fer-1 and RSL-3 (ferroptosis inducer). Then, the role of Fer-1 and RSL-3 in the ferroptosis of VSMCs and AAA formation was analyzed in Ang II-induced mice. Primary mouse VSMCs were cultured in vitro and treated with Ang II, Fer-1, sh-SLC7A11, or sh-GPX4 to assess the effect of Fer-1 via the SLC7A11/GPX axis. Bioinformatics analysis revealed that GPX4 was involved in the fibrosis formation of AAA, and there was an interaction between SLC7A11 and GPX4. In vitro assays showed that Fer-1 alleviated Ang II-induced ferroptosis of VSMCs and retard the consequent AAA formation. The mechanism was associated with activation of the SLC7A11/GPX4 pathway. Silencing of SLC7A11 or GPX4 could inhibit the ameliorating effect of Fer-1 on the ferroptosis of VSMCs. In vivo animal studies further demonstrated that Fer-1 inhibited Ang II-induced ferroptosis and vessel wall structural abnormalities in AAA mouse through activation of the SLC7A11/GPX4 pathway. Fer-1 may prevent AAA formation through activation of the SLC7A11/GPX4 pathway.


Aortic Aneurysm, Abdominal , Ferroptosis , Peptide Hormones , Phenylenediamines , Animals , Mice , Muscle, Smooth, Vascular , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/prevention & control , Cyclohexylamines/pharmacology , Angiotensin II/pharmacology
18.
PeerJ ; 11: e15786, 2023.
Article En | MEDLINE | ID: mdl-37701828

Background: Acute kidney injury (AKI) is a common and serious medical condition with high morbidity and mortality. Recent research has highlighted ferroptosis, a novel form of programmed cell death, as a potential therapeutic target in mitigating renal tubular injury in AKI. Ferrostatin-1, a specific ferroptosis inhibitor, has been demonstrated to prevent renal injury through ferroptosis inhibition. Methods: Utilizing a murine AKI model, we investigated the effects of Ferrostatin-1 by administering it post-injury. Through high-throughput sequencing and pathological analysis, we focused on the critical role of ferroptosis-related pathways in the treatment. Results: Ferrostatin-1 post-conditioning effectively mitigated oxidative damage and reduced iron content associated with AKI. Additionally, critical ferroptosis-related proteins, such as GPX4, SLC7A11, NRF2, and FTH1, exhibited increased expression levels. In vitro, Ferrostatin-1 treatment of HK-2 cells significantly diminished lipid peroxidation and iron accumulation. Furthermore, Ferrostatin-1 was found to downregulate the PI3K signalling pathway. Conclusion: Ferrostatin-1 acted as a potential ferroptosis inhibitor with the capacity to enhance antioxidant defences. This study suggests that Ferrostatin-1 could serve as a promising novel strategy for improving the treatment of AKI and promoting recovery from the condition.


Acute Kidney Injury , Ferritins , Animals , Mice , Iron , Cyclohexylamines/pharmacology , Acute Kidney Injury/drug therapy
19.
Trends Pharmacol Sci ; 44(12): 902-916, 2023 12.
Article En | MEDLINE | ID: mdl-37770317

Ferroptosis is an iron-catalysed form of regulated cell death, which is critically dependent on phospholipid peroxidation of cellular membranes. Ferrostatin 1 was one of the first synthetic radical-trapping antioxidants (RTAs) reported to block ferroptosis and it is widely used as reference compound. Ferroptosis has been linked to multiple diseases and the use of its inhibitors could have therapeutic potential. Although, novel biochemical pathways provide insights for different pharmacological targets, the use of lipophilic RTAs to block ferroptosis remains superior. In this Review, we provide a comprehensive overview of the different classes of ferroptosis inhibitors, focusing on endogenous and synthetic RTAs. A thorough analysis of their chemical, pharmacokinetic, and pharmacological properties and potential for in vivo use is provided.


Ferroptosis , Humans , Lipid Peroxidation , Cyclohexylamines/metabolism , Cyclohexylamines/pharmacology , Antioxidants/pharmacology
20.
Redox Biol ; 62: 102703, 2023 06.
Article En | MEDLINE | ID: mdl-37087975

Ferroptosis is defined as cell death triggered by iron-dependent lipid peroxidation that is preventable by antioxidant compounds such as ferrostatin-1. Endogenous suppressors of ferroptosis include FSP-1 and the selenoprotein GPX4, the latter of which directly enzymatically reduces lipid hydroperoxides. Small molecules that trigger ferroptosis include RSL3, ML162, and ML210; these compounds are often used in studies of ferroptosis and are generally considered as GPX4 inhibitors. Here, we found that RSL3 and ML162 completely lack capacity of inhibiting the enzymatic activity of recombinant selenoprotein GPX4. Surprisingly, these compounds were instead found to be efficient inhibitors of another selenoprotein, TXNRD1. Other known inhibitors of TXNRD1, including auranofin, TRi-1 and TRi-2, are also efficient inducers of cell death but that cell death could not be suppressed with ferrostatin-1. Our results collectively suggest that prior studies using RSL3 and ML162 may need to be reevaluated in the context of ferroptosis with regards to additional enzyme targets and mechanisms of action that may be involved.


Ferroptosis , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Cyclohexylamines/pharmacology , Lipid Peroxidation
...