Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.376
Filtrer
1.
Biomed Environ Sci ; 37(6): 617-627, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38988112

RÉSUMÉ

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


Sujet(s)
Modèles animaux de maladie humaine , Ferroptose , Surcharge en fer , Souris de lignée C57BL , Myocytes cardiaques , Silice , Silicose , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Mâle , Souris , Surcharge en fer/métabolisme , Silice/toxicité , Silicose/métabolisme , Silicose/traitement médicamenteux , Silicose/anatomopathologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Déferoxamine/pharmacologie , Phénylènediamines/pharmacologie , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Stress oxydatif/effets des médicaments et des substances chimiques , Fer/métabolisme , Cyclohexylamines/pharmacologie
2.
Ecotoxicol Environ Saf ; 281: 116680, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38964057

RÉSUMÉ

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


Sujet(s)
Ferroptose , Guanidines , Peroxydation lipidique , Souris de lignée C57BL , Phospholipid hydroperoxide glutathione peroxidase , Fibrose pulmonaire , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/anatomopathologie , Souris , Peroxydation lipidique/effets des médicaments et des substances chimiques , Lignée cellulaire , Guanidines/toxicité , Guanidines/pharmacologie , Mâle , Pneumocytes/effets des médicaments et des substances chimiques , Pneumocytes/anatomopathologie , Cyclohexylamines/pharmacologie , Phénylènediamines , Quinoxalines , Spiranes
3.
J Photochem Photobiol B ; 257: 112975, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38970967

RÉSUMÉ

The physiological parameters such as growth, Chl a content, and photosynthetic performance of the experimental cyanobacterium Anabaenopsis circularis HKAR-22 were estimated to evaluate the cumulative effects of photosynthetically active radiation (PAR) and ultraviolet (UV) radiation. Maximum induction of UV-screening molecules, MAAs, was observed under the treatment condition of PAR + UV-A + UV-B (PAB) radiations. UV/VIS absorption spectroscopy and HPLC-PDA detection primarily confirmed the presence of MAA-shinorine (SN) having absorption maxima (λmax) at 332.3 nm and retention time (RT) of 1.47 min. For further validation of the presence of SN, HRMS, FTIR and NMR were utilized. UV-stress elevated the in vivo ROS scavenging and in vitro enzymatic antioxidant capabilities. SN exhibited substantial and concentration-dependent antioxidant capabilities which was determined utilizing 2,2-diphenyl-1-picryl-hydrazyl (DPPH), 2,2'-azinobis-(3-ethylbenzothiazoline-6-sulfonate (ABTS), ferric reducing power (FRAP) and superoxide radical scavenging assay (SRSA). The density functional theory (DFT) method using B3LYP energy model and 6-311G++(d,p) basis set was implied to perform the quantum chemical calculation to systematically investigate the antioxidant nature of SN. The principal pathways involved in the antioxidant reactions along with the basic molecular descriptors affecting the antioxidant potentials of a compound were also studied. The results favor the potential of SN as an active ingredient to be used in cosmeceutical formulations.


Sujet(s)
Antioxydants , Cyanobactéries , Théorie de la fonctionnelle de la densité , Rayons ultraviolets , Antioxydants/composition chimique , Cyanobactéries/composition chimique , Cyanobactéries/métabolisme , Acides aminés/composition chimique , Acides aminés/métabolisme , Cyclohexanones/composition chimique , Photosynthèse , Espèces réactives de l'oxygène/métabolisme , Chlorophylle A/composition chimique , Chlorophylle A/métabolisme , Dérivés du biphényle/composition chimique , Picrates/antagonistes et inhibiteurs , Picrates/composition chimique , Piégeurs de radicaux libres/composition chimique , Cyclohexylamines , Glycine/analogues et dérivés , Acides sulfoniques , Benzothiazoles
4.
J Oleo Sci ; 73(7): 991-999, 2024.
Article de Anglais | MEDLINE | ID: mdl-38945927

RÉSUMÉ

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


Sujet(s)
Ferroptose , Glutathion , Peroxydation lipidique , Oxydoréduction , Phospholipid hydroperoxide glutathione peroxidase , Humains , Cellules HL-60 , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Peroxydation lipidique/effets des médicaments et des substances chimiques , Glutathion/métabolisme , Oxydoréduction/effets des médicaments et des substances chimiques , Déferoxamine/pharmacologie , Cyclohexylamines/pharmacologie , Lipides , Phénylènediamines/pharmacologie , Lipides membranaires/métabolisme , Agents chélateurs du fer/pharmacologie
5.
PeerJ ; 12: e17551, 2024.
Article de Anglais | MEDLINE | ID: mdl-38887622

RÉSUMÉ

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.


Sujet(s)
Cyclohexylamines , Ferroptose , Fibroblastes , Fibrose , Chéloïde , Facteur-2 apparenté à NF-E2 , Phénylènediamines , Phospholipid hydroperoxide glutathione peroxidase , Humains , Ferroptose/effets des médicaments et des substances chimiques , Chéloïde/anatomopathologie , Chéloïde/métabolisme , Chéloïde/traitement médicamenteux , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Cyclohexylamines/pharmacologie , Fibrose/métabolisme , Fibrose/anatomopathologie , Phénylènediamines/pharmacologie , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/génétique , Mâle , Peroxydation lipidique/effets des médicaments et des substances chimiques , Femelle , Adulte , Fer/métabolisme , Système y+ de transport d'acides aminés/métabolisme , Système y+ de transport d'acides aminés/génétique , Récepteurs à la transferrine/métabolisme , Récepteurs à la transferrine/génétique , Pipérazines/pharmacologie , Actines/métabolisme , Actines/génétique , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques
6.
Sheng Wu Gong Cheng Xue Bao ; 40(6): 1882-1894, 2024 Jun 25.
Article de Chinois | MEDLINE | ID: mdl-38914498

RÉSUMÉ

1,4-cyclohexanedimethylamine (1,4-BAC) is an important monomer for bio-based materials, it finds wide applications in various fields including organic synthesis, medicine, chemical industry, and materials. At present, its synthesis primarily relies on chemical method, which suffer from issues such as expensive metal catalyst, harsh reaction conditions, and safety risks. Therefore, it is necessary to explore greener alternatives for its synthesis. In this study, a two-bacterium three-enzyme cascade conversion pathway was successfully developed to convert 1,4-cyclohexanedicarboxaldehyde to 1,4-cyclohexanedimethylamine. This pathway used Escherichia coli derived aminotransferase (EcTA), Saccharomyces cerevisiae derived glutamate dehydrogenase (ScGlu-DH), and Candida boidinii derived formate dehydrogenase (CbFDH). Through structure-guided protein engineering, a beneficial mutant, EcTAF91Y, was obtained, exhibiting a 2.2-fold increase in specific activity and a 1.9-fold increase in kcat/Km compared to that of the wild type. By constructing recombinant strains and optimizing reaction conditions, it was found that under the optimal conditions, a substrate concentration of 40 g/L could produce (27.4±0.9) g/L of the product, corresponding to a molar conversion rate of 67.5%±2.1%.


Sujet(s)
Escherichia coli , Saccharomyces cerevisiae , Escherichia coli/métabolisme , Escherichia coli/génétique , Saccharomyces cerevisiae/métabolisme , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/enzymologie , Transaminases/métabolisme , Transaminases/génétique , Ingénierie des protéines , Glutamate dehydrogenase/métabolisme , Glutamate dehydrogenase/génétique , Formate dehydrogenases/métabolisme , Formate dehydrogenases/génétique , Candida/enzymologie , Candida/métabolisme , Cyclohexylamines/métabolisme
7.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38928480

RÉSUMÉ

Our study aimed to investigate the role of ferroptosis in sevoflurane-induced hearing impairment and explore the mechanism of the microRNA-182-5p (miR-182-5p)/Glutathione Peroxidase 4 (GPX4) pathway in sevoflurane-induced ototoxicity. Immunofluorescence staining was performed using myosin 7a and CtBP2. Cell viability was assessed using the CCK-8 kit. Fe2+ concentration was measured using FerroOrange and Mi-to-FerroGreen fluorescent probes. The lipid peroxide level was assessed using BODIPY 581/591 C11 and MitoSOX fluorescent probes. The auditory brainstem response (ABR) test was conducted to evaluate the hearing status. Bioinformatics tools and dual luciferase gene reporter analysis were used to confirm the direct targeting of miR-182-5p on GPX4 mRNA. GPX4 and miR-182-5p expression in cells was assessed by qRT-PCR and Western blot. Ferrostatin-1 (Fer-1) pretreatment significantly improved hearing impairment and damage to ribbon synapses in mice caused by sevoflurane exposure. Immunofluorescence staining revealed that Fer-1 pretreatment reduced intracellular and mitochondrial iron overload, as well as lipid peroxide accumulation. Our findings indicated that miR-182-5p was upregulated in sevoflurane-exposed HEI-OC1 cells, and miR-182-5p regulated GPX4 expression by binding to the 3'UTR of GPX4 mRNA. The inhibition of miR-182-5p attenuated sevoflurane-induced iron overload and lipid peroxide accumulation. Our study elucidated that the miR-182-5p/GPX4 pathway was implicated in sevoflurane-induced ototoxicity by promoting ferroptosis.


Sujet(s)
Ferroptose , microARN , Ototoxicité , Phospholipid hydroperoxide glutathione peroxidase , Sévoflurane , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/génétique , microARN/génétique , microARN/métabolisme , Sévoflurane/effets indésirables , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/génétique , Animaux , Souris , Ototoxicité/métabolisme , Ototoxicité/étiologie , Transduction du signal/effets des médicaments et des substances chimiques , Lignée cellulaire , Mâle , Perte d'audition/induit chimiquement , Perte d'audition/génétique , Perte d'audition/métabolisme , Perte d'audition/anatomopathologie , Souris de lignée C57BL , Phénylènediamines/pharmacologie , Cyclohexylamines
8.
Cells ; 13(11)2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38891111

RÉSUMÉ

Ferroptosis hallmarked by lipid peroxidation and iron homeostasis imbalance is involved in the occurrence and development of various diseases. The plant growth regulator chlormequat chloride (CCC) can contribute to the causality and exacerbation of reproductive disorders. However, the mechanism by which CCC may cause Leydig cell attenuation remains poorly understood. In this study, TM3 Leydig cells were used to investigate the inhibitory effect of CCC on cell growth and its possible mechanism. The results showed that CCC caused apoptosis, pyroptosis, ferroptosis and necroinflammation in TM3 cells. By comparing the effects of ferroptosis inhibitor Ferrostatin-1 (Fer-1) and pan-Caspase inhibitor Z-VAD-FMK (ZVF) on lipid peroxidation and Caspase-mediated regulated cell death (RCD), we found that Fer-1 was better at rescuing the growth of TM3 cells than ZVF. Although ZVF reduced mitochondrial ROS level and inhibited the activation of Caspase3 and Caspase1, it could not significantly ameliorate lipid peroxidation and the levels of IL-1ß and HMGB1 like Fer-1. Therefore, ferroptosis might be a key non apoptotic RCD mode responsible for CCC-driven inflammation, leading to weakened viability and proliferation of TM3 cells. In addition, overexpression of ferritin light chain (FTL) promoted the resistance of TM3 cells to CCC-induced ferroptosis-mediated inflammation and to some extent improved the inhibition of viability and proliferation. Altogether, ferroptosis-initiated inflammation might play a key role in CCC-impaired TM3 cell growth.


Sujet(s)
Prolifération cellulaire , Ferroptose , Inflammation , Cellules de Leydig , Ferroptose/effets des médicaments et des substances chimiques , Animaux , Mâle , Souris , Cellules de Leydig/effets des médicaments et des substances chimiques , Cellules de Leydig/métabolisme , Cellules de Leydig/anatomopathologie , Inflammation/anatomopathologie , Inflammation/traitement médicamenteux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Peroxydation lipidique/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire , Apoptose/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Chlorométhyl cétones d'acides aminés/pharmacologie , Cyclohexylamines , Phénylènediamines
9.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 36(5): 491-495, 2024 May.
Article de Chinois | MEDLINE | ID: mdl-38845495

RÉSUMÉ

OBJECTIVE: To investigate the effect of nuclear factor E2-related factor 2 (Nrf2) protein on ferroptosis in mice with sepsis-associated liver injury (SALI). METHODS: he male Sprague-Dawley (SD) mice were divided into 6 groups according to the random number table method, with 6 mice in each group. The SALI model of mice was established by cecal ligation and puncture (CLP), and the Sham group was only treated with laparotomy. CLP+Fer-1 group, CLP+Erastin group, CLP+ML385 group and CLP+Curcumin group were intraperitoneally injected with iron death inhibitor Ferrostatin-1 (Fer-1) 10 mg×kg-1×d-1, iron death activator Erastin 20 mg×kg-1×d-1, Nrf2 inhibitor ML385 30 mg×kg-1×d-1 and Nrf2 activator Curcumin 100 mg×kg-1×d-1 after CLP, respectively; Sham group and CLP group were given normal saline 10 mg×kg-1×d-1, each group was administered continuously for 10 days. Ten days after operation, the serum and liver tissues of mice were collected to detect the levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) in serum, and the levels of malondialdehyde (MDA), glutathione (GSH) and Fe2+; in liver homogenate. The pathological changes of liver tissue were observed under light microscope after hematoxylin-eosin (HE) staining. The shape and length of mitochondria in liver cells were observed under transmission electron microscope. The protein expressions of Nrf2, glutathione peroxidase 4 (GPX4) and prostaglandin-endoperoxide synthase 2 (PTGS2) in liver tissue were detected by Western blotting. RESULTS: Compared with Sham group, the serum levels of ALT and AST in the CLP group were significantly increased; histologically, the hepatic cord was disordered, the cells were swollen and necrotic, and the length of mitochondria was significantly shortened; the levels of MDA and Fe2+ in liver tissue increased significantly, and the content of GSH decreased significantly; the protein expressions of Nrf2 and GPX4 in liver tissue decreased, and the protein expression of PTGS2 increased significantly. Compared with CLP group, the serum levels of ALT and AST in CLP+Fer-1 group and CLP+Curcumin group were significantly decreased [ALT (U/L): 80.65±19.44, 103.45±20.52 vs. 283.50±37.12, AST (U/L): 103.33±11.90, 127.33±15.79 vs. 288.67±36.82, all P < 0.05]; microscopically, the hepatic cord was irregular, the cells were slightly swollen, and the mitochondrial length was significantly increased (µm: 1.42±0.09, 1.43±0.21 vs. 1.07±0.25, both P < 0.05); the levels of MDA and Fe2+; in liver tissue decreased significantly, and the content of GSH increased significantly [MDA (mol/g): 0.87±0.23, 1.85±0.43 vs. 4.47±0.95, Fe2+ (µg/g): 63.80±7.15, 67.48±6.28 vs. 134.52±14.32, GSH (mol/g): 1.95±0.29, 1.95±0.45 vs. 0.55±0.29, all P < 0.05]; the protein expressions of Nrf2 and GPX4 in liver tissue were significantly increased, and the protein expression of PTGS2 was significantly decreased (Nrf2/GAPDH: 1.80±0.28, 2.10±0.43 vs. 0.70±0.24, GPX4/GAPDH: 0.80±0.06, 0.93±0.07 vs. 0.48±0.02, PTGS2/GAPDH: 0.76±0.05, 0.84±0.01 vs. 1.02±0.09, all P < 0.05). However, the results of the above indexes in the CLP+Erastin group and CLP+ML385 group were opposite, and the serum levels of ALT and AST were significantly increased [ALT (U/L): 344.52±40.79, 321.70±21.10 vs. 283.50±37.12, AST (U/L): 333.50±27.90, 333.00±16.67 vs. 288.67±36.82, all P < 0.05]; microscopically, the arrangement of hepatic cords was disordered, the cells were obviously swollen and necrotic, and the length of mitochondria was significantly shortened (µm: 0.78±0.13, 0.67±0.07 vs. 1.07±0.25, both P < 0.05); the levels of MDA and Fe2+ in liver tissue increased significantly, and the content of GSH decreased significantly [MDA (mol/g): 5.92±1.06, 5.62±0.56 vs. 4.47±0.95, Fe2+ (µg/g): 151.40±8.03, 151.88±8.68 vs. 134.52±14.32, GSH (mol/g): 0.25±0.08, 0.23±0.11 vs. 0.55±0.29, all P < 0.05]; the protein expressions of Nrf2 and GPX4 in liver tissue were significantly decreased, and the protein expression of PTGS2 was significantly increased (Nrf2/GAPDH: 0.46±0.09, 0.46±0.11 vs. 0.70±0.24, GPX4/GAPDH: 0.34±0.05, 0.40±0.01 vs. 0.48±0.02, PTGS2/GAPDH: 1.24±0.13, 1.16±0.11 vs. 1.02±0.09, all P < 0.05). CONCLUSIONS: CLP-induced SALI can lead to ferroptosis in mice hepatocytes, and Nrf2 protein in liver tissue can mediate SALI by regulating ferroptosis.


Sujet(s)
Ferroptose , Facteur-2 apparenté à NF-E2 , Sepsie , Animaux , Mâle , Souris , Facteur-2 apparenté à NF-E2/métabolisme , Sepsie/métabolisme , Sepsie/complications , Modèles animaux de maladie humaine , Foie/métabolisme , Rat Sprague-Dawley , Maladies du foie/étiologie , Maladies du foie/métabolisme , Glutathione peroxidase/métabolisme , Malonaldéhyde/métabolisme , Curcumine/pharmacologie , Phénylènediamines/pharmacologie , Cyclohexylamines
10.
J Chromatogr A ; 1728: 465014, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-38797135

RÉSUMÉ

Developing novel chiral stationary phases (CSPs) with versatility is of great importance in enantiomer separation. This study fabricated a dual-chiral covalent organic framework (PA-CA COF) via successive post-synthetic modifications. The chiral trans-1,2-cyclohexanediamine (CA) and (D)-penicillamine (PA) groups were periodically aligned within nanochannels of the COF, allowing selective recognition of enantiomers through intermolecular interactions. It can be a versatile high-performance liquid chromatography (HPLC) CSP for separating a wide range of enantiomers, including chiral pharmaceutical intermediates and chiral drugs. With separation performance comparable to commercial chiral columns and even greater versatility, the PA-CA COF@SiO2 column held promise for practical applications. Chiral separation results combined with molecular simulation indicated that the mixed mode of PA and CA resulted in the broad separation capability of PA-CA COF. The introduction of the dual-chiral COFs concept opens up a new avenue for chiral recognition and separation, holding great potential for practical enantiomer separation.


Sujet(s)
Pénicillamine , Stéréoisomérie , Chromatographie en phase liquide à haute performance/méthodes , Pénicillamine/composition chimique , Pénicillamine/isolement et purification , Cyclohexylamines/composition chimique , Cyclohexylamines/isolement et purification , Silice/composition chimique , Réseaux organométalliques/composition chimique
11.
Ecotoxicol Environ Saf ; 279: 116481, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38788562

RÉSUMÉ

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.


Sujet(s)
Système y+ de transport d'acides aminés , Encéphale , Ferroptose , Sous-unité alpha du facteur-1 induit par l'hypoxie , Manganèse , Souris de lignée ICR , Protéine p53 suppresseur de tumeur , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Cellules PC12 , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Souris , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Manganèse/toxicité , Encéphale/effets des médicaments et des substances chimiques , Système y+ de transport d'acides aminés/métabolisme , Système y+ de transport d'acides aminés/génétique , Rats , Mâle , Neurones dopaminergiques/effets des médicaments et des substances chimiques , Neurones dopaminergiques/anatomopathologie , Cyclohexylamines/pharmacologie , Phénylènediamines/toxicité , Phénylènediamines/pharmacologie , Déferoxamine/pharmacologie , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Acides aminés dicarboxyliques
12.
J Dent Res ; 103(7): 712-722, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38766865

RÉSUMÉ

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.


Sujet(s)
Chondrocytes , Ferroptose , Sous-unité alpha du facteur-1 induit par l'hypoxie , Interleukine-1 bêta , Arthrose , Stress oxydatif , Récepteurs à la transferrine , Troubles de l'articulation temporomandibulaire , Chondrocytes/métabolisme , Chondrocytes/effets des médicaments et des substances chimiques , Animaux , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Rats , Récepteurs à la transferrine/métabolisme , Arthrose/métabolisme , Troubles de l'articulation temporomandibulaire/métabolisme , Mâle , Humains , Rat Sprague-Dawley , Inflammation , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Articulation temporomandibulaire/métabolisme , Articulation temporomandibulaire/anatomopathologie , Cyclohexylamines/pharmacologie , Cartilage articulaire/métabolisme , Collagène de type II , Espèces réactives de l'oxygène/métabolisme , Femelle , Aldéhydes , Phénylènediamines
13.
J Mol Cell Cardiol ; 192: 36-47, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38734062

RÉSUMÉ

AIMS: Ferroptosis is a form of iron-regulated cell death implicated in ischemic heart disease. Our previous study revealed that Sirtuin 3 (SIRT3) is associated with ferroptosis and cardiac fibrosis. In this study, we tested whether the knockout of SIRT3 in cardiomyocytes (SIRT3cKO) promotes mitochondrial ferroptosis and whether the blockade of ferroptosis would ameliorate mitochondrial dysfunction. METHODS AND RESULTS: Mitochondrial and cytosolic fractions were isolated from the ventricles of mice. Cytosolic and mitochondrial ferroptosis were analyzed by comparison to SIRT3loxp mice. An echocardiography study showed that SIRT3cKO mice developed heart failure as evidenced by a reduction of EF% and FS% compared to SIRT3loxp mice. Comparison of mitochondrial and cytosolic fractions of SIRT3cKO and SIRT3loxp mice revealed that, upon loss of SIRT3, mitochondrial, but not cytosolic, total lysine acetylation was significantly increased. Similarly, acetylated p53 was significantly upregulated only in the mitochondria. These data demonstrate that SIRT3 is the primary mitochondrial deacetylase. Most importantly, loss of SIRT3 resulted in significant reductions of frataxin, aconitase, and glutathione peroxidase 4 (GPX4) in the mitochondria. This was accompanied by a significant increase in levels of mitochondrial 4-hydroxynonenal. Treatment of SIRT3cKO mice with the ferroptosis inhibitor ferrostatin-1 (Fer-1) for 14 days significantly improved preexisting heart failure. Mechanistically, Fer-1 treatment significantly increased GPX4 and aconitase expression/activity, increased mitochondrial iron­sulfur clusters, and improved mitochondrial membrane potential and Complex IV activity. CONCLUSIONS: Inhibition of ferroptosis ameliorated cardiac dysfunction by specifically targeting mitochondrial aconitase and iron­sulfur clusters. Blockade of mitochondrial ferroptosis may be a novel therapeutic target for mitochondrial cardiomyopathies.


Sujet(s)
Aconitate hydratase , Ferroptose , Souris knockout , Myocytes cardiaques , Phénylènediamines , Sirtuine-3 , Animaux , Sirtuine-3/métabolisme , Sirtuine-3/génétique , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Aconitate hydratase/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Souris , Acétylation , Phénylènediamines/pharmacologie , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Ferrosulfoprotéines/métabolisme , Ferrosulfoprotéines/génétique , Fer/métabolisme , , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/génétique , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/effets des médicaments et des substances chimiques , Protéines de liaison au fer/métabolisme , Protéines de liaison au fer/génétique , Défaillance cardiaque/métabolisme , Défaillance cardiaque/génétique , Cytosol/métabolisme , Cyclohexylamines
14.
Ann Clin Lab Sci ; 54(2): 190-200, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38802151

RÉSUMÉ

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.


Sujet(s)
Ferroptose , Indoles , Lésion de reperfusion myocardique , NADPH Oxidase 4 , Nanoparticules , Phospholipid hydroperoxide glutathione peroxidase , Polymères , Animaux , NADPH Oxidase 4/métabolisme , Lésion de reperfusion myocardique/traitement médicamenteux , Indoles/pharmacologie , Ferroptose/effets des médicaments et des substances chimiques , Rats , Polymères/composition chimique , Nanoparticules/composition chimique , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Mâle , Rat Sprague-Dawley , Cyclohexylamines/pharmacologie , Régulation négative/effets des médicaments et des substances chimiques , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Phénylènediamines
15.
Microb Cell Fact ; 23(1): 121, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38725068

RÉSUMÉ

BACKGROUND: Mycosporine-like amino acids (MAAs) are a class of strongly UV-absorbing compounds produced by cyanobacteria, algae and corals and are promising candidates for natural sunscreen components. Low MAA yields from natural sources, coupled with difficulties in culturing its native producers, have catalyzed synthetic biology-guided approaches to produce MAAs in tractable microbial hosts like Escherichia coli, Saccharomyces cerevisiae and Corynebacterium glutamicum. However, the MAA titres obtained in these hosts are still low, necessitating a thorough understanding of cellular factors regulating MAA production. RESULTS: To delineate factors that regulate MAA production, we constructed a shinorine (mycosporine-glycine-serine) producing yeast strain by expressing the four MAA biosynthetic enzymes from Nostoc punctiforme in Saccharomyces cerevisiae. We show that shinorine is produced from the pentose phosphate pathway intermediate sedoheptulose 7-phosphate (S7P), and not from the shikimate pathway intermediate 3-dehydroquinate (3DHQ) as previously suggested. Deletions of transaldolase (TAL1) and phosphofructokinase (PFK1/PFK2) genes boosted S7P/shinorine production via independent mechanisms. Unexpectedly, the enhanced S7P/shinorine production in the PFK mutants was not entirely due to increased flux towards the pentose phosphate pathway. We provide multiple lines of evidence in support of a reversed pathway between glycolysis and the non-oxidative pentose phosphate pathway (NOPPP) that boosts S7P/shinorine production in the phosphofructokinase mutant cells. CONCLUSION: Reversing the direction of flux between glycolysis and the NOPPP offers a novel metabolic engineering strategy in Saccharomyces cerevisiae.


Sujet(s)
Acides aminés , Glycolyse , Voie des pentoses phosphates , Saccharomyces cerevisiae , Saccharomyces cerevisiae/métabolisme , Saccharomyces cerevisiae/génétique , Acides aminés/métabolisme , Génie métabolique/méthodes , Nostoc/métabolisme , Nostoc/génétique , Oses phosphates/métabolisme , Glycine/métabolisme , Glycine/analogues et dérivés , Cyclohexylamines
16.
Free Radic Biol Med ; 220: 271-287, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38734267

RÉSUMÉ

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.


Sujet(s)
AlkB Homolog 5, RNA demethylase , Coenzyme A ligases , Ferroptose , Stabilité de l'ARN , Rat Sprague-Dawley , Animaux , Ferroptose/génétique , Rats , Coenzyme A ligases/génétique , Coenzyme A ligases/métabolisme , AlkB Homolog 5, RNA demethylase/métabolisme , AlkB Homolog 5, RNA demethylase/génétique , Cellules PC12 , Cyclohexylamines/pharmacologie , Humains , Déferoxamine/pharmacologie , Stress oxydatif , Lésions encéphaliques/métabolisme , Lésions encéphaliques/génétique , Lésions encéphaliques/anatomopathologie , Lésions encéphaliques/étiologie , Phénylènediamines/pharmacologie , ARN messager/génétique , ARN messager/métabolisme , Mâle , Modèles animaux de maladie humaine , Peroxydation lipidique
17.
Biosci Biotechnol Biochem ; 88(7): 830-838, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38684478

RÉSUMÉ

Mycosporine-like amino acids (MAAs) are the natural UV-absorbing compounds with antioxidant activity found in microalgae and macroalgae. We collected red algae Asparagopsis taxiformis, Meristotheca japonica, and Polysiphonia senticulosa from Nagasaki, where UV radiation is more intense than in Hokkaido, and investigated the effect of UV radiation on MAA content. It was suggested that A. taxiformis and M. japonica contained shinorine and palythine, while UV-absorbing compound in P. senticulosa could not be identified. The amounts of these MAAs were lower compared to those from Hokkaido. Despite an increase in UV radiation in both regions from February to April, MAA contents of red algae from Nagasaki slightly decreased while those from Hokkaido significantly decreased. This difference was suggested the amount of inorganic nitrogen in the ocean. Antioxidant activity of MAAs increased under alkaline conditions. The extract containing MAAs from P. senticulosa showed the highest antioxidant activity among 4 red algae.


Sujet(s)
Acides aminés , Antioxydants , Rhodophyta , Rhodophyta/composition chimique , Acides aminés/analyse , Antioxydants/composition chimique , Antioxydants/pharmacologie , Japon , Rayons ultraviolets , Dérivés du biphényle/antagonistes et inhibiteurs , Concentration en ions d'hydrogène , Cyclohexanols , Cyclohexylamines , Glycine/analogues et dérivés
18.
Exp Cell Res ; 438(1): 114029, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38608805

RÉSUMÉ

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.


Sujet(s)
Asthme , Cadhérines , Modèles animaux de maladie humaine , Ferroptose , Granulocytes , Animaux , Femelle , Souris , Asthme/métabolisme , Asthme/anatomopathologie , Asthme/induit chimiquement , Cadhérines/métabolisme , Cyclohexylamines/pharmacologie , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Ferroptose/effets des médicaments et des substances chimiques , Granulocytes/métabolisme , Granulocytes/anatomopathologie , Souris de lignée BALB C , Ovalbumine , Phénylènediamines/pharmacologie , Quinoxalines , Spiranes
19.
Acta Biomater ; 181: 362-374, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38663684

RÉSUMÉ

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.


Sujet(s)
Ferroptose , Immunothérapie , Nanoparticules , Microenvironnement tumoral , Ferroptose/effets des médicaments et des substances chimiques , Immunothérapie/méthodes , Animaux , Nanoparticules/composition chimique , Souris , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Humains , Lignée cellulaire tumorale , Tumeurs/anatomopathologie , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Acide hyaluronique/composition chimique , Acide hyaluronique/pharmacologie , Cyclohexylamines/pharmacologie , Cyclohexylamines/composition chimique , Imidazoles , Isoindoles
20.
Sci Rep ; 14(1): 7739, 2024 04 02.
Article de Anglais | MEDLINE | ID: mdl-38565869

RÉSUMÉ

Mutations in PINK1 and Parkin cause early-onset Parkinson's Disease (PD). PINK1 is a kinase which functions as a mitochondrial damage sensor and initiates mitochondrial quality control by accumulating on the damaged organelle. There, it phosphorylates ubiquitin, which in turn recruits and activates Parkin, an E3 ubiquitin ligase. Ubiquitylation of mitochondrial proteins leads to the autophagic degradation of the damaged organelle. Pharmacological modulation of PINK1 constitutes an appealing avenue to study its physiological function and develop therapeutics. In this study, we used a thermal shift assay with insect PINK1 to identify small molecules that inhibit ATP hydrolysis and ubiquitin phosphorylation. PRT062607, an SYK inhibitor, is the most potent inhibitor in our screen and inhibits both insect and human PINK1, with an IC50 in the 0.5-3 µM range in HeLa cells and dopaminergic neurons. The crystal structures of insect PINK1 bound to PRT062607 or CYC116 reveal how the compounds interact with the ATP-binding pocket. PRT062607 notably engages with the catalytic aspartate and causes a destabilization of insert-2 at the autophosphorylation dimer interface. While PRT062607 is not selective for PINK1, it provides a scaffold for the development of more selective and potent inhibitors of PINK1 that could be used as chemical probes.


Sujet(s)
Cyclohexylamines , Protein kinases , Pyrimidines , Ubiquitin-protein ligases , Humains , Protein kinases/métabolisme , Cellules HeLa , Ubiquitin-protein ligases/métabolisme , Phosphorylation , Ubiquitine/métabolisme , Adénosine triphosphate/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE