Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 141.121
Filtrer
1.
Clin Exp Rheumatol ; 42(6): 1215-1223, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38966946

RÉSUMÉ

OBJECTIVES: The pathogenesis of fibromyalgia (FM), characterised by chronic widespread pain and fatigue, remains notoriously elusive, hampering attempts to develop disease modifying treatments. Mitochondria are the headquarters of cellular energy metabolism, and their malfunction has been proposed to contribute to both FM and chronic fatigue. Thus, the aim of the current pilot study, was to detect structural changes in mitochondria of peripheral blood mononuclear cells (PBMCs) of FM patients, using transmission electron microscopy (TEM). METHODS: To detect structural mitochondrial alterations in FM, we analysed PBMCs from seven patients and seven healthy controls, using TEM. Patients were recruited from a specialised Fibromyalgia Clinic at a tertiary medical centre. After providing informed consent, participants completed questionnaires including the widespread pain index (WPI), symptoms severity score (SSS), fibromyalgia impact questionnaire (FIQ), beck depression inventory (BDI), and visual analogue scale (VAS), to verify a diagnosis of FM according to ACR criteria. Subsequently, blood samples were drawn and PBMCs were collected for EM analysis. RESULTS: TEM analysis of PBMCs showed several distinct mitochondrial cristae patterns, including total loss of cristae in FM patients. The number of mitochondria with intact cristae morphology was reduced in FM patients and the percentage of mitochondria that completely lacked cristae was increased. These results correlated with the WPI severity. Moreover, in the FM patient samples we observed a high percentage of cells containing electron dense aggregates, which are possibly ribosome aggregates. Cristae loss and possible ribosome aggregation were intercorrelated, and thus may represent reactions to a shared cellular stress condition. The changes in mitochondrial morphology suggest that mitochondrial dysfunction, resulting in inefficient oxidative phosphorylation and ATP production, metabolic and redox disorders, and increased reactive oxygen species (ROS) levels, may play a pathogenetic role in FM. CONCLUSIONS: We describe novel morphological changes in mitochondria of FM patients, including loss of mitochondrial cristae. While these observations cannot determine whether the changes are pathogenetic or represent an epiphenomenon, they highlight the possibility that mitochondrial malfunction may play a causative role in the cascade of events leading to chronic pain and fatigue in FM. Moreover, the results offer the possibility of utilising changes in mitochondrial morphology as an objective biomarker in FM. Further understanding the connection between FM and dysfunction of mitochondria physiology, may assist in developing both novel diagnostic tools as well as specific treatments for FM, such as approaches to improve/strengthen mitochondria function.


Sujet(s)
Fibromyalgie , Mitochondries , Humains , Fibromyalgie/anatomopathologie , Fibromyalgie/physiopathologie , Projets pilotes , Mitochondries/ultrastructure , Mitochondries/anatomopathologie , Femelle , Adulte d'âge moyen , Adulte , Études cas-témoins , Mâle , Microscopie électronique à transmission , Agranulocytes/ultrastructure , Agranulocytes/anatomopathologie , Indice de gravité de la maladie , Mesure de la douleur
2.
PeerJ ; 12: e17637, 2024.
Article de Anglais | MEDLINE | ID: mdl-38966207

RÉSUMÉ

Background: Prostate cancer (PCa) is one of the causes of death in men worldwide. Although treatment strategies have been developed, the recurrence of the disease and consequential side effects remain an essential concern. Diospyros rhodocalyx Kurz, a traditional Thai medicine, exhibits diverse therapeutic properties, including anti-cancer activity. However, its anti-cancer activity against prostate cancer has not been thoroughly explored. This study aims to evaluate the anti-cancer activity and underlying mechanisms of the ethyl acetate extract of D. rhodocalyx Kurz (EADR) related to apoptosis induction in the LNCaP human prostate cancer cell line. Methods: Ethyl acetate was employed to extract the dried bark of D. rhodocalyx Kurz. The cytotoxicity of EADR on both LNCaP and WPMY-1 cells (normal human prostatic myofibroblast cell line) was evaluated using MTS assay. The effect of EADR on the cell cycle, apoptosis induction, and alteration in mitochondrial membrane potential (MMP) was assessed by the staining with propidium iodide (PI), Annexin V-FITC/PI, and JC-1 dye, respectively. Subsequent analysis was conducted using flow cytometry. The expression of cleaved caspase-3, BAX, and Bcl-2 was examined by Western blotting. The phytochemical profiling of the EADR was performed using gas chromatography-mass spectrometry (GC-MS). Results: EADR exhibited a dose-dependent manner cytotoxic effect on LNCaP cells, with IC50 values of 15.43 and 12.35 µg/mL after 24 and 48 h, respectively. Although it also exhibited a cytotoxic effect on WPMY-1 cells, the effect was comparatively lower, with the IC50 values of 34.61 and 19.93 µg/mL after 24 and 48 h of exposure, respectively. Cell cycle analysis demonstrated that EADR did not induce cell cycle arrest in either LNCaP or WPMY-1 cells. However, it significantly increased the sub-G1 population in LNCaP cells, indicating a potential induction of apoptosis. The Annexin V-FITC/PI staining indicated that EADR significantly induced apoptosis in LNCaP cells. Subsequent investigation into the underlying mechanism of EADR-induced apoptosis revealed a reduction in MMP as evidenced by JC-1 staining. Moreover, Western blotting demonstrated that EADR treatment resulted in the upregulation of BAX, downregulation of BCL-2, and elevation of caspase-3 cleavage in LNCaP cells. Notably, the epilupeol was a prominent compound in EADR as identified by GC-MS. Conclusion: The EADR exhibits anti-cancer activity against the LNCaP human prostate cancer cell line by inducing cytotoxicity and apoptosis. Our findings suggest that EADR promotes apoptosis by upregulating pro-apoptotic BAX, whereas downregulation of anti-apoptotic Bcl-2 results in the reduction of MMP and the activation of caspase-3. Of particular interest is the presence of epilupeol, a major compound identified in EADR, which may hold promise as a candidate for the development of therapeutic agents for prostate cancer.


Sujet(s)
Apoptose , Caspase-3 , Diospyros , Extraits de plantes , Tumeurs de la prostate , Protéines proto-oncogènes c-bcl-2 , Protéine Bax , Humains , Mâle , Apoptose/effets des médicaments et des substances chimiques , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/métabolisme , Lignée cellulaire tumorale , Extraits de plantes/pharmacologie , Protéines proto-oncogènes c-bcl-2/métabolisme , Protéine Bax/métabolisme , Caspase-3/métabolisme , Diospyros/composition chimique , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Antinéoplasiques d'origine végétale/pharmacologie
3.
J Nanobiotechnology ; 22(1): 385, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951822

RÉSUMÉ

BACKGROUND: Numerous studies have confirmed the involvement of extracellular vesicles (EVs) in various physiological processes, including cellular death and tissue damage. Recently, we reported that EVs derived from ischemia-reperfusion heart exacerbate cardiac injury. However, the role of EVs from healthy heart tissue (heart-derived EVs, or cEVs) on myocardial ischemia-reperfusion (MI/R) injury remains unclear. RESULTS: Here, we demonstrated that intramyocardial administration of cEVs significantly enhanced cardiac function and reduced cardiac damage in murine MI/R injury models. cEVs treatment effectively inhibited ferroptosis and maintained mitochondrial homeostasis in cardiomyocytes subjected to ischemia-reperfusion injury. Further results revealed that cEVs can transfer ATP5a1 into cardiomyocytes, thereby suppressing mitochondrial ROS production, alleviating mitochondrial damage, and inhibiting cardiomyocyte ferroptosis. Knockdown of ATP5a1 abolished the protective effects of cEVs. Furthermore, we found that the majority of cEVs are derived from cardiomyocytes, and ATP5a1 in cEVs primarily originates from cardiomyocytes of the healthy murine heart. Moreover, we demonstrated that adipose-derived stem cells (ADSC)-derived EVs with ATP5a1 overexpression showed much better efficacy on the therapy of MI/R injury compared to control ADSC-derived EVs. CONCLUSIONS: These findings emphasized the protective role of cEVs in cardiac injury and highlighted the therapeutic potential of targeting ATP5a1 as an important approach for managing myocardial damage induced by MI/R injury.


Sujet(s)
Vésicules extracellulaires , Souris de lignée C57BL , Mitochondrial Proton-Translocating ATPases , Lésion de reperfusion myocardique , Myocytes cardiaques , Animaux , Vésicules extracellulaires/métabolisme , Souris , Lésion de reperfusion myocardique/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Mâle , Mitochondrial Proton-Translocating ATPases/métabolisme , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Myocarde/métabolisme , Myocarde/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine
4.
Cell Metab ; 36(7): 1433-1435, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38959859

RÉSUMÉ

Small peptides have previously been reported to be encoded in mitochondrial rRNA and translated by cytosolic ribosomes. In this issue of Cell Metabolism, Hu et al. use mass spectrometry to identify a cytosolically translated protein, encoded instead in mitochondrial mRNA, that is surprisingly targeted back into the mitochondrial matrix.


Sujet(s)
Mitochondries , ARN messager , ARN messager/métabolisme , ARN messager/génétique , Mitochondries/métabolisme , Mitochondries/génétique , ARN mitochondrial/métabolisme , ARN mitochondrial/génétique , Biosynthèse des protéines , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Humains , Cytosol/métabolisme , Spectrométrie de masse
5.
Cell Metab ; 36(7): 1436-1438, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38959860

RÉSUMÉ

The factors determining levels of pathogenic mitochondrial DNA in cells and tissues are critical to disease pathology but remain poorly understood and contentious. In Nature, Kotrys et al. published a single-cell-based analysis casting fresh light on this thorny problem and introduced a powerful new investigative tool.


Sujet(s)
ADN mitochondrial , ADN mitochondrial/génétique , Humains , Mitochondries/métabolisme , Mitochondries/génétique , Analyse sur cellule unique/méthodes
6.
Sci Rep ; 14(1): 15406, 2024 07 04.
Article de Anglais | MEDLINE | ID: mdl-38965397

RÉSUMÉ

Patients with multiple myeloma (MM) experience relapse and drug resistance; therefore, novel treatments are essential. Clotrimazole (CTZ) is a wide-spectrum antifungal drug with antitumor activity. However, CTZ's effects on MM are unclear. We investigated CTZ's effect on MM cell proliferation and apoptosis induction mechanisms. CTZ's effects on MM.1S, NCI- H929, KMS-11, and U266 cell growth were investigated using Cell Counting Kit-8 (CCK-8) assay. The apoptotic cell percentage was quantified with annexin V-fluorescein isothiocyanate/7-amino actinomycin D staining. Mitochondrial membrane potential (MMP) and cell cycle progression were evaluated. Reactive oxygen species (ROS) levels were measured via fluorescence microscopy. Expression of apoptosis-related and nuclear factor (NF)-κB signaling proteins was analyzed using western blotting. The CCK-8 assay indicated that CTZ inhibited cell proliferation based on both dose and exposure time. Flow cytometry revealed that CTZ decreased apoptosis and MMP and induced G0/G1 arrest. Immunofluorescence demonstrated that CTZ dose-dependently elevated in both total and mitochondrial ROS production. Western blotting showed that CTZ enhanced Bax and cleaved poly ADP-ribose polymerase and caspase-3 while decreasing Bcl-2, p-p65, and p-IκBα. Therefore, CTZ inhibits MM cell proliferation by promoting ROS-mediated mitochondrial apoptosis, inducing G0/G1 arrest, inhibiting the NF-κB pathway, and has the potential for treating MM.


Sujet(s)
Apoptose , Prolifération cellulaire , Clotrimazole , Potentiel de membrane mitochondriale , Mitochondries , Myélome multiple , Espèces réactives de l'oxygène , Humains , Myélome multiple/anatomopathologie , Myélome multiple/traitement médicamenteux , Myélome multiple/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Espèces réactives de l'oxygène/métabolisme , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Clotrimazole/pharmacologie , Phase G0/effets des médicaments et des substances chimiques , Points de contrôle de la phase G1 du cycle cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Antinéoplasiques/pharmacologie , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques
7.
Cell Biol Toxicol ; 40(1): 51, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38958792

RÉSUMÉ

The implementation of Zinc oxide nanoparticles (ZnO NPs) raises concerns regarding their potential toxic effects on human health. Although more and more researches have confirmed the toxic effects of ZnO NPs, limited attention has been given to their impact on the early embryonic nervous system. This study aimed to explore the impact of exposure to ZnO NPs on early neurogenesis and explore its underlying mechanisms. We conducted experiments here to confirm the hypothesis that exposure to ZnO NPs causes neural tube defects in early embryonic development. We first used mouse and chicken embryos to confirm that ZnO NPs and the Zn2+ they release are able to penetrate the placental barrier, influence fetal growth and result in incomplete neural tube closure. Using SH-SY5Y cells, we determined that ZnO NPs-induced incomplete neural tube closure was caused by activation of various cell death modes, including ferroptosis, apoptosis and autophagy. Moreover, dissolved Zn2+ played a role in triggering widespread cell death. ZnO NPs were accumulated within mitochondria after entering cells, damaging mitochondrial function and resulting in the over production of reactive oxygen species, ultimately inducing cellular oxidative stress. The N-acetylcysteine (NAC) exhibits significant efficacy in mitigating cellular oxidative stress, thereby alleviating the cytotoxicity and neurotoxicity brought about by ZnO NPs. These findings indicated that the exposure of ZnO NPs in early embryonic development can induce cell death through oxidative stress, resulting in a reduced number of cells involved in early neural tube closure and ultimately resulting in incomplete neural tube closure during embryo development. The findings of this study could raise public awareness regarding the potential risks associated with the exposure and use of ZnO NPs in early pregnancy.


Sujet(s)
Développement embryonnaire , Anomalies du tube neural , Tube neural , Stress oxydatif , Espèces réactives de l'oxygène , Oxyde de zinc , Oxyde de zinc/toxicité , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Embryon de poulet , Développement embryonnaire/effets des médicaments et des substances chimiques , Souris , Tube neural/effets des médicaments et des substances chimiques , Tube neural/embryologie , Tube neural/métabolisme , Humains , Anomalies du tube neural/induit chimiquement , Anomalies du tube neural/métabolisme , Anomalies du tube neural/embryologie , Anomalies du tube neural/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Mort cellulaire/effets des médicaments et des substances chimiques , Femelle , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Nanoparticules métalliques/toxicité , Autophagie/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Nanoparticules/toxicité
8.
Life Sci Alliance ; 7(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38955468

RÉSUMÉ

In addition to mitochondrial DNA, mitochondrial double-stranded RNA (mtdsRNA) is exported from mitochondria. However, specific channels for RNA transport have not been demonstrated. Here, we begin to characterize channel candidates for mtdsRNA export from the mitochondrial matrix to the cytosol. Down-regulation of SUV3 resulted in the accumulation of mtdsRNAs in the matrix, whereas down-regulation of PNPase resulted in the export of mtdsRNAs to the cytosol. Targeting experiments show that PNPase functions in both the intermembrane space and matrix. Strand-specific sequencing of the double-stranded RNA confirms the mitochondrial origin. Inhibiting or down-regulating outer membrane proteins VDAC1/2 and BAK/BAX or inner membrane proteins PHB1/2 strongly attenuated the export of mtdsRNAs to the cytosol. The cytosolic mtdsRNAs subsequently localized to large granules containing the stress protein TIA-1 and activated the type 1 interferon stress response pathway. Abundant mtdsRNAs were detected in a subset of non-small-cell lung cancer cell lines that were glycolytic, indicating relevance in cancer biology. Thus, we propose that mtdsRNA is a new damage-associated molecular pattern that is exported from mitochondria in a regulated manner.


Sujet(s)
Cytosol , Mitochondries , Prohibitines , ARN double brin , ARN mitochondrial , Humains , Cytosol/métabolisme , Mitochondries/métabolisme , ARN double brin/métabolisme , ARN mitochondrial/métabolisme , ARN mitochondrial/génétique , Lignée cellulaire tumorale , Protéines de répression/métabolisme , Protéines de répression/génétique , Transport des ARN , Exoribonucleases/métabolisme , Exoribonucleases/génétique , Canal anionique-1 voltage-dépendant/métabolisme , Canal anionique-1 voltage-dépendant/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Protéines mitochondriales
9.
Nat Commun ; 15(1): 5571, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956041

RÉSUMÉ

Statin drugs lower blood cholesterol levels for cardiovascular disease prevention. Women are more likely than men to experience adverse statin effects, particularly new-onset diabetes (NOD) and muscle weakness. Here we find that impaired glucose homeostasis and muscle weakness in statin-treated female mice are associated with reduced levels of the omega-3 fatty acid, docosahexaenoic acid (DHA), impaired redox tone, and reduced mitochondrial respiration. Statin adverse effects are prevented in females by administering fish oil as a source of DHA, by reducing dosage of the X chromosome or the Kdm5c gene, which escapes X chromosome inactivation and is normally expressed at higher levels in females than males. As seen in female mice, we find that women experience more severe reductions than men in DHA levels after statin administration, and that DHA levels are inversely correlated with glucose levels. Furthermore, induced pluripotent stem cells from women who developed NOD exhibit impaired mitochondrial function when treated with statin, whereas cells from men do not. These studies identify X chromosome dosage as a genetic risk factor for statin adverse effects and suggest DHA supplementation as a preventive co-therapy.


Sujet(s)
Acide docosahexaénoïque , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase , Mitochondries , Chromosome X , Animaux , Femelle , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/effets indésirables , Mâle , Souris , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Humains , Chromosome X/génétique , Acide docosahexaénoïque/pharmacologie , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Dosage génique , Souris de lignée C57BL , Glycémie/métabolisme , Glycémie/effets des médicaments et des substances chimiques , Glucose/métabolisme , Diabète/génétique , Diabète/induit chimiquement , Diabète/traitement médicamenteux , Diabète/métabolisme
10.
Cell Death Dis ; 15(7): 473, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956064

RÉSUMÉ

Damage to renal tubular epithelial cells (RTECs) signaled the onset and progression of sepsis-associated acute kidney injury (SA-AKI). Recent research on mitochondria has revealed that mitophagy plays a crucial physiological role in alleviating injury to RTECs and it is suppressed progressively by the inflammation response in SA-AKI. However, the mechanism by which inflammation influences mitophagy remains poorly understood. We examined how macrophage migration inhibitory factor (MIF), a pro-inflammatory protein, influences the PINK1-Parkin pathway of mitophagy by studying protein-protein interactions when MIF was inhibited or overexpressed. Surprisingly, elevated levels of MIF were found to directly bind to PINK1, disrupting its interaction with Parkin. This interference hindered the recruitment of Parkin to mitochondria and impeded the initiation of mitophagy. Furthermore, this outcome led to significant apoptosis of RTECs, which could, however, be reversed by an MIF inhibitor ISO-1 and/or a new mitophagy activator T0467. These findings highlight the detrimental impact of MIF on renal damage through its disruption of the interaction between PINK1 and Parkin, and the therapeutic potential of ISO-1 and T0467 in mitigating SA-AKI. This study offers a fresh perspective on treating SA-AKI by targeting MIF and mitophagy.


Sujet(s)
Atteinte rénale aigüe , Facteurs inhibiteurs de la migration des macrophages , Mitophagie , Protein kinases , Sepsie , Ubiquitin-protein ligases , Facteurs inhibiteurs de la migration des macrophages/métabolisme , Facteurs inhibiteurs de la migration des macrophages/génétique , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/anatomopathologie , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Protein kinases/métabolisme , Sepsie/complications , Sepsie/métabolisme , Animaux , Humains , Mitochondries/métabolisme , Tubules rénaux/métabolisme , Tubules rénaux/anatomopathologie , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Apoptose , Liaison aux protéines , Mâle , Intramolecular oxidoreductases/métabolisme
11.
Sci Rep ; 14(1): 15246, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956068

RÉSUMÉ

This study aimed to explore the effects of peroxisome proliferator-activated receptor α (PPAR-α), a known inhibitor of ferroptosis, in Myocardial ischemia/reperfusion injury (MIRI) and its related mechanisms. In vivo and in vitro MIRI models were established. Our results showed that activation of PPAR-α decreased the size of the myocardial infarct, maintained cardiac function, and decreased the serum contents of creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH), and Fe2+ in ischemia/reperfusion (I/R)-treated mice. Additionally, the results of H&E staining, DHE staining, TUNEL staining, and transmission electron microscopy demonstrated that activation of PPAR-α inhibited MIRI-induced heart tissue and mitochondrial damage. It was also found that activation of PPAR-α attenuated MIRI-induced ferroptosis as shown by a reduction in malondialdehyde, total iron, and reactive oxygen species (ROS). In vitro experiments showed that intracellular contents of malondialdehyde, total iron, LDH, reactive oxygen species (ROS), lipid ROS, oxidized glutathione disulphide (GSSG), and Fe2+ were reduced by the activation of PPAR-α in H9c2 cells treated with anoxia/reoxygenation (A/R), while the cell viability and GSH were increased after PPAR-α activation. Additionally, changes in protein levels of the ferroptosis marker further confirmed the beneficial effects of PPAR-α activation on MIRI-induced ferroptosis. Moreover, the results of immunofluorescence and dual-luciferase reporter assay revealed that PPAR-α achieved its activity via binding to the 14-3-3η promoter, promoting its expression level. Moreover, the cardioprotective effects of PPAR-α could be canceled by pAd/14-3-3η-shRNA or Compound C11 (14-3-3η inhibitor). In conclusion, our results indicated that ferroptosis plays a key role in aggravating MIRI, and PPAR-α/14-3-3η pathway-mediated ferroptosis and mitochondrial injury might be an effective therapeutic target against MIRI.


Sujet(s)
Protéines 14-3-3 , Ferroptose , Lésion de reperfusion myocardique , Récepteur PPAR alpha , Ferroptose/effets des médicaments et des substances chimiques , Animaux , Récepteur PPAR alpha/métabolisme , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/anatomopathologie , Protéines 14-3-3/métabolisme , Souris , Mâle , Espèces réactives de l'oxygène/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Lignée cellulaire , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Souris de lignée C57BL , Rats , Modèles animaux de maladie humaine
12.
Sci Rep ; 14(1): 15175, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956251

RÉSUMÉ

In the current study, we aimed to investigate whether disulfiram (DSF) exerts a neuroprotective role in cerebral ischemiareperfusion (CI-RI) injury by modulating ferredoxin 1 (FDX1) to regulate copper ion (Cu) levels and inhibiting inflammatory responses. To simulate CI-RI, a transient middle cerebral artery occlusion (tMCAO) model in C57/BL6 mice was employed. Mice were administered with or without DSF before and after tMCAO. Changes in infarct volume after tMCAO were observed using TTC staining. Nissl staining and hematoxylin-eosin (he) staining were used to observe the morphological changes of nerve cells at the microscopic level. The inhibitory effect of DSF on initial inflammation was verified by TUNEL assay, apoptosis-related protein detection and iron concentration detection. FDX1 is the main regulatory protein of copper death, and the occurrence of copper death will lead to the increase of HSP70 stress and inflammatory response. Cuproptosis-related proteins and downstream inflammatory factors were detected by western blotting, immunofluorescence staining, and immunohistochemistry. The content of copper ions was detected using a specific kit, while electron microscopy was employed to examine mitochondrial changes. We found that DSF reduced the cerebral infarction volume, regulated the expression of cuproptosis-related proteins, and modulated copper content through down regulation of FDX1 expression. Moreover, DSF inhibited the HSP70/TLR-4/NLRP3 signaling pathway. Collectively, DSF could regulate Cu homeostasis by inhibiting FDX1, acting on the HSP70/TLR4/NLRP3 pathway to alleviate CI/RI. Accordingly, DSF could mitigate inflammatory responses and safeguard mitochondrial integrity, yielding novel therapeutic targets and mechanisms for the clinical management of ischemia-reperfusion injury.


Sujet(s)
Cuivre , Disulfirame , Homéostasie , Inflammation , Souris de lignée C57BL , Lésion d'ischémie-reperfusion , Animaux , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/anatomopathologie , Disulfirame/pharmacologie , Souris , Cuivre/métabolisme , Homéostasie/effets des médicaments et des substances chimiques , Mâle , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Régulation négative/effets des médicaments et des substances chimiques , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Modèles animaux de maladie humaine , Ferrosulfoprotéines/métabolisme , Encéphalopathie ischémique/métabolisme , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Récepteur de type Toll-4/métabolisme
13.
Sci Rep ; 14(1): 15215, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956409

RÉSUMÉ

Increasing evidence has shown that many environmental and toxic factors can cause testicular damage, leading to testicular ferroptosis and subsequent male reproductive disorders. Melatonin is a major hormone and plays an vital role in regulating male reproduction. However, there is a lack of research on whether Mel can alleviate testicular cell ferroptosis and its specific mechanism. In this study, the results indicated that Mel could enhance the viability of swine testis cells undergoing ferroptosis, reduce LDH enzyme release, increase mitochondrial membrane potential, and affect the expression of ferroptosis biomarkers. Furthermore, we found that melatonin depended on melatonin receptor 1B to exert these functions. Detection of MMP and ferroptosis biomarker protein expression confirmed that MT2 acted through the downstream Akt signaling pathway. Moreover, inhibition of the Akt signaling pathway can eliminate the protective effect of melatonin on ferroptosis, inhibit AMPK phosphorylation, reduce the expression of mitochondrial gated channel (VDAC2/3), and affect mitochondrial DNA transcription and ATP content. These results suggest that melatonin exerts a beneficial effect on mitochondrial function to mitigate ferroptosis through the MT2/Akt signaling pathway in ST cells.


Sujet(s)
Ferroptose , Mélatonine , Mitochondries , Protéines proto-oncogènes c-akt , Récepteur de la mélatonine de type MT2 , Transduction du signal , Testicule , Animaux , Mélatonine/pharmacologie , Mâle , Ferroptose/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Suidae , Testicule/métabolisme , Testicule/effets des médicaments et des substances chimiques , Récepteur de la mélatonine de type MT2/métabolisme , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques
14.
Stem Cell Res Ther ; 15(1): 189, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956646

RÉSUMÉ

BACKGROUND: Recent studies have proved the role of autophagy in mesenchymal stem cell (MSCs) function and regenerative properties. How and by which mechanism autophagy modulation can affect the juxtacrine interaction of MSCs should be addressed. Here, the role of autophagy was investigated in the formation of tunneling nanotubes (TNTs) and homotypic mitochondrial donation. METHODS: MSCs were incubated with 15 µM Metformin (Met) and/or 3 µM 3-methyladenine (3-MA) for 48 h. The formation of TNTs was assessed using bright-field and SEM images. The mitochondria density and ΔΨ values were monitored using flow cytometry analysis. Using RT-PCR and protein array, the close interaction and shared mediators between autophagy, apoptosis, and Wnt signaling pathways were also monitored. The total fatty acid profile was assessed using gas chromatography. RESULT: Data indicated the increase of TNT length and number, along with other cell projections after the induction of autophagy while these features were blunted in 3-MA-treated MSCs (p < 0.05). Western blotting revealed the significant reduction of Rab8 and p-FAK in 3-MA-treated MSCs (p < 0.05), indicating the inhibition of TNT assembly and vesicle transport. Likewise, the stimulation of autophagy increased autophagic flux and mitochondrial membrane integrity compared to 3-MA-treated MSCs. Despite these findings, protein levels of mitochondrial membrane Miro1 and 2 were unchanged after autophagy inhibition/stimulation (p > 0.05). We found that the inhibition/stimulation of autophagy can affect the protein, and transcription levels of several mediators related to Wnt and apoptosis signaling pathways involved in different cell bioactivities. Data confirmed the profound increase of mono and polyunsaturated/saturated fatty acid ratio in MSCs exposed to autophagy stimulator. CONCLUSIONS: In summary, autophagy modulation could affect TNT formation which is required for homotypic mitochondrial donation. Thus, the modulation of autophagy creates a promising perspective to increase the efficiency of cell-based therapies.


Sujet(s)
Autophagie , Cellules souches mésenchymateuses , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Mitochondries/métabolisme , Adénine/pharmacologie , Adénine/analogues et dérivés , Humains , Nanotubes/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Animaux , Metformine/pharmacologie , Cellules cultivées , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Structures de la membrane cellulaire
15.
Nat Commun ; 15(1): 5664, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38969660

RÉSUMÉ

Mitochondrial gene expression relies on mitoribosomes to translate mitochondrial mRNAs. The biogenesis of mitoribosomes is an intricate process involving multiple assembly factors. Among these factors, GTP-binding proteins (GTPBPs) play important roles. In bacterial systems, numerous GTPBPs are required for ribosome subunit maturation, with EngB being a GTPBP involved in the ribosomal large subunit assembly. In this study, we focus on exploring the function of GTPBP8, the human homolog of EngB. We find that ablation of GTPBP8 leads to the inhibition of mitochondrial translation, resulting in significant impairment of oxidative phosphorylation. Structural analysis of mitoribosomes from GTPBP8 knock-out cells shows the accumulation of mitoribosomal large subunit assembly intermediates that are incapable of forming functional monosomes. Furthermore, fPAR-CLIP analysis reveals that GTPBP8 is an RNA-binding protein that interacts specifically with the mitochondrial ribosome large subunit 16 S rRNA. Our study highlights the role of GTPBP8 as a component of the mitochondrial gene expression machinery involved in mitochondrial large subunit maturation.


Sujet(s)
Protéines G , Mitochondries , Ribosomes mitochondriaux , Phosphorylation oxydative , Humains , Ribosomes mitochondriaux/métabolisme , Mitochondries/métabolisme , Protéines G/métabolisme , Protéines G/génétique , ARN ribosomique 16S/génétique , ARN ribosomique 16S/métabolisme , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Cellules HEK293 , Biosynthèse des protéines , ARN messager/métabolisme , ARN messager/génétique , Cellules HeLa
16.
Int J Nanomedicine ; 19: 6603-6618, 2024.
Article de Anglais | MEDLINE | ID: mdl-38979533

RÉSUMÉ

Objective: Ovarian cancer cells are prone to acquire tolerance to chemotherapeutic agents, which seriously affects clinical outcomes. The development of novel strategies to enhance the targeting of chemotherapeutic agents to overcome drug resistance and minimize side effects is significant for improving the clinical outcomes of ovarian cancer patients. Methods: We employed folic acid (FA)-modified ZIF-90 nanomaterials (FA-ZIF-90) to deliver the chemotherapeutic drug, cisplatin (DDP), via dual targeting to improve its targeting to circumvent cisplatin resistance in ovarian cancer cells, especially by targeting mitochondria. FA-ZIF-90/DDP could rapidly release DDP in response to dual stimulation of acidity and ATP in tumor cells. Results: FA-ZIF-90/DDP showed good blood compatibility. It was efficiently taken up by human ovarian cancer cisplatin-resistant cells A2780/DDP and aggregated in the mitochondrial region. FA-ZIF-90/DDP significantly inhibited the mitochondrial activity and metastatic ability of A2780/DDP cells. In addition, it effectively induced apoptosis in A2780/DDP cells and overcame cisplatin resistance. In vivo experiments showed that FA-ZIF-90/DDP increased the accumulation of DDP in tumor tissues and significantly inhibited tumor growth. Conclusion: FA-modified ZIF-90 nanocarriers can improve the tumor targeting and anti-tumor effects of chemotherapeutic drugs, reduce toxic side effects, and are expected to be a novel therapeutic strategy to reverse drug resistance in ovarian cancer.


Sujet(s)
Antinéoplasiques , Apoptose , Cisplatine , Résistance aux médicaments antinéoplasiques , Acide folique , Imidazoles , Tumeurs de l'ovaire , Zéolites , Femelle , Cisplatine/pharmacologie , Cisplatine/composition chimique , Cisplatine/pharmacocinétique , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/anatomopathologie , Humains , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Animaux , Zéolites/composition chimique , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/administration et posologie , Acide folique/composition chimique , Acide folique/pharmacologie , Imidazoles/composition chimique , Imidazoles/pharmacologie , Imidazoles/administration et posologie , Apoptose/effets des médicaments et des substances chimiques , Systèmes de délivrance de médicaments/méthodes , Mitochondries/effets des médicaments et des substances chimiques , Souris , Souris de lignée BALB C , Souris nude , Vecteurs de médicaments/composition chimique , Réseaux organométalliques/composition chimique , Réseaux organométalliques/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
17.
Biochemistry (Mosc) ; 89(6): 1061-1078, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38981701

RÉSUMÉ

Voltage-dependent anion channels (VDAC1-3) of the outer mitochondrial membrane are a family of pore-forming ß-barrel proteins that carry out controlled "filtration" of small molecules and ions between the cytoplasm and mitochondria. Due to the conformational transitions between the closed and open states and interaction with cytoplasmic and mitochondrial proteins, VDACs not only regulate the mitochondrial membrane permeability for major metabolites and ions, but also participate in the control of essential intracellular processes and pathological conditions. This review discusses novel data on the molecular structure, regulatory mechanisms, and pathophysiological role of VDAC proteins, as well as future directions in this area of research.


Sujet(s)
Membranes mitochondriales , Canaux anioniques voltage-dépendants , Humains , Canaux anioniques voltage-dépendants/métabolisme , Membranes mitochondriales/métabolisme , Animaux , Mitochondries/métabolisme
18.
Biochemistry (Mosc) ; 89(6): 1031-1044, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38981699

RÉSUMÉ

Alzheimer's disease (AD) is a severe neurodegenerative condition affecting millions worldwide. Prevalence of AD correlates with increased life expectancy and aging population in the developed countries. Considering that AD is a multifactorial disease involving various pathological processes such as synaptic dysfunction, neuroinflammation, oxidative stress, and improper protein folding, a comprehensive approach targeting multiple pathways may prove effective in slowing the disease progression. Cellular therapy and its further development in the form of cell vesicle and particularly mitochondrial transplantation represent promising approaches for treating neurodegeneration. The use of synaptosomes, due to uniqueness of their contents, could mark a new stage in the development of comprehensive therapies for neurodegenerative diseases, particularly AD. Synaptosomes contain unique memory mitochondria, which differ not only in size but also in functionality compared to the mitochondria in the neuronal soma. These synaptosomal mitochondria actively participate in cellular communication and signal transmission within synapses. Synaptosomes also contain other elements such as their own protein synthesis machinery, synaptic vesicles with neurotransmitters, synaptic adhesion molecules, and microRNAs - all crucial for synaptic transmission and, consequently, cognitive processes. Complex molecular ensemble ensures maintenance of the synaptic autonomy of mitochondria. Additionally, synaptosomes, with their affinity for neurons, can serve as an optimal platform for targeted drug delivery to nerve cells. This review discusses unique composition of synaptosomes, their capabilities and advantages, as well as limitations of their suggested use as therapeutic agents for treating neurodegenerative pathologies, particularly AD.


Sujet(s)
Maladie d'Alzheimer , Synaptosomes , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/thérapie , Maladie d'Alzheimer/anatomopathologie , Humains , Synaptosomes/métabolisme , Animaux , Mitochondries/métabolisme , Transmission synaptique , Neurones/métabolisme , Synapses/métabolisme
19.
Biol Lett ; 20(7): 20240147, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38982851

RÉSUMÉ

The nucleus interacts with the other organelles to perform essential functions of the eukaryotic cell. Mitochondria have their own genome and communicate back to the nucleus in what is known as mitochondrial retrograde response. Information is transferred to the nucleus in many ways, leading to wide-ranging changes in nuclear gene expression and culminating with changes in metabolic, regulatory or stress-related pathways. RNAs are emerging molecules involved in this signalling. RNAs encode precise information and are involved in highly target-specific signalling, through a wide range of processes known as RNA interference. RNA-mediated mitochondrial retrograde response requires these molecules to exit the mitochondrion, a process that is still mostly unknown. We suggest that the proteins/complexes translocases of the inner membrane, polynucleotide phosphorylase, mitochondrial permeability transition pore, and the subunits of oxidative phosphorylation complexes may be responsible for RNA export.


Sujet(s)
Noyau de la cellule , Mitochondries , Mitochondries/métabolisme , Noyau de la cellule/métabolisme , ARN/métabolisme , ARN/génétique , Animaux , Transport des ARN , Cellules eucaryotes/métabolisme , Eucaryotes/métabolisme , Eucaryotes/génétique , Eucaryotes/physiologie , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...