Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.434
Filter
1.
Circ Res ; 134(10): 1292-1305, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38618716

ABSTRACT

BACKGROUND: During myocardial ischemia/reperfusion (I/R) injury, high levels of matrix Ca2+ and reactive oxygen species (ROS) induce the opening of the mitochondrial permeability transition pore (mPTP), which causes mitochondrial dysfunction and ultimately necrotic death. However, the mechanisms of how these triggers individually or cooperatively open the pore have yet to be determined. METHODS: Here, we use a combination of isolated mitochondrial assays and in vivo I/R surgery in mice. We challenged isolated liver and heart mitochondria with Ca2+, ROS, and Fe2+ to induce mitochondrial swelling. Using inhibitors of the mPTP (cyclosporine A or ADP) lipid peroxidation (ferrostatin-1, MitoQ), we determined how the triggers elicit mitochondrial damage. Additionally, we used the combination of inhibitors during I/R injury in mice to determine if dual inhibition of these pathways is additivity protective. RESULTS: In the absence of Ca2+, we determined that ROS fails to trigger mPTP opening. Instead, high levels of ROS induce mitochondrial dysfunction and rupture independently of the mPTP through lipid peroxidation. As expected, Ca2+ in the absence of ROS induces mPTP-dependent mitochondrial swelling. Subtoxic levels of ROS and Ca2+ synergize to induce mPTP opening. Furthermore, this synergistic form of Ca2+- and ROS-induced mPTP opening persists in the absence of CypD (cyclophilin D), suggesting the existence of a CypD-independent mechanism for ROS sensitization of the mPTP. These ex vivo findings suggest that mitochondrial dysfunction may be achieved by multiple means during I/R injury. We determined that dual inhibition of the mPTP and lipid peroxidation is significantly more protective against I/R injury than individually targeting either pathway alone. CONCLUSIONS: In the present study, we have investigated the relationship between Ca2+ and ROS, and how they individually or synergistically induce mitochondrial swelling. Our findings suggest that Ca2+ mediates mitochondrial damage through the opening of the mPTP, although ROS mediates its damaging effects through lipid peroxidation. However, subtoxic levels both Ca2+ and ROS can induce mPTP-mediated mitochondrial damage. Targeting both of these triggers to preserve mitochondria viability unveils a highly effective therapeutic approach for mitigating I/R injury.


Subject(s)
Lipid Peroxidation , Mice, Inbred C57BL , Mitochondria, Heart , Mitochondria, Liver , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury , Reactive Oxygen Species , Animals , Lipid Peroxidation/drug effects , Mitochondrial Permeability Transition Pore/metabolism , Reactive Oxygen Species/metabolism , Mice , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/pathology , Male , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Mitochondria, Liver/drug effects , Calcium/metabolism , Mitochondrial Swelling/drug effects
2.
Cells ; 10(5)2021 05 17.
Article in English | MEDLINE | ID: mdl-34067674

ABSTRACT

The cAMP analogue 8-Br-cAMP-AM (8-Br) confers marked protection against global ischaemia/reperfusion of isolated perfused heart. We tested the hypothesis that 8-Br is also protective under clinically relevant conditions (regional ischaemia) when applied either before ischemia or at the beginning of reperfusion, and this effect is associated with the mitochondrial permeability transition pore (MPTP). 8-Br (10 µM) was administered to Langendorff-perfused rat hearts for 5 min either before or at the end of 30 min regional ischaemia. Ca2+-induced mitochondria swelling (a measure of MPTP opening) and binding of hexokinase II (HKII) to mitochondria were assessed following the drug treatment at preischaemia. Haemodynamic function and ventricular arrhythmias were monitored during ischaemia and 2 h reperfusion. Infarct size was evaluated at the end of reperfusion. 8-Br administered before ischaemia attenuated ventricular arrhythmias, improved haemodynamic function, and reduced infarct size during ischaemia/reperfusion. Application of 8-Br at the end of ischaemia protected the heart during reperfusion. 8-Br promoted binding of HKII to the mitochondria and reduced Ca2+-induced mitochondria swelling. Thus, 8-Br protects the heart when administered before regional ischaemia or at the beginning of reperfusion. This effect is associated with inhibition of MPTP via binding of HKII to mitochondria, which may underlie the protective mechanism.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/administration & dosage , Cardiovascular Agents/administration & dosage , Mitochondria, Heart/drug effects , Mitochondrial Permeability Transition Pore/metabolism , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , 8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , Animals , Calcium/metabolism , Disease Models, Animal , Drug Administration Schedule , Hemodynamics/drug effects , Hexokinase/metabolism , Isolated Heart Preparation , Male , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondrial Swelling/drug effects , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Wistar , Signal Transduction , Ventricular Function, Left/drug effects
3.
Naunyn Schmiedebergs Arch Pharmacol ; 394(8): 1675-1684, 2021 08.
Article in English | MEDLINE | ID: mdl-33937934

ABSTRACT

The generation of a reactive nitrenium ion by microsomal/mitochondrial cytochrome P450 (CYPs) from clozapine (CLZ) has been suggested as the main cause of cardiotoxicity by this drug. Previous studies indicated that thymoquinone (TQ) as an active constituent of Nigella sativa has pharmacological effects such as antioxidant, reactive oxygen species (ROS) scavenger, and inhibitory effect on CYPs enzymes. Therefore, we hypothesized that TQ with these pharmacological effects can reduce CLZ-induced toxicity in isolated cardiomyocytes and mitochondria. Rat left ventricular cardiomyocytes and mitochondria were isolated by collagenase perfusion and differential centrifugation respectively. Then, isolated cardiomyocytes and mitochondria were pretreated with different concentrations of TQ (1, 5, and 10 µmol/l) for 30 min and then followed by exposure to CLZ (50 µmol/l) for 6 h. After 6 h of incubation, using biochemical evaluations and flow cytometric analysis, the parameters of cellular toxicity including cytotoxicity, the level of oxidized/reduced glutathione (GSH/GSSG), malondialdehyde (MDA), reactive oxygen species (ROS) formation, lysosomal membrane integrity, mitochondria membrane potential (ΔΨm) collapse, and mitochondrial toxicity including succinate dehydrogenase (SDH) activity and mitochondrial swelling were analyzed. We observed a significant toxicity in isolated cardiomyocytes and mitochondria after exposure with CLZ which was related to ROS formation, oxidative stress, GSH depletion, lysosomal and mitochondrial damages, and mitochondrial dysfunction and swelling, while TQ pretreatment reverted the above toxic effect of CLZ on isolated cardiomyocytes and mitochondria. Our results indicate that TQ prevents and reverses CLZ-induced cytotoxicity and mitochondrial damages in isolated cardiomyocytes and mitochondria, providing an experimental basis for clinical treatment on CLZ-induced cardiotoxicity.


Subject(s)
Benzoquinones/pharmacology , Cardiotoxicity/prevention & control , Clozapine/toxicity , Myocytes, Cardiac/drug effects , Animals , Antipsychotic Agents/toxicity , Benzoquinones/administration & dosage , Cardiotoxicity/etiology , Cell Death/drug effects , Dose-Response Relationship, Drug , Glutathione/metabolism , Male , Malondialdehyde/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/drug effects , Mitochondria, Heart/pathology , Mitochondrial Swelling/drug effects , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
4.
Apoptosis ; 26(5-6): 277-292, 2021 06.
Article in English | MEDLINE | ID: mdl-33811561

ABSTRACT

Melanoma is an aggressive tumor with still poor therapy outcomes. δ-tocotrienol (δ-TT) is a vitamin E derivative displaying potent anti-cancer properties. Previously, we demonstrated that δ-TT triggers apoptosis in human melanoma cells. Here, we investigated whether it might also activate paraptosis, a non-canonical programmed cell death. In accordance with the main paraptotic features, δ-TT was shown to promote cytoplasmic vacuolization, associated with endoplasmic reticulum/mitochondrial dilation and protein synthesis, as well as MAPK activation in A375 and BLM cell lines. Moreover, treated cells exhibited a significant reduced expression of OXPHOS complex I and a marked decrease in oxygen consumption and mitochondrial membrane potential, culminating in decreased ATP synthesis and AMPK phosphorylation. This mitochondrial dysfunction resulted in ROS overproduction, found to be responsible for paraptosis induction. Additionally, δ-TT caused Ca2+ homeostasis disruption, with endoplasmic reticulum-derived ions accumulating in mitochondria and activating the paraptotic signaling. Interestingly, by using both IP3R and VDAC inhibitors, a close cause-effect relationship between mitochondrial Ca2+ overload and ROS generation was evidenced. Collectively, these results provide novel insights into δ-TT anti-melanoma activity, highlighting its ability to induce mitochondrial dysfunction-mediated paraptosis. δ-tocotrienol induces paraptotic cell death in human melanoma cells, causing endoplasmic reticulum dilation and mitochondrial swelling. These alterations induce an impairment of mitochondrial function, ROS production and calcium overload.


Subject(s)
Antineoplastic Agents/pharmacology , Calcium/metabolism , Melanoma/metabolism , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Regulated Cell Death/drug effects , Vitamin E/analogs & derivatives , Cell Line, Tumor , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Humans , Melanoma/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Swelling/drug effects , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/drug effects , Vitamin E/pharmacology
5.
Eur J Pharmacol ; 899: 174025, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33722590

ABSTRACT

Cerebral ischemia constitutes the most frequent type of cerebrovascular disease. The reduction of blood supply to the brain initiates the ischemic cascade starting from ionic imbalance to subsequent glutamate excitotoxicity, neuroinflammation and oxidative stress, eventually causing neuronal death. Previously, the authors have demonstrated the in vitro cytoprotective and antioxidant effects of a new arylidene malonate derivative, KM-34, against oxidizing agents like hydrogen peroxide, glutamate or Fe3+/ascorbate. Here, we examined for the first time the neuroprotective effect of KM-34 on ischemia/reperfusion models. In vitro, treatment with 10 and 50 µM KM-34 reduced the cellular death (propidium iodide incorporation) induced by oxygen glucose deprivation (OGD) in rat organotypic hippocampal slices cultures. In vivo, stroke was induced in male Wistar rats through middle cerebral artery occlusion (MCAO), followed by 23 h of reperfusion. KM-34 was orally administered 105 min after MCAO onset. We noticed that 1 mg/kg KM-34 reduced infarct volume and neurological score, and increased the latency to fall in the Hanging Wire test compared to vehicle-treated ischemic animals. While ischemic and sham-operated groups showed similar horizontal locomotor activity, vertical counts decreased after MCAO, suggesting that vertical movements are more sensitive to the ischemic injury. Treatment with KM-34 also alleviated the mitochondrial impairment (ROS generation, swelling and membrane potential dissipation) induced by transient MCAO but not significant alterations were found in oxidative stress parameters. Overall, the study provides preclinical evidences confirming the neuroprotective effects of a novel synthetic molecule and paved the way for future investigations regarding its therapeutic potential against brain ischemia/reperfusion injury.


Subject(s)
Brain/drug effects , Catechols/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/pharmacology , Reperfusion Injury/prevention & control , Animals , Behavior, Animal/drug effects , Brain/metabolism , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Locomotion/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Swelling/drug effects , Motor Activity/drug effects , Oxidative Stress/drug effects , Rats, Wistar , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Tissue Culture Techniques
6.
Methods Mol Biol ; 2240: 231-241, 2021.
Article in English | MEDLINE | ID: mdl-33423237

ABSTRACT

Mitochondria are the center for all metabolic pathways within the eukaryotic cell. Being responsible for the production of over 95% of the cell's requirement of adenosine triphosphate any effect on the function of mitochondria is sure to cause disruption of cellular activity and even viability. As such, it comes as no surprise that many diseases have mitochondrial dysfunction at their core. Understanding mitochondrial function and capacity in the context of a study is key for perceiving and explaining the behavior of said disease or toxic effect. Here, we describe a wide array of simple and yet elegant assays that can be easily implemented to ascertain the function of mitochondria and thus greatly improve the understanding of how a certain disease or compound causes its effects on the cellular function.


Subject(s)
Biological Assay , Energy Metabolism/drug effects , Mitochondria, Liver/drug effects , Toxicity Tests , Animals , Calcium/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Mitochondrial Swelling/drug effects , Oxygen Consumption/drug effects , Rats
7.
Biomed Pharmacother ; 135: 111186, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33395606

ABSTRACT

Trypanosoma cruzi is a protozoan parasite that causes Chagas disease, a neglected tropical disease that is endemic in Latin America and spreading worldwide due to globalization. The current treatments are based on benznidazole and nifurtimox; however, these drugs have important limitations and limited efficacy during the chronic phase, reinforcing the necessity of an alternative chemotherapy. For the last 30 years, our group has been evaluating the biological activity of naphthoquinones and derivatives on T. cruzi, and of the compounds tested, N1, N2 and N3 were found to be the most active in vitro. Here, we show the synthesis of a novel ß-lapachone-derived naphthoimidazolium named N4 and assess its activity on T. cruzi stages and the mechanism of action. The new compound was very active on all parasite stages (IC50/24 h in the range of 0.8-7.9 µM) and had a selectivity index of 5.4. Mechanistic analyses reveal that mitochondrial ROS production begins after short treatment starts and primarily affects the activity of complexes II-III. After 24 h treatment, a partial restoration of mitochondrial physiology (normal complexes II-III and IV activities and controlled H2O2 release) was observed; however, an extensive injury in its morphology was still detected. During treatment with N4, we also observed that trypanothione reductase activity increased in a time-dependent manner and concomitant with increased oxidative stress. Molecular docking calculations indicated the ubiquinone binding site of succinate dehydrogenase as an important interaction point with N4, as with the FMN binding site of dihydroorotate dehydrogenase. The results presented here may be a good starting point for the development of alternative treatments for Chagas disease and for understanding the mechanism of naphthoimidazoles in T. cruzi.


Subject(s)
Chagas Disease/drug therapy , Electron Transport Chain Complex Proteins/metabolism , Energy Metabolism/drug effects , Mitochondria/drug effects , Naphthoquinones/pharmacology , Protozoan Proteins/metabolism , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects , Chagas Disease/enzymology , Chagas Disease/parasitology , Dihydroorotate Dehydrogenase , Mitochondria/enzymology , Mitochondria/ultrastructure , Mitochondrial Swelling/drug effects , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Reactive Oxygen Species/metabolism , Succinate Dehydrogenase/metabolism , Trypanosoma cruzi/enzymology , Trypanosoma cruzi/growth & development , Trypanosoma cruzi/ultrastructure
8.
Cardiovasc Diabetol ; 20(1): 7, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33413413

ABSTRACT

BACKGROUND: Sodium-glucose linked transporter type 2 (SGLT-2) inhibition has been shown to reduce cardiovascular mortality in heart failure independently of glycemic control and prevents the onset of atrial arrhythmias, a common co-morbidity in heart failure with preserved ejection fraction (HFpEF). The mechanism behind these effects is not fully understood, and it remains unclear if they could be further enhanced by additional SGLT-1 inhibition. We investigated the effects of chronic treatment with the dual SGLT-1&2 inhibitor sotagliflozin on left atrial (LA) remodeling and cellular arrhythmogenesis (i.e. atrial cardiomyopathy) in a metabolic syndrome-related rat model of HFpEF. METHODS: 17 week-old ZSF-1 obese rats, a metabolic syndrome-related model of HFpEF, and wild type rats (Wistar Kyoto), were fed 30 mg/kg/d sotagliflozin for 6 weeks. At 23 weeks, LA were imaged in-vivo by echocardiography. In-vitro, Ca2+ transients (CaT; electrically stimulated, caffeine-induced) and spontaneous Ca2+ release were recorded by ratiometric microscopy using Ca2+-sensitive fluorescent dyes (Fura-2) during various experimental protocols. Mitochondrial structure (dye: Mitotracker), Ca2+ buffer capacity (dye: Rhod-2), mitochondrial depolarization (dye: TMRE) and production of reactive oxygen species (dye: H2DCF) were visualized by confocal microscopy. Statistical analysis was performed with 2-way analysis of variance followed by post-hoc Bonferroni and student's t-test, as applicable. RESULTS: Sotagliflozin ameliorated LA enlargement in HFpEF in-vivo. In-vitro, LA cardiomyocytes in HFpEF showed an increased incidence and amplitude of arrhythmic spontaneous Ca2+ release events (SCaEs). Sotagliflozin significantly reduced the magnitude of SCaEs, while their frequency was unaffected. Sotagliflozin lowered diastolic [Ca2+] of CaT at baseline and in response to glucose influx, possibly related to a ~ 50% increase of sodium sodium-calcium exchanger (NCX) forward-mode activity. Sotagliflozin prevented mitochondrial swelling and enhanced mitochondrial Ca2+ buffer capacity in HFpEF. Sotagliflozin improved mitochondrial fission and reactive oxygen species (ROS) production during glucose starvation and averted Ca2+ accumulation upon glycolytic inhibition. CONCLUSION: The SGLT-1&2 inhibitor sotagliflozin ameliorated LA remodeling in metabolic HFpEF. It also improved distinct features of Ca2+-mediated cellular arrhythmogenesis in-vitro (i.e. magnitude of SCaEs, mitochondrial Ca2+ buffer capacity, diastolic Ca2+ accumulation, NCX activity). The safety and efficacy of combined SGLT-1&2 inhibition for the treatment and/or prevention of atrial cardiomyopathy associated arrhythmias should be further evaluated in clinical trials.


Subject(s)
Arrhythmias, Cardiac/prevention & control , Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Glycosides/pharmacology , Heart Atria/drug effects , Heart Failure/drug therapy , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2/metabolism , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Calcium Signaling/drug effects , Disease Models, Animal , Heart Atria/metabolism , Heart Atria/physiopathology , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/physiopathology , Metabolic Syndrome/complications , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondrial Dynamics/drug effects , Mitochondrial Swelling/drug effects , Rats, Inbred WKY , Rats, Zucker , Reactive Oxygen Species/metabolism , Sodium-Calcium Exchanger/metabolism , Sodium-Glucose Transporter 1/metabolism
9.
Article in English | MEDLINE | ID: mdl-33171298

ABSTRACT

Even though marine dinoflagellates are important primary producers, many toxic species may alter the natural equilibrium of aquatic ecosystems and even generate human intoxication incidents, as they are the major causative agents of harmful algal blooms. In order to deepen the knowledge regarding benthic dinoflagellate adverse effects, the present study aims to clarify the influence of Gambierdiscus excentricus strain UNR-08, Ostreopsis cf. ovata strain UNR-03 and Prorocentrum lima strain UNR-01 crude extracts on rat mitochondrial energetic function and permeability transition pore (mPTP) induction. Our results, expressed in number of dinoflagellate cell toxic compounds tested in a milligram of mitochondrial protein, revealed that 934 cells mg prot-1 of G. excentricus, and 7143 cells mg prot-1 of both O. cf. ovata and P. lima negatively affect mitochondrial function, including by decreasing ATP synthesis-related membrane potential variations. Moreover, considerably much lower concentrations of dinoflagellate extracts (117 cells mg prot-1 of G. excentricus, 1429 cells mg prot-1 of O. cf. ovata and 714 cells mg prot-1 of P. lima) produced mPTP-induced swelling in Ca2+-loaded isolated mitochondria. The present study clearly demonstrates the toxicity of G. excentricus, O. cf. ovata and P. lima extracts at the mitochondrial level, which may lead to mitochondrial failure and consequent cell toxicity, and that G. excentricus always provide much more severe effects than O. cf. ovata and P. lima.


Subject(s)
Dinoflagellida/chemistry , Marine Toxins/toxicity , Mitochondria, Liver/drug effects , Mitochondrial Proteins/metabolism , Animals , Dinoflagellida/classification , Electron Transport Chain Complex Proteins/metabolism , Female , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/metabolism , Mitochondrial Swelling/drug effects , Oxygen Consumption/drug effects , Rats, Wistar , Seawater , Species Specificity
10.
Drug Res (Stuttg) ; 71(4): 219-227, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33276407

ABSTRACT

The possible action of polyphenolic compounds in the reduction of reactive oxygen species (ROS) and mitochondrial toxicity may suggest them as putative agents for the treatment of drug-induced mitochondrial dysfunction and cardiotoxicity. This study was designed to explore protective effect of ellagic acid (EA) against celecoxib-induced cellular and mitochondrial toxicity in cardiomyocytes and their isolated mitochondria. In order to do this, isolated cardiomyocytes and mitochondria were pretreated with 3 different concentrations of EA (10, 50 and 100 µM), after which celecoxib (16 µg/ml) was added to promote deleterious effects on cells and mitochondria. Using flow cytometry and biochemical methods, the parameters of cellular and mitochondrial toxicity were investigated. Our results showed that celecoxib (16 µg/ml) caused a significant decrease in cell viability, mitochondrial membrane potential (MMP), glutathione (GSH) in intact cardiomyocytes and succinate dehydrogenase (SDH) activity, MMP collapse, and mitochondrial swelling, and a significant increase in reactive oxygen species (ROS) formation, lipid peroxidation (LP) and oxidative stress in isolated mitochondria. Also, our results revealed that co-administration of EA (50 and 100 µM) with celecoxib significantly attenuated the cellular and mitochondrial toxicity effects. In this study, we showed that simultaneous treatment with of EA ameliorated the cellular and mitochondrial toxicity induced by celecoxib, with cardiomyocytes presenting normal activity compared to the control group, and mitochondria retaining their normal activity.


Subject(s)
Cardiotoxicity/prevention & control , Celecoxib/adverse effects , Ellagic Acid/pharmacology , Mitochondria/drug effects , Myocytes, Cardiac/drug effects , Animals , Cardiotoxicity/etiology , Cardiotoxicity/pathology , Cell Fractionation , Cells, Cultured , Ellagic Acid/therapeutic use , Lipid Peroxidation/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/pathology , Mitochondrial Swelling/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Primary Cell Culture , Rats , Reactive Oxygen Species/metabolism
11.
Oxid Med Cell Longev ; 2020: 5872645, 2020.
Article in English | MEDLINE | ID: mdl-33133347

ABSTRACT

The prefrontal cortex is the largest lobe of the brain and is consequently involved in stroke. There is no comprehensive practical pharmacological strategy for ameliorating prefrontal cortex injury induced by cerebral ischemia. Therefore, we studied the neuroprotective properties of verapamil (Ver) on mitochondrial dysfunction and morphological features of apoptosis in transient global ischemia/reperfusion (I/R). Ninety-six Wistar rats were allocated into four groups: control, I/R, I/R+Ver (10 mg/kg twice 1 hour prior to ischemia and 1 hour after reperfusion phase), and I/R+NaCl (vehicle). Animals were sacrificed, and mitochondrial dysfunction parameters (i.e., mitochondrial swelling, mitochondrial membrane potential, ATP concentration, ROS production, and cytochrome c release), antioxidant defense (i.e., superoxide dismutase, malondialdehyde, glutathione peroxidase, catalase, and caspase-3 activation), and morphological features of apoptosis were determined. The results showed that mitochondrial damage, impairment of antioxidant defense system, and apoptosis were significantly more prevalent in the I/R group in comparison with the other groups. Ver decreased mitochondrial damage by reducing oxidative stress, augmented the activity of antioxidant enzymes in the brain, and decreased apoptosis in the I/R neurons. The current study confirmed the role of oxidative stress and mitochondrial dysfunction in I/R progression and indicated the possible antioxidative mechanism of the neuroprotective activities of Ver.


Subject(s)
Apoptosis , Ischemic Attack, Transient/pathology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/pathology , Verapamil/pharmacology , Adenosine Triphosphate/metabolism , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Brain/drug effects , Brain/enzymology , Caspase 3/metabolism , Cell Survival/drug effects , Cytochromes c/metabolism , Ischemic Attack, Transient/complications , Male , Malondialdehyde/metabolism , Mitochondria/drug effects , Mitochondrial Swelling/drug effects , Nerve Degeneration/complications , Nerve Degeneration/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Prefrontal Cortex/drug effects , Prefrontal Cortex/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Reperfusion Injury/complications , Verapamil/administration & dosage
12.
Eur J Pharmacol ; 886: 173439, 2020 Nov 05.
Article in English | MEDLINE | ID: mdl-32871175

ABSTRACT

Takotsubo Syndrome (TS) is a kind of acute cardiac syndrome with a complex pathophysiological mechanism that remains to be elucidated. The relationship between TS and reactive oxygen species has received increasing attention over in recent years. Therefore, the relationship between TS and reactive oxygen species was investigated in vivo and in vitro. Isoprenaline (ISO) was used to induce TS and tempol (quercetin) was selected as a scavenger to eliminate reactive oxygen species in animal experiments, and echocardiography was used to determine the incidence of TS. The H9C2 cells were cultured with different reagents to investigate the detailed mechanism; Reactive oxygen species levels and mitochondrial function were evaluated. Cell apoptosis rate was analyzed by TUNEL staining and the proteins involved in the signaling pathways were examined by Western blotting. It was found that a high dose of tempol almost eliminated TS and protected the cardiac function. Moreover, tempol also decreased the reactive oxygen species levels and reduced lipid droplet deposition in myocardial tissue. In terms of the cultured cells, tempol preconditioning decreased reactive oxygen species production as well as lipid droplet deposition, and protected the mitochondrial function by reducing mitochondrial swelling, thereby maintaining the mitochondrial membrane potential (ΔΨm) at a level that was higher than that of controls. Furthermore, tempol could reduce cells apoptosis after ISO treatment and decrease the protein level of p38, which is a member of the MAPK family, which and thus plays an important role in regulating cells apoptosis. This antiapoptotic effect of tempol was similar to that of a control reagent, SB203580, which is a specific inhibitor of phospha-p38 (p-p38). This study demonstrated, for the first time, a sudden increase in reactive oxygen species and effects of the downstream cascades play core roles in the development of TS.


Subject(s)
Adrenergic beta-Agonists , Apoptosis/drug effects , Cyclic N-Oxides/therapeutic use , Free Radical Scavengers/therapeutic use , Isoproterenol , Mitochondria, Heart/drug effects , Signal Transduction/drug effects , Takotsubo Cardiomyopathy/chemically induced , Takotsubo Cardiomyopathy/prevention & control , p38 Mitogen-Activated Protein Kinases/drug effects , Animals , Cell Line , Lipid Droplets/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Swelling/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Spin Labels , Takotsubo Cardiomyopathy/mortality
13.
Toxicol Appl Pharmacol ; 405: 115205, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32835763

ABSTRACT

Triclosan (TCS) is an antimicrobial agent that was effectively banned by the FDA from hand soaps in 2016, hospital soaps in 2017, and hand sanitizers in 2019; however, TCS can still be found in a few products. At consumer-relevant, non-cytotoxic doses, TCS inhibits the functions of both mitochondria and mast cells, a ubiquitous cell type. Via the store-operated Ca2+ entry mechanism utilized by many immune cells, mast cells undergo antigen-stimulated Ca2+ influx into the cytosol, for proper function. Previous work showed that TCS inhibits Ca2+ dynamics in mast cells, and here we show that TCS also inhibits Ca2+ mobilization in human Jurkat T cells. However, the biochemical mechanism behind the Ca2+ dampening has yet to be elucidated. Three-dimensional super-resolution microscopy reveals that TCS induces mitochondrial swelling, in line with and extending the previous finding of TCS inhibition of mitochondrial membrane potential via its proton ionophoric activity. Inhibition of plasma membrane potential (PMP) by the canonical depolarizer gramicidin can inhibit mast cell function. However, use of the genetically encoded voltage indicators (GEVIs) ArcLight (pH-sensitive) and ASAP2 (pH-insensitive), indicates that TCS does not disrupt PMP. In conjunction with data from a plasma membrane-localized, pH-sensitive reporter, these results indicate that TCS, instead, induces cytosolic acidification in mast cells and T cells. Acidification of the cytosol likely inhibits Ca2+ influx by uncoupling the STIM1/ORAI1 interaction that is required for opening of plasma membrane Ca2+ channels. These results provide a mechanistic explanation of TCS disruption of Ca2+ influx and, thus, of immune cell function.


Subject(s)
Anti-Infective Agents/toxicity , Calcium/metabolism , Cytoplasm/drug effects , Mast Cells/drug effects , T-Lymphocytes/drug effects , Triclosan/toxicity , Calcium Channels/metabolism , Cell Degranulation/drug effects , Cell Line , Cell Membrane/drug effects , Cytoplasm/metabolism , Humans , Hydrogen-Ion Concentration , Mast Cells/metabolism , Membrane Potentials/drug effects , Mitochondrial Swelling/drug effects , T-Lymphocytes/metabolism
14.
J Orthop Surg Res ; 15(1): 275, 2020 Jul 23.
Article in English | MEDLINE | ID: mdl-32703256

ABSTRACT

BACKGROUND: Ischemia-reperfusion injury of the spinal cord (SCII) often leads to unalterable neurological deficits, which may be associated with apoptosis induced by oxidative stress and inflammation. Astaxanthin (AST) is a strong antioxidant and anti-inflammatory agent with multitarget neuroprotective effects. This study aimed to investigate the potential therapeutic effects of AST for SCII and the molecular mechanism. METHODS: Rat models of SCII with abdominal aortic occlusion for 40 min were carried out to investigate the effects of AST on the recovery of SCII. Tarlov's scores were used to assess the neuronal function; HE and TUNEL staining were used to observe the pathological morphology of lesions. Neuron oxidative stress and inflammation were measured using commercial detection kits. Flow cytometry was conducted to assess the mitochondrial swelling degree. Besides, Western blot assay was used to detect the expression of PI3K/Akt/GSK-3ß pathway-related proteins, as well as NOX2 and NLRP3 proteins. RESULTS: The results demonstrated that AST pretreatment promoted the hind limb motor function recovery and alleviated the pathological damage induced by SCII. Moreover, AST significantly enhanced the antioxidative stress response and attenuated mitochondrial swelling. However, AST pretreatment hardly inhibited the levels of proinflammatory cytokines after SCII. Most importantly, AST activated p-Akt and p-GSK-3ß expression levels. Meanwhile, cotreatment with LY294002 (a PI3K inhibitor) was found to abolish the above protective effects observed with the AST pretreatment. CONCLUSION: Overall, these results suggest that AST pretreatment not only mitigates pathological tissue damage but also effectively improves neural functional recovery following SCII, primarily by alleviating oxidative stress but not inhibiting inflammation. A possible underlying molecular mechanism of AST may be mainly attributed to the activation of PI3K/Akt/GSK-3ß pathway.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reperfusion Injury/drug therapy , Signal Transduction/drug effects , Spinal Cord Ischemia/drug therapy , Animals , Anti-Inflammatory Agents , Antioxidants , Apoptosis/drug effects , Male , Mitochondrial Swelling/drug effects , Neuroprotective Agents , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/genetics , Spinal Cord Ischemia/genetics , Spinal Cord Ischemia/metabolism , Spinal Cord Ischemia/pathology , Xanthophylls/administration & dosage , Xanthophylls/pharmacology
15.
J Biochem Mol Toxicol ; 34(10): e22557, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32583933

ABSTRACT

Today, it has been proven that the nanoparticles such as superparamagnetic iron oxide nanoparticles (SPIONs) have widespread use in biomedical applications, for instance, in magnetic resonance imaging and targeted delivery of drugs. Despite many studies on SPIONs in diagnosing some diseases like cancer, it has not been investigated on the oral tongue squamous cell carcinoma (OTSCC) detection by the NPs. Hence, the present study has been designed to assess the in vitro cytotoxicity of SPIONs on the isolated mitochondria of OTSCC by mitochondrial tests. Isolated mitochondria were removed from the separated cancer and control tissues from the squamous cells of tango in male Wistar rats (6 or 8 weeks) and exposed to the different concentrations of SPIONs (30, 60, and 120 nM). A rise in the production of reactive oxygen species is one of the significant mechanisms of this study, followed by a collapse of mitochondrial membrane potential, the escape of mitochondrial cytochrome c, and mitochondrial swelling in the exposed isolated mitochondria of OTSCC with SPIONs. Furthermore, our results indicated that the exposure to the SPIONs reduced the activity of succinate dehydrogenase in complex II of the mitochondria obtained from cancerous oral tongue squamous. So the SPIONs can induce selective cytotoxicity on the OTSCC mitochondria without significant effects on the control mitochondria. Based on the results and further studies about in vivo experiments in this regard, it is concluded the SPIONs may be a hopeful therapeutic candidate for the treatment of OTSCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Magnetic Iron Oxide Nanoparticles , Mitochondria/drug effects , Oxidative Stress/drug effects , Squamous Cell Carcinoma of Head and Neck/drug therapy , Tongue Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cytochromes c/metabolism , In Vitro Techniques , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondrial Swelling/drug effects , Rats , Reactive Oxygen Species/metabolism , Squamous Cell Carcinoma of Head and Neck/enzymology , Squamous Cell Carcinoma of Head and Neck/metabolism , Succinate Dehydrogenase/metabolism , Tongue Neoplasms/enzymology , Tongue Neoplasms/metabolism
16.
Environ Toxicol ; 35(10): 1114-1124, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32432806

ABSTRACT

Apart from the anticancer, antioxidant, anti-inflammatory effects, and inhibition of aromatase, chrysin is involved in the protection of cardiovascular disorders. Cardiovascular complications are the main cause of death induced by aluminum phosphide (AlP) which is related to oxidative stress and mitochondrial damages. For this purpose, we investigated the effect of chrysin as an antioxidant and mitochondrial protective agent against AlP-induced toxicity in isolated cardiomyocytes and mitochondria obtained from rat heart ventricular. Using by biochemical and flow cytometry, cell viability, reactive oxygen species (ROS) formation, mitochondria membrane potential (MMP), lysosomal membrane integrity, malondialdehyde (MDA) content, and glutathione (GSH) and oxidized glutathione (GSSG) content were measured in isolated cardiomyocytes. Also, mitochondrial toxicity parameters such as mitochondrial NADH/succinate dehydrogenase activity, mitochondrial swelling, ROS formation, MMP collapse, and lipid peroxidation were analyzed in isolated mitochondria. Our results showed that the administration of chrysin (up to 10 µM) efficiently decreased (P < 0.05) cytotoxicity, oxidative, lysosomal, and mitochondrial damages induced by AlP, in isolated cardiomyocytes. Also, our finding in isolated mitochondria showed that chrysin (up to 10 µM) significantly (P < 0.05) decreased AlP-induced mitochondrial toxicity. These findings demonstrated that chrysin as an antioxidant and mitochondrial protective agent exert protective effect in wild-type cardiomyocyte treated with AlP. It was concluded that chrysin significantly reduced the toxicity of AlP in isolated cardiomyocytes and mitochondria. Due to the very low toxicity of chrysin for humans, it could be a promising agent in treatment of AlP poisoning.


Subject(s)
Aluminum Compounds/toxicity , Flavonoids/pharmacology , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Phosphines/toxicity , Protective Agents/pharmacology , Animals , Cardiotoxicity , Cells, Cultured , Glutathione/metabolism , Humans , Lipid Peroxidation/drug effects , Lysosomes/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/metabolism , Mitochondrial Swelling/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
17.
Biosci Rep ; 40(5)2020 05 29.
Article in English | MEDLINE | ID: mdl-32368781

ABSTRACT

Amino acid sequence from 65th to 76th residue of the N-terminus of Chromogranin A (CGA-N12) is an antimicrobial peptide (AMP). Our previous studies showed that CGA-N12 reduces Candida tropicalis mitochondrial membrane potential. Here, we explored the mechanism that CGA-N12 collapsed the mitochondrial membrane potential by investigations of its action on the mitochondrial permeability transition pore (mPTP) complex of C. tropicalis. The results showed that CGA-N12 induced cytochrome c (Cyt c) leakage, mitochondria swelling and led to polyethylene glycol (PEG) of molecular weight 1000 Da penetrate mitochondria. mPTP opening inhibitors bongkrekic acid (BA) could contract the mitochondrial swelling induced by CGA-N12, but cyclosporin A (CsA) could not. Therefore, we speculated that CGA-N12 could induce C. tropicolis mPTP opening by preventing the matrix-facing (m) conformation of adenine nucleotide transporter (ANT), thereby increasing the permeability of the mitochondrial membrane and resulted in the mitochondrial potential dissipation.


Subject(s)
Antifungal Agents/pharmacology , Candida tropicalis/drug effects , Chromogranin A/pharmacology , Fungal Proteins/agonists , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondrial Permeability Transition Pore/agonists , Peptide Fragments/pharmacology , Pore Forming Cytotoxic Proteins/pharmacology , Candida tropicalis/metabolism , Candida tropicalis/ultrastructure , Cytochromes c/metabolism , Fungal Proteins/metabolism , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial ADP, ATP Translocases/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Mitochondrial Swelling/drug effects
18.
Clin Exp Pharmacol Physiol ; 47(9): 1554-1563, 2020 09.
Article in English | MEDLINE | ID: mdl-32314438

ABSTRACT

Erythromycin (ERY) is a risk factor for cardiotoxicity through the mitochondria pathway. In the current study, we tested the hypothesis that erythromycin could impair mitochondrial function and oxidative stress and 1,25-dihydroxivitamin D3 (calcitriol) treatment could prevent these effects in rat heart isolated mitochondria. Rat heart mitochondria were isolated with mechanical lysis and differential centrifugation. Then isolated mitochondria were first pretreated with three different concentrations of 1,25-dihydroxivitamin D3 (2.5, 5 and 10 µmol/L) for 5 minutes at 37°C, after which erythromycin (10 µmol/L) was added to promote deleterious effects on mitochondria. During 1 hour of incubation, using by flow cytometry and biochemical evaluations, the parameters of mitochondrial toxicity were evaluated, including: succinate dehydrogenase (SDH) activity, mitochondrial swelling, mitochondrial membrane potential (MMP) collapse, reactive oxygen species (ROS) formation and lipid peroxidation (LP). The results showed that erythromycin (10 µmol/L) caused a significant change in mitochondrial function, ROS formation, mitochondrial swelling, MMP collapse, increasing lipid peroxidation and oxidative stress. 1,25-dihydroxivitamin D3 (10 µmol/L) reverted the effect of erythromycin on the tested parameters . In this study, we showed that erythromycin impairs mitochondrial function and induces mitochondrial toxicity in rat heart isolated mitochondria, which were reverted by calcitriol. These findings suggest that 1,25-dihydroxivitamin D3 may be a preventive/therapeutic strategy for cardiotoxicity complications caused by erythromycin.


Subject(s)
Antioxidants/pharmacology , Calcitriol/pharmacology , Erythromycin/toxicity , Heart Diseases/prevention & control , Mitochondria, Heart/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Animals , Cardiotoxicity , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/pathology , Lipid Peroxidation/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondrial Swelling/drug effects , Rats, Wistar
19.
Toxicol Mech Methods ; 30(6): 427-437, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32312132

ABSTRACT

Di-(2-ethylhexyl) phthalate (DEHP) and its main metabolite, monoethylhexyl phthalic acid (MEHP), are a serious threat to human and animals' health in the current century. However, their exact mechanism to induce nephrotoxicity is not clear. In the current study, we addressed toxic effects of MEHP and DEHP on embryonic human kidney cells (HEK-293 cell line) and kidney tissue of rats, respectively. In the HEK-293, MTT assay and oxidative stress parameters were measured after treatment with different concentrations of MEHP. For in vivo study, rats were treated with different doses of DEHP (50, 100, 200, 400 mg/kg) via gavage administration for 45 days. The renal function biomarkers (BUN and creatinine) were determined in serum of rats. Mitochondrial toxic parameters including MTT, mitochondrial membrane potential (MMP), mitochondrial swelling, and also oxidative stress parameters were measured in isolated kidney mitochondria. Histopathological effects of DEHP were also evaluated in rats' kidneys. We demonstrated that MEHP induced oxidative stress and cytotoxicity in HEK-293 cells in a concentration dependent manner. The administration of DEHP led to histopathological changes in kidney tissue, which concurred with BUN and creatinine alternations in serum of rats. The results of present study showed a significant mitochondrial dysfunction and oxidative stress confirmed by enhancement of mitochondrial swelling, mitochondrial reactive oxygen species (ROS) and malondialdehyde (MDA), and reduction of MMP and mitochondrial glutathione (GSH). Taken together, this study showed that DEHP/MEHP resulted in mitochondrial dysfunction and oxidative damage, which suggest a vital role of mitochondria in DEHP/MEHP-induced nephrotoxicity.


Subject(s)
Diethylhexyl Phthalate/analogs & derivatives , Kidney Diseases/chemically induced , Kidney/drug effects , Mitochondria/drug effects , Oxidative Stress/drug effects , Animals , Biomarkers/blood , Blood Urea Nitrogen , Creatinine/blood , Diethylhexyl Phthalate/toxicity , HEK293 Cells , Humans , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Lipid Peroxidation/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Swelling/drug effects , Rats, Wistar , Reactive Oxygen Species/metabolism
20.
Metabolism ; 107: 154227, 2020 06.
Article in English | MEDLINE | ID: mdl-32275974

ABSTRACT

OBJECTIVE: L5, a highly electronegative subtype of low-density lipoprotein (LDL), is likely associated with the development of atherosclerosis and cardiovascular diseases. Normal LDL is composed mainly of apolipoprotein (Apo) B, but L5 has additional proteins such as ApoE. We previously demonstrated that L5 induces endothelial cell senescence by increasing mitochondrial reactive oxygen species. In the present study, we examined the effect of L5 on mitochondrial function in cardiomyocytes. METHODS: We used the Seahorse XF24 extracellular flux analyzer to examine the effect of L5 and its components on mitochondrial energy production. The effects of L5 on mitochondrial morphology were examined by immunofluorescence using MitoTracker Green FM and the corresponding probes in H9c2 cardiomyoblasts. Mitochondrial permeability was assessed by using a calcium-induced swelling assay with a voltage-dependent anion-selective channel (VDAC) inhibitor to determine VDAC-dependence both in vitro and in vivo. L5 without ApoE, referred to as △L5, was used to clarify the role of ApoE in L5-induced mitochondrial dysfunction. RESULTS: L5 not only significantly decreased basal (P < 0.05) and maximal respiration (P < 0.01) but also reduced spare respiratory capacity (P < 0.01) in H9c2 cells. Additionally, L5 caused phosphorylation of Drp1 and mitochondrial fission. Recombinant ApoE mimicked the mitochondrial effects of L5, but △L5 did not cause similar effects. After entering cells, ApoE on L5 colocalized with mitochondrial VDAC and caused mitochondria swelling both in vitro and in vivo. This effect was also seen with recombinant ApoE but not △L5. CONCLUSIONS: ApoE may play an important role in electronegative LDL-induced mitochondrial dysfunction through the opening of the mitochondrial permeability transition pore via the interaction of ApoE and VDAC.


Subject(s)
Apolipoproteins E/metabolism , Lipoproteins, LDL/pharmacology , Mitochondrial Diseases/chemically induced , Mitochondrial Diseases/metabolism , Myocytes, Cardiac/metabolism , Animals , Cell Line , Energy Metabolism/drug effects , Humans , In Vitro Techniques , Lipoproteins, LDL/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Membranes/metabolism , Mitochondrial Swelling/drug effects , Myocytes, Cardiac/drug effects , Oxygen Consumption/drug effects , Reactive Oxygen Species
SELECTION OF CITATIONS
SEARCH DETAIL
...