Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Med Sci ; 17(16): 2454-2467, 2020.
Article in English | MEDLINE | ID: mdl-33029088

ABSTRACT

Aims: Dexmedetomidine (Dex) as a highly selective α2-adrenoceptor agonist, was widely used anesthetic in perioperative settings, whether Dex induces cardiac hypertrophy during perioperative administration is unknown. Methods: The effects of Dex on cardiac hypertrophy were explored using the transverse aortic constriction model and neonatal rat cardiomyocytes. Results: We reported that Dex induces cardiomyocyte hypertrophy with activated ERK, AKT, PKC and inactivated AMPK in both wild-type mice and primary cultured rat cardiomyocytes. Additionally, pre-administration of Dex protects against transverse aortic constriction induced-heart failure in mice. We found that Dex up-regulates the activation of ERK, AKT, and PKC via suppression of AMPK activation in rat cardiomyocytes. However, suppression of mitochondrial coupling efficiency and membrane potential by FCCP blocks Dex induced AMPK inactivation as well as ERK, AKT, and PKC activation. All of these effects are blocked by the α2-adrenoceptor antagonist atipamezole. Conclusion: The present study demonstrates Dex preconditioning induces cardiac hypertrophy that protects against heart failure through mitochondria-AMPK pathway in perioperative settings.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adrenergic alpha-2 Receptor Agonists/pharmacology , Cardiomegaly/chemically induced , Dexmedetomidine/pharmacology , Heart Failure/prevention & control , Adrenergic alpha-2 Receptor Agonists/therapeutic use , Animals , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/administration & dosage , Cells, Cultured , Dexmedetomidine/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Heart Failure/etiology , Heart Failure/pathology , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Primary Cell Culture , Rats , Signal Transduction/drug effects
2.
PLoS One ; 12(8): e0184246, 2017.
Article in English | MEDLINE | ID: mdl-28859158

ABSTRACT

Metabolic stress evoked by myocardial ischemia leads to impairment of cardiac excitation and contractility. We studied the mechanisms by which metabolic inhibition affects the activity of L-type Ca2+ channels (LTCCs) in frog ventricular myocytes. Metabolic inhibition induced by the protonophore FCCP (as well as by 2,4- dinitrophenol, sodium azide or antimycin A) resulted in a dose-dependent reduction of LTCC current (ICa,L) which was more pronounced during ß-adrenergic stimulation with isoprenaline. ICa,L was still reduced by metabolic inhibition even in the presence of 3 mM intracellular ATP, or when the cell was dialysed with cAMP or ATP-γ-S to induce irreversible thiophosphorylation of LTCCs, indicating that reduction in ICa,L is not due to ATP depletion and/or reduced phosphorylation of the channels. However, the effect of metabolic inhibition on ICa,L was strongly attenuated when the mitochondrial F1F0-ATP-synthase was blocked by oligomycin or when the cells were dialysed with the non-hydrolysable ATP analogue AMP-PCP. Moreover, increasing the intracellular pH buffering capacity or intracellular dialysis of the myocytes with an alkaline solution strongly attenuated the inhibitory effect of FCCP on ICa,L. Thus, our data demonstrate that metabolic inhibition leads to excessive ATP hydrolysis by the mitochondrial F1F0-ATP-synthase operating in the reverse mode and this results in intracellular acidosis causing the suppression of ICa,L. Limiting ATP break-down by F1F0-ATP-synthase and the consecutive development of intracellular acidosis might thus represent a potential therapeutic approach for maintaining a normal cardiac function during ischemia.


Subject(s)
Calcium Channels, L-Type/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Myocardial Contraction/genetics , Myocardial Ischemia/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium Channels, L-Type/genetics , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/administration & dosage , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Isoproterenol/administration & dosage , Mitochondria/enzymology , Muscle Cells/drug effects , Muscle Cells/metabolism , Myocardial Contraction/drug effects , Myocardial Ischemia/genetics , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rana esculenta , Stress, Physiological/drug effects , Stress, Physiological/genetics
3.
J Assist Reprod Genet ; 33(12): 1621-1631, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27639998

ABSTRACT

PURPOSE: Temporary and reversible downregulation of metabolism may improve the survival of tissues exposed to non-physiological conditions during transport, in vitro culture, and cryopreservation. The objectives of the study were to (1) optimize the concentration and duration of carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP-a mitochondrial uncoupling agent) exposures for biopsies of domestic cat ovarian tissue and (2) examine the effects of FCCP pre-exposures on follicle integrity after tissue culture and/or cryopreservation. METHODS: Biopsies of cat ovarian tissue were first treated with various concentrations of FCCP (0, 10, 40, or 200 nM) for 10 or 120 min to determine the most suitable pre-exposure conditions. Based on these results, tissues were pre-exposed to 200 nM FCCP for 120 min for the subsequent studies on culture and cryopreservation. In all experiments and for each treatment group, tissue activity and integrity were measured by mitochondrial membrane potential (relative optical density of rhodamine 123 fluorescence), follicular viability (calcein assay), follicular morphology (histology), granulosa cell proliferation (Ki-67 immunostaining), and follicular density. RESULTS: Ovarian tissues incubated with 200 nM FCCP for 120 min led to the lowest mitochondrial activity (1.17 ± 0.09; P < 0.05) compared to control group (0 nM; 1.30 ± 0.12) while maintaining a constant percentage of viable follicles (75.3 ± 7.8 %) similar to the control group (71.8 ± 11.7 %; P > 0.05). After 2 days of in vitro culture, percentage of viable follicles (78.8 ± 8.9 %) in similar pre-exposure conditions was higher (P < 0.05) than in the absence of FCCP (61.2 ± 12.0 %) with percentages of morphologically normal follicles (57.6 ± 17.3 %) not different from the fresh tissue (70.2 ± 7.1 %; P > 0.05). Interestingly, percentages of cellular proliferation and follicular density were unaltered by the FCCP exposures. Based on the indicators mentioned above, the FCCP-treated tissue fragments did not have a better follicle integrity after freezing and thawing. CONCLUSIONS: Pre-exposure to 200 nM FCCP during 120 min protects and enhances the follicle integrity in cat ovarian tissue during short-term in vitro culture. However, FCCP does not appear to exert a beneficial or detrimental effect during ovarian tissue cryopreservation.


Subject(s)
Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/administration & dosage , Cryopreservation , Ovarian Follicle/growth & development , Ovary/growth & development , Animals , Cats , Cell Proliferation/drug effects , Female , Freezing , Humans , Membrane Potential, Mitochondrial/drug effects , Ovarian Follicle/drug effects , Ovary/drug effects , Tissue Culture Techniques/methods
4.
Free Radic Biol Med ; 60: 233-45, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23376234

ABSTRACT

The Nlrp3 inflammasome is activated in response to an array of environmental and endogenous molecules leading to caspase-1-dependent IL-1ß processing and secretion by myeloid cells. Several identified Nlrp3 inflammasome activators also trigger reactive oxygen species (ROS) production. However, the initial concept that NADPH oxidases are the primary source of ROS production during inflammasome activation is becoming less accepted. Therefore, the importance of mitochondria-derived ROS has been recently explored. In this study, we explore the impact of mitochondria dysfunction and ROS production on Nlrp3 inflammasome stimulation and IL-1ß secretion induced by serum amyloid A (SAA) in primary mouse peritoneal macrophages. To induce mitochondrial dysfunction, we utilized antimycin A, which blocks electron flow at complex III, and carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP), a mitochondrial oxidative phosphorylation uncoupler. We also utilized a superoxide dismutase mimetic, MnTBAP, which targets the mitochondria, as well as the broad-spectrum antioxidants DPI (diphenyleneiodonium chloride) and ebselen. Our findings demonstrate that SAA alone induces mitochondrial ROS in a time-dependent manner. We observed that MnTBAP and ebselen blocked IL-1ß secretion caused by SAA only when added before stimulation, and DPI augmented IL-1ß secretion. Surprisingly, these effects were not directly related to intracellular or mitochondrial ROS levels. We also found that mitochondria-targeted drugs increased IL-1ß secretion regardless of their impact on mitochondrial function and ROS levels, suggesting that mitochondrial ROS-dependent and -independent mechanisms play a role in the Nlrp3 inflammasome/IL-1ß secretion axis in SAA-stimulated cells. Finally, we found that FCCP significantly sustained the association of the Nlrp3 inflammasome complex, which could explain the most robust effect among the drugs tested in enhancing IL-1ß secretion in SAA-treated cells. Overall, our data suggest that the Nlrp3 inflammasome/IL-1ß secretion axis is a very highly regulated inflammatory pathway that is susceptible not only to changes in mitochondrial or intracellular ROS, but also to changes in overall mitochondrial function.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Mitochondria/metabolism , Animals , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/administration & dosage , Carrier Proteins/genetics , Energy Metabolism/drug effects , Inflammasomes/drug effects , Inflammasomes/genetics , Interleukin-1beta/genetics , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Mice , Mitochondria/drug effects , Mitochondria/pathology , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidation-Reduction/drug effects , Primary Cell Culture , Reactive Oxygen Species/metabolism , Serum Amyloid A Protein/pharmacology
5.
Exp Neurol ; 218(2): 381-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19477175

ABSTRACT

Experimental traumatic brain injury (TBI) leads to a rapid and extensive necrosis at the primary site of injury that appears to be driven in part by significant mitochondrial dysfunction. The present study is based on the hypothesis that TBI-induced, aberrant glutamate release increases mitochondrial Ca(2+) cycling/overload ultimately leading to mitochondrial damage. Previous work from our laboratory demonstrates that mitochondrial uncoupling during the acute phases of TBI-induced excitotoxicity can reduce mitochondrial Ca(2+) uptake (cycling), ROS production and mitochondrial damage resulting in neuroprotection and improved behavioral outcome. The current study was designed to determine the optimal dosage and therapeutic window of opportunity for the potent mitochondrial uncoupler FCCP following moderate TBI. For this study, we used young adult male Sprague-Dawley rats (300-350 g); either sham-operated or moderately (1.5 mm) injured using the controlled cortical impactor (CCI) model of TBI. In the first set of studies animals were injected with either vehicle (100% DMSO) or different concentrations of FCCP (0.5, 1, 2.5 and 5 mg/kg in 100% DMSO) intraperitoneally at 5 min post-injury; tested behaviorally at 10 days and cortical sparing assessed at 18 days post-injury. The results demonstrate that of all the dosages tested, 2.5 mg/kg rendered the maximum improvement in behavioral outcomes and tissue spared. Using this optimal dose (2.5 mg/kg) and time point for intervention (5 min post-injury), we assessed mitochondrial bioenergetics and mitochondrial structural integrity 24 h post-injury. Furthermore, using this dosage we assessed mitochondrial bioenergetics and Ca(2+) loading at 3 and 6 h post-injury to further verify our target mechanism and establish these assessments as a valid endpoint to use as a means to determine the therapeutic window of FCCP. To begin to address the window of opportunity for maintaining mitochondrial homeostasis, the optimal dose of FCCP was then administered at 5 min, 3, 6, or 24 h post-injury and several parameters of mitochondrial function were used as outcome measures. The results demonstrate that a prolonged window of opportunity exists for targeting mitochondrial dysfunction using uncouplers following TBI and give insight into the cellular pathology associated with TBI.


Subject(s)
Brain Injuries/drug therapy , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Homeostasis/drug effects , Mitochondria/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Uncoupling Agents/pharmacology , Animals , Brain Injuries/metabolism , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Male , Mitochondria/metabolism , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley , Time Factors , Uncoupling Agents/administration & dosage
6.
Toxicol Appl Pharmacol ; 125(2): 309-21, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8171438

ABSTRACT

Cytosolic Ca2+ overload may play a key role in the process of lead-induced retinal injury and degeneration. We report that retinal calcium content was elevated following developmental and in vitro lead exposure. To determine the concentration-dependent effects of Ca2+ (5-1000 nM) on retinal mitochondrial bioenergetics an isolation procedure was developed. Isolated mitochondria were efficiently coupled; had good respiratory control ratios with the NAD-linked substrates, glutamate or pyruvate plus malate (G/M or P/M), and the FAD-linked substrate, succinate plus rotenone (S/R); and possessed a Na+/Ca2+ exchanger. The major finding was that at equimolar [Ca2+] > or = 35 nM, mitochondria were more sensitive to and exhibited a greater degree of inhibition of coupled and uncoupled respiration with NAD-linked substrates compared to S/R. At all [Ca2+], decreases in State 3 and uncoupled respiration were similar, thereby eliminating the ATP synthase and ADP/ATP translocase as sites of inhibition and suggesting that opening the mitochondrial permeability transition pore (MTP) did not contribute to the inhibition. The effects of toxicological [Ca2+] were: (1) blocked by ruthenium red, (2) blocked by dibucaine only in the presence of NAD-linked substrates, and (3) partially reversed by NAD+ with G/M after opening the MTP. Results with G/M suggest that Ca2+ acts on the inner membrane phospholipase A2 to decrease NADH CoQ reductase activity and/or produce a NAD+ leak, whereas with S/R, Ca2+ may inhibit succinate dehydrogenase. In conclusion, Ca2+ inhibits retinal mitochondrial ATP production, which may contribute to the retinal cell injury and death observed in developmentally lead-exposed rats.


Subject(s)
Calcium/pharmacology , Mitochondria/metabolism , Retina/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium/physiology , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/administration & dosage , Energy Metabolism/drug effects , Energy Metabolism/physiology , Female , In Vitro Techniques , Lead/analysis , Microscopy, Electron , Mitochondria/drug effects , Mitochondria/ultrastructure , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Rats , Rats, Inbred Strains , Retina/drug effects , Retina/ultrastructure , Subcellular Fractions/enzymology , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...