Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Antioxidants (Basel) ; 12(8)2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37627587

ABSTRACT

The incidence of kidney disease is increasing worldwide. Acute kidney injury (AKI) can strongly favor cardio-renal syndrome (CRS) type 3 development. However, the mechanism involved in CRS development is not entirely understood. In this sense, mitochondrial impairment in both organs has become a central axis in CRS physiopathology. This study aimed to elucidate the molecular mechanisms associated with cardiac mitochondrial impairment and its role in CRS development in the folic acid-induced AKI (FA-AKI) model. Our results showed that 48 h after FA-AKI, the administration of N-acetyl-cysteine (NAC), a mitochondrial glutathione regulator, prevented the early increase in inflammatory and cell death markers and oxidative stress in the heart. This was associated with the ability of NAC to protect heart mitochondrial bioenergetics, principally oxidative phosphorylation (OXPHOS) and membrane potential, through complex I activity and the preservation of glutathione balance, thus preventing mitochondrial dynamics shifting to fission and the decreases in mitochondrial biogenesis and mass. Our data show, for the first time, that mitochondrial bioenergetics impairment plays a critical role in the mechanism that leads to heart damage. Furthermore, NAC heart mitochondrial preservation during an AKI event can be a valuable strategy to prevent CRS type 3 development.

2.
Front Microbiol ; 13: 949272, 2022.
Article in English | MEDLINE | ID: mdl-36118191

ABSTRACT

A challenge in the study of gastrointestinal microbiota (GITm) is the validation of the genomic data with metabolic studies of the microbial communities to understand how the microbial networks work during health and sickness. To gain insights into the metabolism of the GITm, feces from healthy and sick rats with cancer were inoculated in a defined synthetic medium directed for anaerobic prokaryote growth (INC-07 medium). Significant differences between cultures of healthy and sick individuals were found: 1) the consumption of the carbon source and the enzyme activity involved in their catabolism (e.g., sucrase, lactase, lipases, aminotransferases, and dehydrogenases); 2) higher excretion of acetic, propionic, isobutyric, butyric, valeric, and isovaleric acids; 3) methane production; 4) ability to form biofilms; and 5) up to 500 amplicon sequencing variants (ASVs) identified showed different diversity and abundance. Moreover, the bowel inflammation induced by cancer triggered oxidative stress, which correlated with deficient antioxidant machinery (e.g., NADPH-producing enzymes) determined in the GITm cultures from sick individuals in comparison with those from control individuals. Altogether, the data suggested that to preserve the microbial network between bacteria and methanogenic archaea, a complete oxidation of the carbon source may be essential for healthy microbiota. The correlation of 16S rRNA gene metabarcoding between cultures and feces, as well as metabolomic data found in cultures, suggest that INC-07 medium may be a useful tool to understand the metabolism of microbiota under gut conditions.

3.
FEBS J ; 288(13): 4064-4080, 2021 07.
Article in English | MEDLINE | ID: mdl-33400378

ABSTRACT

Under physiological conditions, cells produce low basal levels of reactive oxygen species (ROS); however, in pathologic conditions ROS production increases dramatically, generating high concentrations of toxic unsaturated aldehydes. Aldehyde dehydrogenases (ALDHs) are responsible for detoxification of these aldehydes protecting the cell. Due to the physiological relevance of these enzymes, it is important to design strategies to modulate their activity. It was previously reported that omeprazole activation of ALDH1A1 protected Escherichia coli cells overexpressing this enzyme, from oxidative stress generated by H2 O2 . In this work, omeprazole cell protection potential was evaluated in eukaryotic cells. AS-30D cell or hepatocyte suspensions were subjected to a treatment with omeprazole and exposure to light (that is required to activate omeprazole in the active site of ALDH) and then exposed to H2 O2 . Cells showed viability similar to control cells, total activity of ALDH was preserved, while cell levels of lipid aldehydes and oxidative stress markers were maintained low. Cell protection by omeprazole was avoided by inhibition of ALDHs with disulfiram, revealing the key role of these enzymes in the protection. Additionally, omeprazole also preserved ALDH2 (mitochondrial isoform) activity, diminishing lipid aldehyde levels and oxidative stress in this organelle, protecting mitochondrial respiration and transmembrane potential formation capacity, from the stress generated by H2 O2 . These results highlight the important role of ALDHs as part of the antioxidant system of the cell, since if the activity of these enzymes decreases under stress conditions, the viability of the cell is compromised.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Lipid Peroxidation/drug effects , Omeprazole/pharmacology , Oxidative Stress/drug effects , Aldehyde Dehydrogenase 1 Family/genetics , Animals , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Female , Humans , Hydrogen Peroxide/pharmacology , Light , Oxidants/pharmacology , Rats, Wistar , Reactive Oxygen Species/metabolism
4.
Biochem Res Int ; 2020: 5253108, 2020.
Article in English | MEDLINE | ID: mdl-33489376

ABSTRACT

Mitochondrial permeability transition is characterized by the opening of a transmembranal pore that switches membrane permeability from specific to nonspecific. This structure allows the free traffic of ions, metabolites, and water across the mitochondrial inner membrane. The opening of the permeability transition pore is triggered by oxidative stress along with calcium overload. In this work, we explored if oxidative stress is a consequence, rather than an effector of the pore opening, by evaluating the interaction of agaric acid with the adenine nucleotide translocase, a structural component of the permeability transition pore. We found that agaric acid induces transition pore opening, increases the generation of oxygen-derived reactive species, augments the oxidation of unsaturated fatty acids in the membrane, and promotes the detachment of cytochrome c from the inner membrane. The effect of agaric acid was inhibited by the antioxidant tamoxifen in association with decreased binding of the thiol reagent eosin-3 maleimide to the adenine nucleotide translocase. We conclude that agaric acid promotes the opening of the pore, increasing ROS production that exerts oxidative modification of critical thiols in the adenine nucleotide translocase.

5.
Toxicol Appl Pharmacol ; 370: 65-77, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30878505

ABSTRACT

The resveratrol (RSV) efficacy to affect the proliferation of several cancer cell lines was initially examined. RSV showed higher potency to decrease growth of metastatic HeLa and MDA-MB-231 (IC50 = 200-250 µM) cells than of low metastatic MCF-7, SiHa and A549 (IC50 = 400-500 µM) and non-cancer HUVEC and 3T3 (IC50≥600 µM) cells after 48 h exposure. In order to elucidate the biochemical mechanisms underlying RSV anti-cancer effects, the energy metabolic pathways and the oxidative stress metabolism were analyzed in HeLa cells as metastatic-type cell model. RSV (200 µM/48 h) significantly decreased both glycolysis and oxidative phosphorylation (OxPhos) protein contents (30-90%) and fluxes (40-70%) vs. non-treated cells. RSV (100 µM/1-5 min) also decreased at a greater extent OxPhos flux (net ADP-stimulated respiration) of isolated tumor mitochondria (> 50%) than of non-tumor mitochondria (< 50%), particularly with succinate as oxidizable substrate. In addition, RSV promoted an excessive cellular ROS (2-3 times) production corresponding with a significant decrement in the SOD activity (but not in its content) and GSH levels; whereas the catalase, glutahione reductase, glutathione peroxidase and glutathione-S-transferase activities (but not their contents) remained unchanged. RSV (200 µM/48 h) also induced cellular death although not by apoptosis but rather by promoting a strong mitophagy activation (65%). In conclusion, RSV impaired OxPhos by inducing mitophagy and ROS over-production, which in turn halted metastatic HeLa cancer cell growth.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Proliferation/drug effects , Neoplasms/pathology , Oxidative Phosphorylation/drug effects , Oxidative Stress/drug effects , Resveratrol/pharmacology , 3T3 Cells , Animals , Cell Line, Tumor , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , MCF-7 Cells , Mice , Mitophagy/drug effects , Neoplasm Metastasis/prevention & control , Phytochemicals/pharmacology
6.
Biochem Cell Biol ; 97(2): 187-192, 2019 04.
Article in English | MEDLINE | ID: mdl-30332552

ABSTRACT

In the kidney, the accumulation of heavy metals such as Cd2+ produces mitochondrial dysfunctions, i.e., uncoupling of the oxidative phosphorylation, inhibition of the electron transport through the respiratory chain, and collapse of the transmembrane electrical gradient. This derangement may be due to the fact that Cd2+ induces the transition of membrane permeability from selective to nonselective via the opening of a transmembrane pore. In fact, Cd2+ produces this injury through the stimulation of oxygen-derived radical generation, inducing oxidative stress. Several molecules have been used to avoid or even reverse Cd2+-induced mitochondrial injury, for instance, cyclosporin A, resveratrol, dithiocarbamates, and even EDTA. The aim of this study was to explore the possibility that the antioxidant tamoxifen could protect mitochondria from the deleterious effects of Cd2+. Our results indicate that the addition of 1 µmol/L Cd2+ to mitochondria collapsed the transmembrane electrical gradient, induced the release of cytochrome c, and increased both the generation of H2O2 and the oxidative damage to mitochondrial DNA (among other measured parameters). Of interest, these mitochondrial dysfunctions were ameliorated after the addition of tamoxifen.


Subject(s)
Cadmium/toxicity , Hydrogen Peroxide/metabolism , Kidney/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Animals , Kidney/pathology , Mitochondria/pathology , Oxidation-Reduction/drug effects
7.
Cell Biochem Biophys ; 76(4): 445-450, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30159781

ABSTRACT

Several studies have demonstrated that the mitochondrial membrane switches from selective to non-selective permeability because of its improved matrix Ca2+ accumulation and oxidative stress. This process, known as permeability transition, evokes severe dysfunction in mitochondria through the opening of a non-specific pore, whose chemical nature is still under discussion. There are some proposals regarding the components of the pore structure, e.g., the adenine nucleotide translocase and dimers of the F1 Fo-ATP synthase. Our results reveal that Ca2+ induces oxidative stress, which not only increases lipid peroxidation and ROS generation but also brings about both the collapse of the transmembrane potential and the membrane release of cytochrome c. Additionally, it is shown that Ca2+ increases the binding of the probe eosin-5-maleimide to adenine nucleotide translocase. Interestingly, these effects are diminished after the addition of ADP. It is suggested that pore opening is caused by the binding of Ca2+ to the adenine nucleotide translocase.


Subject(s)
Calcium/pharmacology , Mitochondria/metabolism , Mitochondrial ADP, ATP Translocases/metabolism , Oxidative Stress/drug effects , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Animals , Cytochromes c/metabolism , Kidney/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondrial ADP, ATP Translocases/chemistry , Protein Binding , Rats , Reactive Oxygen Species/metabolism , Succinate Dehydrogenase/chemistry , Succinate Dehydrogenase/metabolism , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/metabolism
8.
Cell Biol Int ; 41(12): 1356-1366, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28884894

ABSTRACT

Heavy metal ions are known to produce harmful alterations on kidney function. Specifically, the accumulation of Hg2+ in kidney tissue may induce renal failure. In this work, the protective effect of CDP-choline against the deleterious effects induced by Hg2+ on renal function was studied. CDP-choline administered ip at a dose of 125 mg/kg body weight prevented the damage induced by Hg2+ administration at a dose of 3 mg/kg body weight. The findings indicate that CDP-choline guards mitochondria against Hg2+ -toxicity by preserving their ability to retain matrix content, such as accumulated Ca2+ . This nucleotide also protected mitochondria from Hg2+ -induced loss of the transmembrane electric gradient and from the generation of hydrogen peroxide and membrane TBARS. In addition, CDP-choline avoided the oxidative damage of mtDNA and inhibited the release of the interleukins IL-1 and IL6, recognized as markers of acute inflammatory reaction. After the administration of Hg2+ and CDP, CDP-choline maintained nearly normal levels of renal function and creatinine clearance, as well as blood urea nitrogen (BUN) and serum creatinine.


Subject(s)
Cytidine Diphosphate Choline/pharmacology , Kidney/drug effects , Mercury/toxicity , Mitochondria/drug effects , Animals , Creatine/metabolism , Interleukin-1/metabolism , Interleukin-6/metabolism , Kidney/metabolism , Kidney/pathology , Kidney Function Tests , Male , Membrane Potentials/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Oxidation-Reduction , Rats , Rats, Wistar , Thiobarbituric Acid Reactive Substances/metabolism
9.
Biochem Cell Biol ; 95(5): 556-562, 2017 10.
Article in English | MEDLINE | ID: mdl-28595020

ABSTRACT

In this work, we studied the protective effects of tamoxifen (TAM) on disulfiram (Dis)-induced mitochondrial membrane insult. The results indicate that TAM circumvents the inner membrane leakiness manifested as Ca2+ release, mitochondrial swelling, and collapse of the transmembrane electric gradient. Furthermore, it was found that TAM prevents inactivation of the mitochondrial enzyme aconitase and detachment of cytochrome c from the inner membrane. Interestingly, TAM also inhibited Dis-promoted generation of hydrogen peroxide. Given that TAM is an antioxidant molecule, it is plausible that its protection may be due to the inhibition of Dis-induced oxidative stress.


Subject(s)
Disulfiram/adverse effects , Mitochondrial Membranes/drug effects , Tamoxifen/pharmacology , Animals , Calcium/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/pathology , Oxidative Stress/drug effects , Rats , Rats, Wistar
10.
Cell Biol Int ; 40(12): 1349-1356, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27730705

ABSTRACT

In this work, we studied the effect of tamoxifen and cyclosporin A on mitochondrial permeability transition caused by addition of the thiol-oxidizing pair Cu2+ -orthophenanthroline. The findings indicate that tamoxifen and cyclosporin A circumvent the oxidative membrane damage manifested by matrix Ca2+ release, mitochondrial swelling, and transmembrane electrical gradient collapse. Furthermore, it was found that tamoxifen and cyclosporin A prevent the generation of TBARs promoted by Cu2+ -orthophenanthroline, as well as the inactivation of the mitochondrial enzyme aconitase and disruption of mDNA. Electrophoretic analysis was unable to demonstrate a cross-linking reaction between membrane proteins. Yet, it was found that Cu2+ -orthophenanthroline induced the generation of reactive oxygen species. It is thus plausible that membrane leakiness is due to an oxidative stress injury.


Subject(s)
Copper/toxicity , Mitochondria/drug effects , Mitochondria/metabolism , Organometallic Compounds/toxicity , Oxidative Stress/drug effects , Phenanthrolines/toxicity , Tamoxifen/pharmacology , Blotting, Western , Calcium/metabolism , Cyclosporine/pharmacology , DNA, Mitochondrial/metabolism , Electrophoresis, Polyacrylamide Gel , Glutathione/metabolism , Hydrogen Peroxide/metabolism , Mitochondria/pathology , Protective Agents/pharmacology , Thiobarbituric Acid Reactive Substances/metabolism
11.
Life Sci ; 139: 108-13, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26316446

ABSTRACT

AIMS: Mitochondrial permeability transition is a process established through massive Ca(2+) load in addition to an inducer reagent. Ebselen (Ebs), an antioxidant seleno compound, has been introduced as a reagent which inhibits mitochondrial dysfunction induced by permeability transition. Paradoxically enough, it has been shown that Ebs may also be able to induce the opening of the mitochondrial non-selective pores. This study was performed with the purpose of establishing the membrane system involved in Ebs-induced pore opening. MAIN METHODS: Permeability transition was appraised by analyzing the following: i) matrix Ca(2+) release, and mitochondrial swelling, ii) efflux of cytochrome c, and iii) the inhibition of superoxide dismutase. All of these adverse reactions were inhibited by N-ethylmaleimide and cyclosporin A. KEY FINDINGS: At concentrations from 5 to 20 µM, we found that Ebs induces non-specific membrane permeability. Remarkably, Ebs blocks the binding of the fluorescent reagent eosin-5-maleimide to the thiol groups of the adenine nucleotide translocase. SIGNIFICANCE: Based on the above, it is tempting to hypothesize that Ebs induces pore opening through its binding to the ADP/ATP carrier.


Subject(s)
Antioxidants/pharmacology , Azoles/pharmacology , Mitochondria/drug effects , Mitochondrial ADP, ATP Translocases/metabolism , Mitochondrial Swelling/drug effects , Organoselenium Compounds/pharmacology , Permeability/drug effects , Animals , Atractyloside , Calcium/metabolism , Isoindoles , Mitochondria/metabolism , Rats
12.
Int J Biochem Cell Biol ; 65: 209-21, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26073129

ABSTRACT

Oxidative phosphorylation (OxPhos) is functional and sustains tumor proliferation in several cancer cell types. To establish whether mitochondrial ß-oxidation of free fatty acids (FFAs) contributes to cancer OxPhos functioning, its protein contents and enzyme activities, as well as respiratory rates and electrical membrane potential (ΔΨm) driven by FFA oxidation were assessed in rat AS-30D hepatoma and liver (RLM) mitochondria. Higher protein contents (1.4-3 times) of ß-oxidation (CPT1, SCAD) as well as proteins and enzyme activities (1.7-13-times) of Krebs cycle (KC: ICD, 2OGDH, PDH, ME, GA), and respiratory chain (RC: COX) were determined in hepatoma mitochondria vs. RLM. Although increased cholesterol content (9-times vs. RLM) was determined in the hepatoma mitochondrial membranes, FFAs and other NAD-linked substrates were oxidized faster (1.6-6.6 times) by hepatoma mitochondria than RLM, maintaining similar ΔΨm values. The contents of ß-oxidation, KC and RC enzymes were also assessed in cells. The mitochondrial enzyme levels in human cervix cancer HeLa and AS-30D cells were higher than those observed in rat hepatocytes whereas in human breast cancer biopsies, CPT1 and SCAD contents were lower than in human breast normal tissue. The presence of CPT1 and SCAD in AS-30D mitochondria and HeLa cells correlated with an active FFA utilization in HeLa cells. Furthermore, the ß-oxidation inhibitor perhexiline blocked FFA utilization, OxPhos and proliferation in HeLa and other cancer cells. In conclusion, functional mitochondria supported by FFA ß-oxidation are essential for the accelerated cancer cell proliferation and hence anti-ß-oxidation therapeutics appears as an alternative promising approach to deter malignant tumor growth.


Subject(s)
Fatty Acids/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Neoplasms/pathology , 3T3 Cells , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Female , HeLa Cells , Humans , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , MCF-7 Cells , Mice , Oxidative Phosphorylation , Rats , Rats, Wistar
13.
Biochem Cell Biol ; 93(3): 185-91, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25589288

ABSTRACT

Hyperthyroidism represents an increased risk factor for cardiovascular morbidity, especially when the heart is subjected to an ischemia/reperfusion process. The aim of this study was to explore the possible protective effect of the nucleotide citicoline on the susceptibility of hyperthyroid rat hearts to undergo reperfusion-induced damage, which is associated with mitochondrial dysfunction. Hence, we analyzed the protective effect of citicoline on the electrical behavior and on the mitochondrial function in rat hearts. Hyperthyroidism was established after a daily i.p. injection of triiodothyronine (at 2 mg/kg of body weight) during 5 days. Thereafter, citicoline was administered i.p. (at 125 mg/kg of body weight) for 5 days. In hyperthyroid rat hearts, citicoline protected against reperfusion-induced ventricular arrhythmias. Moreover, citicoline maintained the accumulation of mitochondrial Ca(2+), allowing mitochondria to reach a high transmembrane electric gradient that protected against the release of cytochrome c. It also preserved the activity of the enzyme aconitase that inhibited the release of cytokines. The protection also included the inhibition of oxidative stress-induced mDNA disruption. We conclude that citicoline protects against the reperfusion damage that is found in the hyperthyroid myocardium. This effect might be due to its inhibitory action on the permeability transition in mitochondria.


Subject(s)
Cardiotonic Agents/pharmacology , Cytidine Diphosphate Choline/pharmacology , Heart/drug effects , Hyperthyroidism/physiopathology , Mitochondria, Heart/drug effects , Aconitate Hydratase/metabolism , Animals , Calcium/metabolism , DNA, Mitochondrial/metabolism , Hyperthyroidism/chemically induced , Hyperthyroidism/complications , Mitochondria, Heart/metabolism , Myocardial Reperfusion , Myocardial Reperfusion Injury/prevention & control , Rats , Superoxide Dismutase/metabolism , Triiodothyronine/adverse effects
14.
J Steroid Biochem Mol Biol ; 143: 416-23, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24923730

ABSTRACT

Hyperthyroidism, known to have deleterious effects on heart function, and is associated with an enhanced metabolic state, implying an increased production of reactive oxygen species. Tamoxifen is a selective antagonist of estrogen receptors. These receptors make the hyperthyroid heart more susceptible to ischemia/reperfusion. Tamoxifen is also well-known as an antioxidant. The aim of the present study was to explore the possible protective effect of tamoxifen on heart function in hyperthyroid rats. Rats were injected daily with 3,5,3'-triiodothyronine at 2mg/kg body weight during 5 days to induce hyperthyroidism. One group was treated with 10mg/kg tamoxifen and another was not. The protective effect of the drug on heart rhythm was analyzed after 5 min of coronary occlusion followed by 5 min reperfusion. In hyperthyroid rats not treated with tamoxifen, ECG tracings showed post-reperfusion arrhythmias, and heart mitochondria isolated from the ventricular free wall lost the ability to accumulate and retain matrix Ca(2+) and to form a high electric gradient. Both of these adverse effects were avoided with tamoxifen treatment. Hyperthyroidism-induced oxidative stress caused inhibition of cis-aconitase and disruption of mitochondrial DNA, effects which were also avoided by tamoxifen treatment. The current results support the idea that tamoxifen inhibits the hypersensitivity of hyperthyroid rat myocardium to reperfusion damage, probably because its antioxidant activity inhibits the mitochondrial permeability transition.


Subject(s)
Arrhythmias, Cardiac/drug therapy , Estrogen Antagonists/therapeutic use , Hyperthyroidism/complications , Mitochondria, Heart/drug effects , Myocardial Reperfusion Injury/drug therapy , Tamoxifen/therapeutic use , Animals , Arrhythmias, Cardiac/etiology , Cytochromes c/metabolism , Female , Mitochondria, Heart/pathology , Myocardial Reperfusion Injury/etiology , Oxidative Stress , Rats , Rats, Wistar , Thiobarbituric Acid Reactive Substances/metabolism
15.
Life Sci ; 96(1-2): 53-8, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24389400

ABSTRACT

AIMS: Oxidative stress emerges after reperfusion of an organ following an ischemic period and results in tissue damage. In the heart, an amplified generation of reactive oxygen species and a significant Ca(2+) accumulation cause ventricular arrhythmias and mitochondrial dysfunction. This occurs in consequence of increased non-specific permeability. A number of works have shown that permeability transition is a common substrate that underlies the reperfusion-induced heart injury. The aim of this work was to explore the possibility that CDP-choline may circumvent heart damage and mitochondrial permeability transition. MAIN METHODS: Rats were injected i.p. with CDP-choline at 20 mg/kg body weight. Heart electric behavior was followed during a closure/opening cycle of the left coronary descendent artery. Heart mitochondria were isolated from rats treated with CDP-choline, and their function was evaluated by analyzing Ca(2+) movements, achievement of a high level of the transmembrane potential, and respiratory control. Oxidative stress was estimated following the activity of the enzymes cis-aconitase and superoxide dismutase, as well as the disruption of mitochondrial DNA. KEY FINDINGS: This study shows that CDP-choline avoided ventricular arrhythmias and drop of blood pressure. Results also show that mitochondria, isolated from CDP-choline-treated rats, maintained selective permeability, retained accumulated Ca(2+), an elevated value of transmembrane potential, and a high ratio of respiratory control. Furthermore, activity of cis-aconitase enzyme and mDNA structure were preserved. SIGNIFICANCE: This work introduces CDP-choline as a useful tool to preserve heart function from reperfusion damage by inhibiting mitochondrial permeability transition.


Subject(s)
Cardiotonic Agents/therapeutic use , Cell Membrane Permeability/drug effects , Cytidine Diphosphate Choline/therapeutic use , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Myocardial Reperfusion Injury/prevention & control , Myocardium , Animals , Cardiotonic Agents/pharmacology , Cytidine Diphosphate Choline/pharmacology , Male , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Myocardium/pathology , Rats , Rats, Wistar
16.
Cell Biol Int ; 38(3): 287-95, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23765583

ABSTRACT

Chemical modification of primary amino groups of mitochondrial membrane proteins by the fluorescent probe fluorescamine induces non-specific membrane permeabilisation. Titration of the lysine ϵ-amino group promoted efflux of accumulated Ca(2+), collapse of transmembrane potential and mitochondrial swelling. Ca(2+) release was inhibited by cyclosporin A. Considering the latter, we assumed that fluorescamine induces permeability transition. Carboxyatractyloside also inhibited the reaction. Using a polyclonal antibody for adenine nucleotide translocase, Western blot analysis showed that the carrier appeared labelled with the fluorescent probe. The results point out the importance of the ϵ-amino group of lysine residues, located in the adenine nucleotide carrier, on the modulation of membrane permeability, since its blockage suffices to promote opening of the non-specific nanopore.


Subject(s)
Cell Membrane Permeability/drug effects , Fluorescamine/pharmacology , Lysine/metabolism , Membrane Potentials/drug effects , Mitochondrial ADP, ATP Translocases/metabolism , Animals , Atractyloside/analogs & derivatives , Atractyloside/metabolism , Calcium/metabolism , Cell Membrane Permeability/physiology , Ion Transport/drug effects , Ion Transport/physiology , Male , Membrane Potentials/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial ADP, ATP Translocases/drug effects , Mitochondrial Swelling/drug effects , Mitochondrial Swelling/physiology , Rats , Rats, Wistar
17.
Peptides ; 53: 202-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23880546

ABSTRACT

Cecropin 3 (Ccrp3) is an antimicrobial peptide from Anopheles albimanus, which is expressed during Plasmodium berghei infection. Here, we report that synthetic Ccrp3, aside from antibacterial activity, also shows cardio regulatory functions. In rats, Ccrp3 significantly diminishes blood pressure as well as the heartbeat frequency at nanomolar concentration. Ccrp3 affect the rat cardiac muscle mitochondria, inducing uncoupling of oxidative phosphorylation, oxygen consumption and transport of Ca(2). Ccrp3 treatment of the mitochondria causes mitochondrial damage promoting oxidative stress, causing overproduction of reactive oxygen species (ROS) and inhibition of superoxide dismutase. At nM concentration, Ccrp3 inhibits superoxide dismutase activity through direct interaction, diminishing by its enzymatic activity. Ccrp3 induces the release of the pro-apoptotic marker Bax from the mitochondria. Altogether, these results suggest that Ccrp3 pro-oxidative activity on cardiac muscle mitochondria could be responsible for triggering the heartbeat frequency and blood pressure lowering observed the Ccrp3 injected rats.


Subject(s)
Cecropins/pharmacology , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Oxidative Stress/drug effects , Animals , Anopheles , Biological Transport/drug effects , Blood Pressure/drug effects , Calcium/metabolism , Male , Oxygen Consumption/drug effects , Rats , Rats, Wistar , Thiobarbituric Acid Reactive Substances/metabolism
18.
FEBS J ; 280(3): 927-38, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23206332

ABSTRACT

Succinate-driven oxidation via complex II (CII) may have a significant contribution towards the high rates of production of reactive oxygen species (ROS) by mitochondria. Here, we show that the CII Q site inhibitor thenoyltrifluoroacetone (TTFA) blocks succinate + rotenone-driven ROS production, whereas the complex III (CIII) Qo inhibitor stigmatellin has no effect, indicating that CII, not CIII, is the ROS-producing site. The complex I (CI) inhibitor rotenone partially reduces the ROS production driven by high succinate levels (5 mm), which is commonly interpreted as being due to inhibition of a reverse electron flow from CII to CI. However, experimental evidence presented here contradicts the model of reverse electron flow. First, ROS levels produced using succinate + rotenone were significantly higher than those produced using glutamate + malate + rotenone. Second, in tumor mitochondria, succinate-driven ROS production was significantly increased (not decreased) by rotenone. Third, in liver mitochondria, rotenone had no effects on succinate-driven ROS production. Fourth, using isolated heart or hepatoma (AS-30D) mitochondria, the CII Qp anti-cancer drug mitochondrially targeted vitamin E succinate (MitoVES) induced elevated ROS production in the presence of low levels of succinate(0.5 mm), but rotenone had no effect. Using sub-mitochondrial particles, the Cu-based anti-cancer drug Casiopeina II-gly enhanced succinate-driven ROS production. Thus, the present results are inconsistent with and question the interpretation of reverse electron flow from CII to CI and the rotenone effect on ROS production supported by succinate oxidation. Instead, a thermodynamically more favorable explanation is that, in the absence of CIII or complex IV (CIV) inhibitors (which, when added, facilitate reverse electron flow by inducing accumulation of ubiquinol, the CI product), the CII redox centers are the major source of succinate-driven ROS production.


Subject(s)
Electron Transport Complex II/metabolism , Electron Transport Complex I/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Animals , Cattle , Cell Line, Tumor , Electron Transport/drug effects , Glutamic Acid/pharmacology , Hydrogen Peroxide/metabolism , Malates/pharmacology , Mitochondria/drug effects , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Polyenes/pharmacology , Rats , Rotenone/pharmacology , Succinic Acid/pharmacology , Thenoyltrifluoroacetone/pharmacology
19.
Biochim Biophys Acta ; 1817(9): 1597-607, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22627082

ABSTRACT

The effects of α-tocopheryl succinate (α-TOS), α-tocopheryl acetyl ether (α-TEA) and triphenylphosphonium-tagged vitamin E succinate (mitochondrially targeted vitamin E succinate; MitoVES) on energy-related mitochondrial functions were determined in mitochondria isolated from AS-30D hepatoma and rat liver, bovine heart sub-mitochondrial particles (SMPs), and in rodent and human carcinoma cell lines and rat hepatocytes. In isolated mitochondria, MitoVES stimulated basal respiration and ATP hydrolysis, but inhibited net state 3 (ADP-stimulated) respiration and Ca(2+) uptake, by collapsing the membrane potential at low doses (1-10µM). Uncoupled mitochondrial respiration and basal respiration of SMPs were inhibited by the three drugs at concentrations at least one order of magnitude higher and with different efficacy: MitoVES>α-TEA>α-TOS. At high doses (>10µM), the respiratory complex II (CII) was the most sensitive MitoVES target. Acting as an uncoupler at low doses, this agent stimulated total O(2) uptake, collapsed ∆ψ(m), inhibited oxidative phosphorylation and induced ATP depletion in rodent and human cancer cells more potently than in normal rat hepatocytes. These findings revealed that in situ tumor mitochondria are preferred targets of the drug, indicating its clinical relevance.


Subject(s)
Mitochondria/drug effects , Neoplasms/drug therapy , Vitamin E/analogs & derivatives , Adenosine Triphosphate/chemistry , Animals , Calcium/metabolism , Cattle , Cell Line, Tumor , Cell Respiration/drug effects , Electron Transport Complex I/metabolism , Electron Transport Complex II/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/physiology , Rats
20.
J Bioenerg Biomembr ; 43(6): 757-64, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22108703

ABSTRACT

Permeability transition was examined in heart mitochondria isolated from neonate rats. We found that these mitochondria were more susceptible to Ca(2+)-induced membrane leakiness than mitochondria from adult rats. In K(+) containing medium, at 25 °C, mitochondria were unable to accumulate Ca(2+). Conversely, in Na(+) containing medium, mitochondria accumulated effectively Ca(2+). At 15 °C mitochondria accumulated Ca(2+) regardless of the presence of K(+). Kinetics of Ca(2+) accumulation showed a similar Vmax as that of adult mitochondria. Lipid milieu of inner membrane contained more unsaturated fatty acids than adult mitochondria. Aconitase inhibition and high thiobarbituric acid-reactive substances (TBARS) indicate that oxidative stress caused mitochondrial damage. In addition, proteomics analysis showed that there is a considerable diminution of succinate dehydrogenase C and subunit 4 of cytochrome oxidase in neonate mitochondria. Our proposal is that dysfunction of the respiratory chain makes neonate mitochondria more susceptible to damage by oxidative stress.


Subject(s)
Calcium/pharmacology , Mitochondria, Heart/metabolism , Mitochondrial Membranes/metabolism , Oxidative Stress/drug effects , Animals , Electron Transport/drug effects , Permeability/drug effects , Potassium/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...