Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(16)2021 Aug 07.
Article in English | MEDLINE | ID: mdl-34445229

ABSTRACT

Symbiosis between the mitochondrion and the ancestor of the eukaryotic cell allowed cellular complexity and supported life. Mitochondria have specialized in many key functions ensuring cell homeostasis and survival. Thus, proper communication between mitochondria and cell nucleus is paramount for cellular health. However, due to their archaebacterial origin, mitochondria possess a high immunogenic potential. Indeed, mitochondria have been identified as an intracellular source of molecules that can elicit cellular responses to pathogens. Compromised mitochondrial integrity leads to release of mitochondrial content into the cytosol, which triggers an unwanted cellular immune response. Mitochondrial nucleic acids (mtDNA and mtRNA) can interact with the same cytoplasmic sensors that are specialized in recognizing genetic material from pathogens. High-energy demanding cells, such as neurons, are highly affected by deficits in mitochondrial function. Notably, mitochondrial dysfunction, neurodegeneration, and chronic inflammation are concurrent events in many severe debilitating disorders. Interestingly in this context of pathology, increasing number of studies have detected immune-activating mtDNA and mtRNA that induce an aberrant production of pro-inflammatory cytokines and interferon effectors. Thus, this review provides new insights on mitochondria-driven inflammation as a potential therapeutic target for neurodegenerative and primary mitochondrial diseases.


Subject(s)
Mitochondria/immunology , Neurodegenerative Diseases/immunology , Animals , Cytokines/immunology , DNA, Mitochondrial/immunology , Humans , Mitochondria/pathology , Neurodegenerative Diseases/pathology , RNA, Mitochondrial/immunology
2.
Mol Ther Methods Clin Dev ; 17: 1071-1078, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32478122

ABSTRACT

Leigh syndrome, or infantile necrotizing subacute encephalopathy (OMIM #256000), is one of the most common manifestations of mitochondrial dysfunction, due to mutations in more than 75 genes, with mutations in respiratory complex I subunits being the most common cause. In the present study, we used the recently described PHP.B serotype, characterized by efficient capacity to cross the blood-brain barrier, to express the hNDUFS4 gene in the Ndufs4 -/- mouse model of Leigh disease. A single intravenous injection of PHP.B-hNDUFS4 in adult Ndufs4 -/- mice led to a normalization of the body weight, marked amelioration of the rotarod performance, delayed onset of neurodegeneration, and prolongation of the lifespan up to 1 year of age. hNDUFS4 protein was expressed in virtually all brain regions, leading to a partial recovery of complex I activity. Our findings strongly support the feasibility and effectiveness of adeno-associated viral vector (AAV)-mediated gene therapy for mitochondrial disease, particularly with new serotypes showing increased permeability to the blood-brain barrier in order to achieve widespread expression in the central nervous system.

3.
Mol Ther ; 28(8): 1918-1930, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32562616

ABSTRACT

Moderate overexpression of Opa1, the master regulator of mitochondrial cristae morphology, significantly improved mitochondrial damage induced by drugs, surgical denervation, or oxidative phosphorylation (OXPHOS) defects due to specific impairment of a single mitochondrial respiratory chain complex. Here, we investigated the effectiveness of this approach in the Mpv17-/- mouse, characterized by profound, multisystem mitochondrial DNA (mtDNA) depletion. After the crossing with Opa1tg mice, we found a surprising anticipation of the severe, progressive focal segmental glomerulosclerosis, previously described in Mpv17-/- animals as a late-onset clinical feature (after 12-18 months of life). In contrast, Mpv17-/- animals from this new "mixed" strain died at 8-9 weeks after birth because of severe kidney failure However, Mpv17-/-::Opa1tg mice lived much longer than Mpv17-/- littermates and developed the kidney dysfunction much later. mtDNA content and OXPHOS activities were significantly higher in Mpv17-/-::Opa1tg than in Mpv17-/- kidneys and similar to those for wild-type (WT) littermates. Mitochondrial network and cristae ultrastructure were largely preserved in Mpv17-/-::Opa1tg versus Mpv17-/- kidney and isolated podocytes. Mechanistically, the protective effect of Opa1 overexpression in this model was mediated by a block in apoptosis due to the stabilization of the mitochondrial cristae. These results demonstrate that strategies aiming at increasing Opa1 expression or activity can be effective against mtDNA depletion syndromes.


Subject(s)
GTP Phosphohydrolases/genetics , Gene Expression , Kidney Diseases/etiology , Kidney Diseases/metabolism , Membrane Proteins/deficiency , Animals , Apoptosis/genetics , DNA, Mitochondrial , Disease Models, Animal , Disease Susceptibility , GTP Phosphohydrolases/metabolism , Immunohistochemistry , Kidney Diseases/pathology , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Biological , Oxidative Phosphorylation , Podocytes/metabolism , Podocytes/pathology , Podocytes/ultrastructure
4.
Sci Rep ; 8(1): 14013, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30228311

ABSTRACT

Mitohormesis is an adaptive response induced by a mild mitochondrial stress that promotes longevity and metabolic health in different organisms. This mechanism has been proposed as the cause of the increase in the survival in Coq7+/- (Mclk1+/-) mice, which show hepatic reduction of COQ7, early mitochondrial dysfunction and increased oxidative stress. Our study shows that the lack of COQ9 in Coq9Q95X mice triggers the reduction of COQ7, COQ6 and COQ5, which results in an increase in life expectancy. However, our results reveal that the hepatic CoQ levels are not decreased and, therefore, neither mitochondrial dysfunction or increased oxidative stress are observed in liver of Coq9Q95X mice. These data point out the tissue specific differences in CoQ biosynthesis. Moreover, our results suggest that the effect of reduced levels of COQ7 on the increased survival in Coq9Q95X mice may be due to mitochondrial mechanisms in non-liver tissues or to other unknown mechanisms.


Subject(s)
Longevity , Mitochondria, Liver/metabolism , Ubiquinone/biosynthesis , Animals , Antioxidants/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Liver/pathology , Ubiquinone/physiology
5.
Front Physiol ; 8: 525, 2017.
Article in English | MEDLINE | ID: mdl-28790927

ABSTRACT

Coenzyme Q (CoQ) is a lipid present in all cell membranes. One of the multiple metabolic functions of CoQ is to transport electrons in the reaction catalyzed by sulfide:quinone oxidoreductase (SQOR), the first enzyme of the oxidation pathway of sulfides (hydrogen sulfide, H2S). Early evidence of a defect in the metabolism of H2S in primary CoQ deficiency came from yeast studies in Schizosaccharomyces pombe strains defective for dps1 and ppt1 (homologs of PDSS1 and COQ2, respectively), which have H2S accumulation. Our recent studies in human skin fibroblasts and in murine models of primary CoQ deficiency show that, also in mammals, decreased CoQ levels cause impairment of H2S oxidation. Patient fibroblasts carrying different mutations in genes encoding proteins involved in CoQ biosynthesis show reduced SQOR activity and protein levels proportional to the levels of CoQ. In Pdss2kd/kd mice, kidney, the only organ clinically affected, shows reduced SQOR levels and downstream enzymes, accumulation of H2S, and glutathione depletion. Pdss2kd/kd mice have also low levels of thiosulfate in plasma and urine, and increased C4-C6 acylcarnitines in blood, due to inhibition of short-chain acyl-CoA dehydrogenase. Also in Coq9R239X mice, the symptomatic organ, cerebrum, shows accumulation of H2S, reduced SQOR, increase in thiosulfate sulfurtransferase and sulfite oxidase, and reduction in the levels of glutathione and glutathione enzymes, leading to alteration of the biosynthetic pathways of glutamate, serotonin, and catecholamines. Coq9R239X mice have also reduced blood pressure, possible consequence of H2S-induced vasorelaxation. Since liver is not clinically affected in Pdss2 and Coq9 mutant mice, the effects of the impairment of H2S oxidation in this organ were not investigated, despite its critical role in metabolism. In conclusion, in vitro and in vivo studies of CoQ deficient models provide evidence of tissue-specific H2S oxidation impairment, an additional pathomechanism that should be considered in the understanding and treatment of primary CoQ deficiency.

6.
Sci Rep ; 7: 45076, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28345620

ABSTRACT

Mitochondria are key cellular signaling platforms, affecting fundamental processes such as cell proliferation, differentiation and death. However, it remains unclear how mitochondrial signaling affects other organelles, particularly lysosomes. Here, we demonstrate that mitochondrial respiratory chain (RC) impairments elicit a stress signaling pathway that regulates lysosomal biogenesis via the microphtalmia transcription factor family. Interestingly, the effect of mitochondrial stress over lysosomal biogenesis depends on the timeframe of the stress elicited: while RC inhibition with rotenone or uncoupling with CCCP initially triggers lysosomal biogenesis, the effect peaks after few hours and returns to baseline. Long-term RC inhibition by long-term treatment with rotenone, or patient mutations in fibroblasts and in a mouse model result in repression of lysosomal biogenesis. The induction of lysosomal biogenesis by short-term mitochondrial stress is dependent on TFEB and MITF, requires AMPK signaling and is independent of calcineurin signaling. These results reveal an integrated view of how mitochondrial signaling affects lysosomes, which is essential to fully comprehend the consequences of mitochondrial malfunction, particularly in the context of mitochondrial diseases.


Subject(s)
Electron Transport , Lysosomes/metabolism , Mitochondrial Diseases/metabolism , Organelle Biogenesis , AMP-Activated Protein Kinase Kinases , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Microphthalmia-Associated Transcription Factor/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Protein Kinases/metabolism , Rotenone/pharmacology , Uncoupling Agents/pharmacology
7.
EMBO Mol Med ; 9(1): 78-95, 2017 01.
Article in English | MEDLINE | ID: mdl-27856619

ABSTRACT

Coenzyme Q (CoQ) is a key component of the mitochondrial respiratory chain, but it also has several other functions in the cellular metabolism. One of them is to function as an electron carrier in the reaction catalyzed by sulfide:quinone oxidoreductase (SQR), which catalyzes the first reaction in the hydrogen sulfide oxidation pathway. Therefore, SQR may be affected by CoQ deficiency. Using human skin fibroblasts and two mouse models with primary CoQ deficiency, we demonstrate that severe CoQ deficiency causes a reduction in SQR levels and activity, which leads to an alteration of mitochondrial sulfide metabolism. In cerebrum of Coq9R239X mice, the deficit in SQR induces an increase in thiosulfate sulfurtransferase and sulfite oxidase, as well as modifications in the levels of thiols. As a result, biosynthetic pathways of glutamate, serotonin, and catecholamines were altered in the cerebrum, and the blood pressure was reduced. Therefore, this study reveals the reduction in SQR activity as one of the pathomechanisms associated with CoQ deficiency syndrome.


Subject(s)
Ataxia/physiopathology , Mitochondria/metabolism , Mitochondrial Diseases/physiopathology , Muscle Weakness/physiopathology , Quinone Reductases/metabolism , Sulfides/metabolism , Ubiquinone/deficiency , Animals , Blood Pressure , Cells, Cultured , Cerebrum/physiopathology , Disease Models, Animal , Fibroblasts/metabolism , Humans , Mice , Oxidation-Reduction
8.
Mitochondrion ; 27: 56-63, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26748191

ABSTRACT

Analysis of mitochondrial function is crucial to understand their involvement in a given disease. High-resolution respirometry of permeabilized myocardial fibers in septic mice allows the evaluation of the bioenergetic system, maintaining mitochondrial ultrastructure and intracellular interactions, which are critical for an adequate functionality. OXPHOS and electron transport system (ETS) capacities were assessed using different substrate combinations. Our findings show a severe septic-dependent impairment in OXPHOS and ETS capacities with mitochondrial uncoupling at early and late phases of sepsis. Moreover, sepsis triggers complex III (CIII)-linked alterations in supercomplexes structure, and loss of mitochondrial density. In these conditions, melatonin administration to septic mice prevented sepsis-dependent mitochondrial injury in mitochondrial respiration. Likewise, melatonin improved cytochrome b content and ameliorated the assembly of CIII in supercomplexes. These results support the use of permeabilized fibers to identify properly the respiratory deficits and specific melatonin effects in sepsis.


Subject(s)
Antioxidants/administration & dosage , Melatonin/administration & dosage , Mitochondria, Heart/metabolism , Myocardium/metabolism , Myofibrils/metabolism , Sepsis/drug therapy , Sepsis/physiopathology , Animals , Disease Models, Animal , Electron Transport , Electron Transport Complex III/metabolism , Male , Mice, Inbred C57BL , Oxidative Phosphorylation
9.
EMBO Mol Med ; 7(5): 670-87, 2015 May.
Article in English | MEDLINE | ID: mdl-25802402

ABSTRACT

Primary coenzyme Q10 (CoQ10) deficiency is due to mutations in genes involved in CoQ biosynthesis. The disease has been associated with five major phenotypes, but a genotype-phenotype correlation is unclear. Here, we compare two mouse models with a genetic modification in Coq9 gene (Coq9(Q95X) and Coq9(R239X)), and their responses to 2,4-dihydroxybenzoic acid (2,4-diHB). Coq9(R239X) mice manifest severe widespread CoQ deficiency associated with fatal encephalomyopathy and respond to 2,4-diHB increasing CoQ levels. In contrast, Coq9(Q95X) mice exhibit mild CoQ deficiency manifesting with reduction in CI+III activity and mitochondrial respiration in skeletal muscle, and late-onset mild mitochondrial myopathy, which does not respond to 2,4-diHB. We show that these differences are due to the levels of COQ biosynthetic proteins, suggesting that the presence of a truncated version of COQ9 protein in Coq9(R239X) mice destabilizes the CoQ multiprotein complex. Our study points out the importance of the multiprotein complex for CoQ biosynthesis in mammals, which may provide new insights to understand the genotype-phenotype heterogeneity associated with human CoQ deficiency and may have a potential impact on the treatment of this mitochondrial disorder.


Subject(s)
Ataxia/pathology , Genetic Variation , Genotype , Mitochondrial Diseases/pathology , Muscle Weakness/pathology , Ubiquinone/deficiency , Animals , Disease Models, Animal , Hydroxybenzoates/administration & dosage , Hydroxybenzoates/toxicity , Mammals , Mice , Mice, Transgenic , Mutation, Missense , Ubiquinone/genetics
10.
J Pineal Res ; 58(1): 34-49, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25388914

ABSTRACT

Mucositis is a common and distressing side effect of chemotherapy or radiotherapy that has potentially severe consequences, and no treatment is available. The purpose of this study was to analyze the molecular pathways involved in the development of oral mucositis and to evaluate whether melatonin can prevent this pathology. The tongue of male Wistar rats was subjected to irradiation (X-ray YXLON Y.Tu 320-D03 irradiator; the animals received a dose of 7.5 Gy/day for 5 days). Rats were treated with 45 mg/day melatonin or vehicle for 21 days postirradiation, either by local application into their mouths (melatonin gel) or by subcutaneous injection. A connection between reactive oxygen species, generating mitochondria and the NLRP3 (NLR-related protein 3 nucleotide-binding domain leucine-rich repeat containing receptor-related protein 3) inflammasome, has been reported in mucositis. Here, we show that mitochondrial oxidative stress, bioenergetic impairment and subsequent NLRP3 inflammasome activation are involved in the development of oral mucositis after irradiation and that melatonin synthesized in the rat tongue is depleted after irradiation. The application of melatonin gel restores physiological melatonin levels in the tongue and prevents mucosal disruption and ulcer formation. Melatonin gel protects the mitochondria from radiation damage and blunts the NF-κB/NLRP3 inflammasome signaling activation in the tongue. Our results suggest new molecular pathways involved in radiotherapy-induced mucositis that are inhibited by topical melatonin application, suggesting a potential preventive therapy for mucositis in patients with cancer.


Subject(s)
Antioxidants/pharmacology , Melatonin/pharmacology , Mitochondria/metabolism , Radiation Injuries, Experimental/prevention & control , Receptors, Cytoplasmic and Nuclear/metabolism , Stomatitis/prevention & control , Animals , Carrier Proteins , Inflammasomes/metabolism , Male , Mitochondria/pathology , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/radiation effects , Stomatitis/metabolism , Stomatitis/pathology , Tongue/metabolism , Tongue/pathology , X-Rays/adverse effects
11.
Life Sci ; 121: 158-65, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25498899

ABSTRACT

AIMS: Previous data showed that melatonin maintains liver mitochondrial homeostasis during sepsis, but neither the mechanisms underlying mitochondrial dysfunction nor the target of melatonin are known. MAIN METHODS: Here, we analyzed mitochondrial respiration in isolated mouse liver mitochondria with different substrate combinations (glutamate/malate, glutamate/malate/sucinate or succinate/rotenone) to identify mitochondrial defects and melatonin targets during sepsis. Other bioenergetic parameters including a + a3, b, and c + c1 content, mitochondrial mass, and mitochondrial supercomplexes formation were analyzed. Mitochondrial function was assessed during experimental sepsis induced by cecal ligation and puncture (CLP) in livers of 3 mo. C57BL/6 mice at early and late phases of sepsis, i.e., at 8 and 24 h after sepsis induction. KEY FINDINGS: Septic mice showed mitochondrial injury with a decrease in state 3, respiratory control rate, mitochondrial mass, and cytochrome b and c + c1 content, which was prevented by melatonin treatment. Mitochondrial dysfunction in sepsis was mainly linked to complex I damage, because complex II was far less impaired. These mitochondria preserved the respiratory supramolecular organization, maintaining their electron transport system capacity. SIGNIFICANCE: This work strengthens the use of substrate combinations to identify specific respiratory defects and selective melatonin actions in septic mitochondria. Targeting mitochondrial complex I should be a main therapeutical approach in the treatment of sepsis, whereas the use of melatonin should be considered in the therapy of clinical sepsis.


Subject(s)
Liver/metabolism , Mitochondrial Diseases/diagnosis , Receptors, Melatonin/drug effects , Sepsis/diagnosis , Animals , Citrate (si)-Synthase/metabolism , Cytochromes/metabolism , Liver/physiopathology , Male , Mice , Mice, Inbred C57BL , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Mitochondrial Diseases/physiopathology , Mitochondrial Membranes/drug effects , Oxygen Consumption/drug effects , Sepsis/metabolism , Sepsis/physiopathology , Spirometry
12.
Proc Natl Acad Sci U S A ; 111(44): E4697-705, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25339443

ABSTRACT

Coenzyme Q (CoQ) is an isoprenylated quinone that is essential for cellular respiration and is synthesized in mitochondria by the combined action of at least nine proteins (COQ1-9). Although most COQ proteins are known to catalyze modifications to CoQ precursors, the biochemical role of COQ9 remains unclear. Here, we report that a disease-related COQ9 mutation leads to extensive disruption of the CoQ protein biosynthetic complex in a mouse model, and that COQ9 specifically interacts with COQ7 through a series of conserved residues. Toward understanding how COQ9 can perform these functions, we solved the crystal structure of Homo sapiens COQ9 at 2.4 Å. Unexpectedly, our structure reveals that COQ9 has structural homology to the TFR family of bacterial transcriptional regulators, but that it adopts an atypical TFR dimer orientation and is not predicted to bind DNA. Our structure also reveals a lipid-binding site, and mass spectrometry-based analyses of purified COQ9 demonstrate that it associates with multiple lipid species, including CoQ itself. The conserved COQ9 residues necessary for its interaction with COQ7 comprise a surface patch around the lipid-binding site, suggesting that COQ9 might serve to present its bound lipid to COQ7. Collectively, our data define COQ9 as the first, to our knowledge, mammalian TFR structural homolog and suggest that its lipid-binding capacity and association with COQ7 are key features for enabling CoQ biosynthesis.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Lipid Metabolism/physiology , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Ubiquinone/biosynthesis , Animals , Carrier Proteins/genetics , Crystallography, X-Ray , Humans , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Mitochondrial Proteins/genetics , Mixed Function Oxygenases , Protein Structure, Tertiary , Ubiquinone/genetics
13.
Mol Syndromol ; 5(3-4): 163-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25126049

ABSTRACT

Primary coenzyme Q10 (CoQ10) deficiency is a rare mitochondrial disorder associated with 5 major clinical phenotypes: (1) encephalomyopathy, (2) severe infantile multisystemic disease, (3) cerebellar ataxia, (4) isolated myopathy, and (5) steroid-resistant nephrotic syndrome. Growth retardation, deafness and hearing loss have also been described in CoQ10-deficient patients. This heterogeneity in the clinical presentations suggests that multiple pathomechanisms may exist. To investigate the biochemical and molecular consequences of CoQ10 deficiency, different laboratories have studied cultures of skin fibroblasts from patients with CoQ10 deficiency. In this review, we summarize the results obtained in these studies over the last decade.

14.
Biochim Biophys Acta ; 1842(7): 893-901, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24576561

ABSTRACT

Coenzyme Q10 (CoQ10) deficiency (MIM 607426) causes a mitochondrial syndrome with variability in the clinical presentations. Patients with CoQ10 deficiency show inconsistent responses to oral ubiquinone-10 supplementation, with the highest percentage of unsuccessful results in patients with neurological symptoms (encephalopathy, cerebellar ataxia or multisystemic disease). Failure in the ubiquinone-10 treatment may be the result of its poor absorption and bioavailability, which may be improved by using different pharmacological formulations. In a mouse model (Coq9(X/X)) of mitochondrial encephalopathy due to CoQ deficiency, we have evaluated oral supplementation with water-soluble formulations of reduced (ubiquinol-10) and oxidized (ubiquinone-10) forms of CoQ10. Our results show that CoQ10 was increased in all tissues after supplementation with ubiquinone-10 or ubiquinol-10, with the tissue levels of CoQ10 with ubiquinol-10 being higher than with ubiquinone-10. Moreover, only ubiquinol-10 was able to increase the levels of CoQ10 in mitochondria from cerebrum of Coq9(X/X) mice. Consequently, ubiquinol-10 was more efficient than ubiquinone-10 in increasing the animal body weight and CoQ-dependent respiratory chain complex activities, and reducing the vacuolization, astrogliosis and oxidative damage in diencephalon, septum-striatum and, to a lesser extent, in brainstem. These results suggest that water-soluble formulations of ubiquinol-10 may improve the efficacy of CoQ10 therapy in primary and secondary CoQ10 deficiencies, other mitochondrial diseases and neurodegenerative diseases.


Subject(s)
Ataxia/drug therapy , Mitochondria/drug effects , Mitochondrial Diseases/drug therapy , Mitochondrial Encephalomyopathies/drug therapy , Muscle Weakness/drug therapy , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Animals , Brain Diseases/drug therapy , Brain Stem/drug effects , Corpus Striatum/drug effects , Electron Transport/drug effects , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Ubiquinone/pharmacology
15.
J Pineal Res ; 56(1): 71-81, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24117944

ABSTRACT

While it is accepted that the high production of nitric oxide (NO˙) by the inducible nitric oxide synthase (iNOS) impairs cardiac mitochondrial function during sepsis, the role of neuronal nitric oxide synthase (nNOS) may be protective. During sepsis, there is a significantly increase in the expression and activity of mitochondrial iNOS (i-mtNOS), which parallels the changes in cytosolic iNOS. The existence of a constitutive NOS form (c-mtNOS) in heart mitochondria has been also described, but its role in the heart failure during sepsis remains unclear. Herein, we analyzed the changes in mitochondrial oxidative stress and bioenergetics in wild-type and nNOS-deficient mice during sepsis, and the role of melatonin, a known antioxidant, in these changes. Sepsis was induced by cecal ligation and puncture, and heart mitochondria were analyzed for NOS expression and activity, nitrites, lipid peroxidation, glutathione and glutathione redox enzymes, oxidized proteins, and respiratory chain activity in vehicle- and melatonin-treated mice. Our data show that sepsis produced a similar induction of iNOS/i-mtNOS and comparable inhibition of the respiratory chain activity in wild-type and in nNOS-deficient mice. Sepsis also increased mitochondrial oxidative/nitrosative stress to a similar extent in both mice strains. Melatonin administration inhibited iNOS/i-mtNOS induction, restored mitochondrial homeostasis in septic mice, and preserved the activity of nNOS/c-mtNOS. The effects of melatonin were unrelated to the presence or the absence of nNOS. Our observations show a lack of effect of nNOS on heart bioenergetic impairment during sepsis and further support the beneficial actions of melatonin in sepsis.


Subject(s)
Melatonin/pharmacology , Mitochondria/drug effects , Myocardium/cytology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type I/metabolism , Sepsis/metabolism , Analysis of Variance , Animals , Antioxidants/pharmacology , Cytosol/chemistry , Cytosol/drug effects , Cytosol/metabolism , Disease Models, Animal , Glutathione/analysis , Glutathione/metabolism , Lipid Peroxidation/drug effects , Mice , Mice, Knockout , Mitochondria/chemistry , Mitochondria/metabolism , Mitochondrial Proteins/analysis , Mitochondrial Proteins/metabolism , Nitric Oxide , Nitric Oxide Synthase Type I/analysis , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type II/analysis , Nitric Oxide Synthase Type II/genetics , Oxidative Stress/drug effects
16.
Nat Prod Commun ; 8(1): 47-50, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23472457

ABSTRACT

The powerful antioxidant capacity of virgin argan oil is attributed to its content of antioxidant molecules. Recent investigations have identified CoQ10 and melatonin as some of these antioxidant molecules. In this review, we summarize the most recent data about the content of CoQ10 and melatonin in virgin argan oil and the differences found in samples extracted by the traditional and half-industrialized methods. We also emphasize the importance of these two molecules for human health, focusing on their actions in mitochondria. Finally, we refer to other abundant antioxidants in virgin argan oil: tocopherols and polyphenols.


Subject(s)
Antioxidants/pharmacology , Melatonin/pharmacology , Mitochondria/drug effects , Plant Oils/chemistry , Ubiquinone/analogs & derivatives , Antioxidants/analysis , Homeostasis/drug effects , Humans , Melatonin/analysis , Mitochondria/metabolism , Plant Oils/pharmacology , Reactive Oxygen Species/metabolism , Ubiquinone/analysis , Ubiquinone/pharmacology
17.
Hum Mol Genet ; 22(6): 1233-48, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23255162

ABSTRACT

Coenzyme Q10 (CoQ(10)) or ubiquinone is a well-known component of the mitochondrial respiratory chain. In humans, CoQ(10) deficiency causes a mitochondrial syndrome with an unexplained variability in the clinical presentations. To try to understand this heterogeneity in the clinical phenotypes, we have generated a Coq9 Knockin (R239X) mouse model. The lack of a functional Coq9 protein in homozygous Coq9 mutant (Coq9(X/X)) mice causes a severe reduction in the Coq7 protein and, as consequence, a widespread CoQ deficiency and accumulation of demethoxyubiquinone. The deficit in CoQ induces a brain-specific impairment of mitochondrial bioenergetics performance, a reduction in respiratory control ratio, ATP levels and ATP/ADP ratio and specific loss of respiratory complex I. These effects lead to neuronal death and demyelinization with severe vacuolization and astrogliosis in the brain of Coq9(X/X) mice that consequently die between 3 and 6 months of age. These results suggest that the instability of mitochondrial complex I in the brain, as a primary event, triggers the development of mitochondrial encephalomyopathy associated with CoQ deficiency.


Subject(s)
Mitochondrial Encephalomyopathies/enzymology , Ubiquinone/deficiency , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Encephalomyopathies/genetics , Ubiquinone/genetics , Ubiquinone/metabolism
18.
Horm Mol Biol Clin Investig ; 16(2): 91-100, 2013 Dec.
Article in English | MEDLINE | ID: mdl-25436750

ABSTRACT

Abstract Mitochondrial dysfunction and oxidative/nitrosative stress are common features of senescence, and they explain some of the pathophysiological events during aging. In different animal models of aging, the existence of oxidative stress, inflammation, and mitochondrial dysfunction has been reported. There is no information, however, regarding the age when these symptoms begin and if they account for gender differences in aging. Here we analyzed oxidative/nitrosative stress markers and bioenergetics in the brain mitochondria of normal mice during the first 10 months of life, looking for early signs of senescence. Male and female mice were treated with vehicle or melatonin during the first 9 months of life, starting at weaning. Mice were sacrificed at 5 and 10 months of life, and pure brain mitochondria were prepared and assayed for respiratory chain activity, ATP production, and oxidative/nitrosative stress status. The results showed that the brain mitochondria from male mice have a better glutathione cycle than female mice, whereas female mice have higher electron transport chain activity and ATP production at 5 months old. Five months later, however, oxidative/nitrosative stress markers increased in both male and female mice, thus eliminating the differences between the genders. More importantly, these changes were prevented by chronic melatonin administration, which also restored the gender differences found in 5-month-old mice. Thus, melatonin administration as a single therapy can maintain the full function of the brain mitochondria during the early events of aging, a finding that has important consequences in the pathophysiology of brain senescence.

SELECTION OF CITATIONS
SEARCH DETAIL
...