Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Hum Mol Genet ; 29(19): 3296-3311, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32975579

ABSTRACT

Abnormalities of one carbon, glutathione and sulfide metabolisms have recently emerged as novel pathomechanisms in diseases with mitochondrial dysfunction. However, the mechanisms underlying these abnormalities are not clear. Also, we recently showed that sulfide oxidation is impaired in Coenzyme Q10 (CoQ10) deficiency. This finding leads us to hypothesize that the therapeutic effects of CoQ10, frequently administered to patients with primary or secondary mitochondrial dysfunction, might be due to its function as cofactor for sulfide:quinone oxidoreductase (SQOR), the first enzyme in the sulfide oxidation pathway. Here, using biased and unbiased approaches, we show that supraphysiological levels of CoQ10 induces an increase in the expression of SQOR in skin fibroblasts from control subjects and patients with mutations in Complex I subunits genes or CoQ biosynthetic genes. This increase of SQOR induces the downregulation of the cystathionine ß-synthase and cystathionine γ-lyase, two enzymes of the transsulfuration pathway, the subsequent downregulation of serine biosynthesis and the adaptation of other sulfide linked pathways, such as folate cycle, nucleotides metabolism and glutathione system. These metabolic changes are independent of the presence of sulfur aminoacids, are confirmed in mouse models, and are recapitulated by overexpression of SQOR, further proving that the metabolic effects of CoQ10 supplementation are mediated by the overexpression of SQOR. Our results contribute to a better understanding of how sulfide metabolism is integrated in one carbon metabolism and may explain some of the benefits of CoQ10 supplementation observed in mitochondrial diseases.


Subject(s)
Ataxia/pathology , Carbon/metabolism , Electron Transport Complex I/metabolism , Mitochondria/pathology , Mitochondrial Diseases/pathology , Muscle Weakness/pathology , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Sulfides/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Animals , Ataxia/genetics , Ataxia/metabolism , Electron Transport , Electron Transport Complex I/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Glutathione/metabolism , Humans , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Muscle Weakness/genetics , Muscle Weakness/metabolism , Oxidoreductases Acting on Sulfur Group Donors/genetics , Skin/drug effects , Skin/metabolism , Skin/pathology , Transcriptome , Ubiquinone/genetics , Ubiquinone/metabolism , Ubiquinone/pharmacology , Vitamins/pharmacology
2.
J Pineal Res ; 73(3): e12824, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35986493

ABSTRACT

The oncostatic effects of melatonin correlate with increased reactive oxygen species (ROS) levels, but how melatonin induces this ROS generation is unknown. In the present study, we aimed to elucidate the two seemingly opposing actions of melatonin regarding its relationship with free radicals. We analyzed the effects of melatonin on head and neck squamous cell carcinoma cell lines (Cal-27 and SCC-9), which were treated with 0.5 or 1 mM melatonin. We further examined the potential effects of melatonin to induce ROS and apoptosis in Cal-27 xenograft mice. Here we report that melatonin mediates apoptosis in head and neck cancer by driving mitochondrial reverse electron transport (RET) to induce ROS production. Melatonin-induced changes in tumoral metabolism led to increased mitochondrial activity, which, in turn, induced ROS-dependent mitochondrial uncoupling. Interestingly, mitochondrial complex inhibitors, including rotenone, abolished the ROS elevation indicating that melatonin increased ROS generation via RET. Melatonin also increased membrane potential and CoQ10 H2 /CoQ10 ratio to elevate mitochondrial ROS production, which are essential conditions for RET. We found that genetic manipulation of cancer cells with alternative oxidase, which transfers electrons from QH2 to oxygen, inhibited melatonin-induced ROS generation, and apoptosis. RET restored the melatonin-induced oncostatic effect, highlighting the importance of RET as the site of ROS production. These results illustrate that RET and ROS production are crucial factors in melatonin's effects in cancer cells and establish the dual effect of melatonin in protecting normal cells and inducing apoptosis in cancer cells.


Subject(s)
Head and Neck Neoplasms , Melatonin , Animals , Apoptosis , Electron Transport , Head and Neck Neoplasms/drug therapy , Humans , Melatonin/pharmacology , Mice , Reactive Oxygen Species/metabolism
3.
Environ Res ; 211: 113115, 2022 08.
Article in English | MEDLINE | ID: mdl-35292247

ABSTRACT

BACKGROUND: Numerous contemporary non-persistent pesticides may elicit neurodevelopmental impairments. Brain-derived neurotrophic factor (BDNF) has been proposed as a novel effect biomarker of neurological function that could help to understand the biological responses of some environmental exposures. OBJECTIVES: To investigate the relationship between exposure to various non-persistent pesticides, BDNF, and behavioral functioning among adolescents. METHODS: The concentrations of organophosphate (OP) insecticide metabolites 3,5,6-trichloro-2-pyridinol (TCPy), 2-isopropyl-4-methyl-6-hydroxypyrimidine (IMPy), malathion diacid (MDA), and diethyl thiophosphate (DETP); metabolites of pyrethroids 3-phenoxybenzoic acid (3-PBA) and dimethylcyclopropane carboxylic acid (DCCA), the metabolite of insecticide carbaryl 1-naphthol (1-N), and the metabolite of ethylene-bis-dithiocarbamate fungicides ethylene thiourea (ETU) were measured in spot urine samples, as well as serum BDNF protein levels and blood DNA methylation of Exon IV of BDNF gene in 15-17-year-old boys from the INMA-Granada cohort in Spain. Adolescents' behavior was reported by parents using the Child Behavior Check List (CBCL/6-18). This study included 140 adolescents of whom 118 had data on BDNF gene DNA methylation. Multivariable linear regression, weighted quantile sum (WQS) for mixture effects, and mediation models were fit. RESULTS: IMPy, MDA, DCCA, and ETU were detected in more than 70% of urine samples, DETP in 53%, and TCPy, 3-PBA, and 1-N in less than 50% of samples. Higher levels of IMPy, TCPy, and ETU were significantly associated with more behavioral problems as social, thought problems, and rule-breaking symptoms. IMPy, MDA, DETP, and 1-N were significantly associated with decreased serum BDNF levels, while MDA, 3-PBA, and ETU were associated with higher DNA methylation percentages at several CpGs. WQS models suggest a mixture effect on more behavioral problems and BDNF DNA methylation at several CpGs. A mediated effect of serum BDNF within IMPy-thought and IMPy-rule breaking associations was suggested. CONCLUSION: BDNF biomarkers measured at different levels of biological complexity provided novel information regarding the potential disruption of behavioral function due to contemporary pesticides, highlighting exposure to diazinon (IMPy) and the combined effect of IMPy, MDA, DCCA, and ETU. However, further research is warranted.


Subject(s)
Adolescent Behavior , Brain-Derived Neurotrophic Factor , Pesticides , Adolescent , Adolescent Behavior/drug effects , Biomarkers , Brain-Derived Neurotrophic Factor/genetics , Environmental Exposure/adverse effects , Ethylenes , Humans , Male , Organophosphorus Compounds/urine , Pesticides/toxicity , Pesticides/urine , Pyrethrins/urine
4.
Mol Genet Metab ; 123(3): 289-291, 2018 03.
Article in English | MEDLINE | ID: mdl-29246431

ABSTRACT

Primary disorders of the human coenzyme Q10 (CoQ10) biosynthesis pathway are a known cause of severe pediatric diseases. So far, oral administration of CoQ10 is the only treatment strategy for affected individuals. However, the real benefit of CoQ10 supplementation remains questionable and clinical studies regarding efficiency are lacking. Here we provide an outlook on novel treatment approaches using CoQ precursor compounds. These metabolic bypass strategies might be a promising alternative for oral CoQ10 supplementation regimens.


Subject(s)
Ataxia/drug therapy , Hydroxybenzoates/therapeutic use , Mitochondrial Diseases/drug therapy , Muscle Weakness/drug therapy , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Animals , Apoptosis/drug effects , Ataxia/genetics , Ataxia/pathology , Biosynthetic Pathways/drug effects , Biosynthetic Pathways/genetics , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Disease Models, Animal , Humans , Hydroxybenzoates/pharmacology , Mice , Mitochondria/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Muscle Weakness/genetics , Muscle Weakness/pathology , Pyrimidines/metabolism , Solubility , Treatment Outcome , Ubiquinone/biosynthesis , Ubiquinone/genetics , Ubiquinone/metabolism , Ubiquinone/therapeutic use , Vitamins/therapeutic use
5.
J Pineal Res ; 64(3)2018 Apr.
Article in English | MEDLINE | ID: mdl-29247557

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) clearly involves activation of the Akt mammalian target of rapamycin (mTOR) signalling pathway. However, the effectiveness of treatment with the mTOR inhibitor rapamycin is often limited by chemoresistance. Melatonin suppresses neoplastic growth via different mechanisms in a variety of tumours. In this study, we aimed to elucidate the effects of melatonin on rapamycin-induced HNSCC cell death and to identify potential cross-talk pathways. We analysed the dose-dependent effects of melatonin in rapamycin-treated HNSCC cell lines (Cal-27 and SCC-9). These cells were treated with 0.1, 0.5 or 1 mmol/L melatonin combined with 20 nM rapamycin. We further examined the potential synergistic effects of melatonin with rapamycin in Cal-27 xenograft mice. Relationships between inhibition of the mTOR pathway, reactive oxygen species (ROS), and apoptosis and mitophagy reportedly increased the cytotoxic effects of rapamycin in HNSCC. Our results demonstrated that combined treatment with rapamycin and melatonin blocked the negative feedback loop from the specific downstream effector of mTOR activation S6K1 to Akt signalling, which decreased cell viability, proliferation and clonogenic capacity. Interestingly, combined treatment with rapamycin and melatonin-induced changes in mitochondrial function, which were associated with increased ROS production, increasing apoptosis and mitophagy. This led to increase cell death and cellular differentiation. Our data further indicated that melatonin administration reduced rapamycin-associated toxicity to healthy cells. Overall, our findings suggested that melatonin could be used as an adjuvant agent with rapamycin, improving effectiveness while minimizing its side effects.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/pathology , Mitophagy/drug effects , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Melatonin/pharmacology , Mice , Mice, Nude , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Sirolimus/pharmacology , Squamous Cell Carcinoma of Head and Neck , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
6.
Cell Mol Life Sci ; 74(21): 3965-3987, 2017 11.
Article in English | MEDLINE | ID: mdl-28785808

ABSTRACT

After the characterization of the central pacemaker in the suprachiasmatic nucleus, the expression of clock genes was identified in several peripheral tissues including the immune system. The hierarchical control from the central clock to peripheral clocks extends to other functions including endocrine, metabolic, immune, and mitochondrial responses. Increasing evidence links the disruption of the clock genes expression with multiple diseases and aging. Chronodisruption is associated with alterations of the immune system, immunosenescence, impairment of energy metabolism, and reduction of pineal and extrapineal melatonin production. Regarding sepsis, a condition coursing with an exaggerated response of innate immunity, experimental and clinical data showed an alteration of circadian rhythms that reflects the loss of the normal oscillation of the clock. Moreover, recent data point to that some mediators of the immune system affects the normal function of the clock. Under specific conditions, this control disappears reactivating the immune response. So, it seems that clock gene disruption favors the innate immune response, which in turn induces the expression of proinflammatory mediators, causing a further alteration of the clock. Here, the clock control of the mitochondrial function turns off, leading to a bioenergetic decay and formation of reactive oxygen species that, in turn, activate the inflammasome. This arm of the innate immunity is responsible for the huge increase of interleukin-1ß and entrance into a vicious cycle that could lead to the death of the patient. The broken clock is recovered by melatonin administration, that is accompanied by the normalization of the innate immunity and mitochondrial homeostasis. Thus, this review emphasizes the connection between clock genes, innate immunity and mitochondria in health and sepsis, and the role of melatonin to maintain clock homeostasis.


Subject(s)
Antioxidants/pharmacology , CLOCK Proteins/metabolism , Circadian Rhythm/physiology , Melatonin/pharmacology , Mitochondria/metabolism , Sepsis/prevention & control , Animals , CLOCK Proteins/genetics , Humans , Mitochondria/drug effects , Sepsis/genetics , Sepsis/metabolism
7.
Hum Mol Genet ; 24(16): 4516-29, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25976310

ABSTRACT

Ataxia oculomotor apraxia type 1 (AOA1) is an autosomal recessive disease caused by mutations in APTX, which encodes the DNA strand-break repair protein aprataxin (APTX). CoQ10 deficiency has been identified in fibroblasts and muscle of AOA1 patients carrying the common W279X mutation, and aprataxin has been localized to mitochondria in neuroblastoma cells, where it enhances preservation of mitochondrial function. In this study, we show that aprataxin deficiency impairs mitochondrial function, independent of its role in mitochondrial DNA repair. The bioenergetics defect in AOA1-mutant fibroblasts and APTX-depleted Hela cells is caused by decreased expression of SDHA and genes encoding CoQ biosynthetic enzymes, in association with reductions of APE1, NRF1 and NRF2. The biochemical and molecular abnormalities in APTX-depleted cells are recapitulated by knockdown of APE1 in Hela cells and are rescued by overexpression of NRF1/2. Importantly, pharmacological upregulation of NRF1 alone by 5-aminoimidazone-4-carboxamide ribonucleotide does not rescue the phenotype, which, in contrast, is reversed by the upregulation of NRF2 by rosiglitazone. Accordingly, we propose that the lack of aprataxin causes reduction of the pathway APE1/NRF1/NRF2 and their target genes. Our findings demonstrate a critical role of APTX in transcription regulation of mitochondrial function and the pathogenesis of AOA1 via a novel pathomechanistic pathway, which may be relevant to other neurodegenerative diseases.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , DNA-Binding Proteins/deficiency , Down-Regulation , Fibroblasts/metabolism , Mitochondria/metabolism , NF-E2-Related Factor 2/biosynthesis , Nuclear Proteins/deficiency , Nuclear Respiratory Factor 1/biosynthesis , Signal Transduction , Ataxia/genetics , Ataxia/metabolism , Ataxia/pathology , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-Binding Proteins/genetics , Female , Fibroblasts/pathology , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Humans , Male , Mitochondria/pathology , NF-E2-Related Factor 2/genetics , Nuclear Proteins/genetics , Nuclear Respiratory Factor 1/genetics
8.
Mol Genet Metab ; 121(3): 216-223, 2017 07.
Article in English | MEDLINE | ID: mdl-28552678

ABSTRACT

Coenzyme Q10 (CoQ10) is an essential cofactor of the mitochondrial oxidative phosphorylation (OXPHOS) system and its deficiency has important implications for several inherited metabolic disorders of childhood. The biosynthesis of CoQ10 is a complicated process, which involves at least 12 different enzymes. One of the metabolic intermediates that are formed during CoQ10 biosynthesis is the molecule 6-demethoxyubiquinone (6-DMQ). This CoQ precursor is processed at the level of COQ7 and COQ9. We selected this metabolite as a marker substance for metabolic analysis of cell lines with inherited genetic defects (COQ2, COQ4, COQ7 and COQ9) or siRNA knockdown in CoQ biosynthesis enzymes using ultra-performance liquid chromatography coupled to tandem mass spectrometry (UPLC-MS/MS). In COQ4, COQ7 and COQ9 deficient cell lines, we detected significantly elevated levels of 6-DMQ. This suggests a functional interplay of these proteins. However, additional siRNA studies demonstrated that elevated 6-DMQ levels are not an exclusive marker of the COQ7/COQ9 enzymatic step of CoQ10 biosynthesis but constitute a more general phenomenon that occurs in disorders impairing the function or stability of the CoQ-synthome. To further investigate the interdependence of CoQ10 biosynthesis enzyme expression, we performed immunoblotting in various cell lines with CoQ10 deficiency, indicating that COQ4, COQ7 and COQ9 protein expression levels are highly regulated depending on the underlying defect. Supplementation of cell lines with synthetic CoQ precursor compounds demonstrated beneficial effects of 2,4-dihydroxybenzoic acid in COQ7 and COQ9 deficiency. Moreover, vanillic acid selectively stimulated CoQ10 biosynthesis and improved cell viability in COQ9 deficiency. However, compounds tested in this study failed to rescue COQ4 deficiency.


Subject(s)
Ataxia/metabolism , Mitochondrial Diseases/metabolism , Muscle Weakness/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Cell Line , Cell Survival/drug effects , Fibroblasts/drug effects , Humans , Hydroxybenzoates/pharmacology , Mitochondria/metabolism , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidative Phosphorylation , Tandem Mass Spectrometry , Ubiquinone/biosynthesis , Ubiquinone/metabolism , Vanillic Acid/pharmacology
9.
J Pineal Res ; 63(1)2017 Aug.
Article in English | MEDLINE | ID: mdl-28370493

ABSTRACT

The NLRP3 inflammasome is involved in the innate immune response during inflammation. Moreover, melatonin blunts the NF-κB/NLRP3 connection during sepsis. Thus, we compared the roles of the NLRP3 inflammasome and/or melatonin treatment in the septic response of wild-type and NLRP3-/- mice. Mouse myocardial tissue was used for this purpose. The nuclear turnover of NF-κB was enhanced during sepsis, with an increase in TNFα, iNOS, and pro-IL-1ß. The lack of inflammasome in NLRP3-/- mice significantly reduced that response and blunted IL-1ß maturation due to the lack of caspase-1. Clock and Bmal1 did not change in both mouse strains, enhancing Chrono expression in mutants. RORα, which positively regulates Bmal1, was enhanced at a similar extend in both mouse strains, whereas the expression of the Bmal1 repressor, Rev-Erbα, increased in WT but was depressed in NLRP3-/- mice. Nampt, transcriptionally controlled by Bmal1, increased in WT mice together with Sirt1, whereas they remained unchanged in NLRP3-/- mice. Melatonin treatment reduced the septic response in a comparable manner as did the lack of NLRP3, but unlike the latter, it normalized the clock genes turnover through the induction of RORα and repression of Rev-Erbα and Per2, leading to enhanced Nampt and Sirt1. The lack of NLRP3 inflammasome converts sepsis to a moderate inflammatory disease and identifies NLRP3 as a main target for the treatment of sepsis. The efficacy of melatonin in counteracting the NLRP3 inflammasome activation further confirms the indoleamine as a useful therapeutic drug against this serious condition.


Subject(s)
Inflammasomes/drug effects , Melatonin/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sepsis/metabolism , Signal Transduction/drug effects , Animals , Female , Heart/drug effects , Inflammasomes/metabolism , Mice , Mice, Inbred C57BL , Mutation , Myocardium/cytology , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
10.
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
11.
FASEB J ; 29(9): 3863-75, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26045547

ABSTRACT

We determined the NF-κB- and NOD-like receptor (NLR)P3-dependent molecular mechanisms involved in sepsis and evaluated the role of retinoid-related orphan receptor (ROR)-α in melatonin's anti-inflammatory actions. Western blot, RT-PCR, ELISA, and spectrophotometric analysis revealed that NF-κB and NLRP3 closely interact, leading to proinflammatory and pro-oxidant status in heart tissue of septic C57BL/6J mice. Moreover, mitochondrial oxygen consumption was reduced by 80% in septic mice. In vivo and in vitro analysis showed that melatonin administration blunts NF-κB transcriptional activity through a sirtuin1-dependent NF-κB deacetylation in septic mice. Melatonin also decreased NF-κB-dependent proinflammatory response and restored redox balance and mitochondrial homeostasis, thus inhibiting the NLRP3 inflammasome. In an important finding, the inhibition of NF-κB by melatonin, but not that of NLRP3, was blunted in RORα (sg/sg) mice, indicating that functional RORα transcription factor is necessary for the initiation of the innate immune response against inflammation. Our results are evidence of the NF-κB/NLRP3 connection during sepsis and identify NLRP3 as a novel molecular target for melatonin. The multiple molecular targets of melatonin in this study explain its potent anti-inflammatory efficacy against systemic innate immune activation and herald a promising therapeutic application for melatonin in the treatment of sepsis.


Subject(s)
Carrier Proteins/metabolism , Immunity, Innate/drug effects , Melatonin/pharmacology , NF-kappa B/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Sepsis/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , Immunity, Innate/genetics , Mice , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/immunology , Oxidation-Reduction/drug effects , Sepsis/genetics , Sepsis/immunology , Sepsis/pathology
12.
J Pineal Res ; 61(1): 96-107, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27064726

ABSTRACT

Multiple studies reporting mitochondrial impairment in Parkinson's disease (PD) involve knockout or knockdown models to inhibit the expression of mitochondrial-related genes, including parkin, PINK1, and DJ-1 ones. Melatonin has significant neuroprotective properties, which have been related to its ability to boost mitochondrial bioenergetics. The meaning and molecular targets of melatonin in PD are yet unclear. Zebrafish are an outstanding model of PD because they are vertebrates, their dopaminergic system is comparable to the nigrostriatal system of humans, and their brains express the same genes as mammals. The exposure of 24 hpf zebrafish embryos to MPTP leads to a significant inhibition of the mitochondrial complex I and the induction of sncga gene, responsible for enhancing γ-synuclein accumulation, which is related to mitochondrial dysfunction. Moreover, MPTP inhibited the parkin/PINK1/DJ-1 expression, impeding the normal function of the parkin/PINK1/DJ-1/MUL1 network to remove the damaged mitochondria. This situation remains over time, and removing MPTP from the treatment did not stop the neurodegenerative process. On the contrary, mitochondria become worse during the next 2 days without MPTP, and the embryos developed a severe motor impairment that cannot be rescued because the mitochondrial-related gene expression remained inhibited. Melatonin, added together with MPTP or added once MPTP was removed, prevented and recovered, respectively, the parkinsonian phenotype once it was established, restoring gene expression and normal function of the parkin/PINK1/DJ-1/MUL1 loop and also the normal motor activity of the embryos. The results show, for the first time, that melatonin restores brain function in zebrafish suffering with Parkinson-like disease.


Subject(s)
Embryo, Nonmammalian/metabolism , MPTP Poisoning/drug therapy , Melatonin/pharmacology , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , MPTP Poisoning/genetics , MPTP Poisoning/metabolism , Nerve Tissue Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Protein Ligases/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
13.
J Pineal Res ; 60(2): 193-205, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26681113

ABSTRACT

The connection between the innate immune system, clock genes, and mitochondrial bioenergetics was analyzed during aging and sepsis in mouse heart. Our results suggest that the sole NF-κB activation does not explain the inflammatory process underlying aging; the former also triggers the NLRP3 inflammasome that enhances caspase-1-dependent maturation of IL-1ß. In this way, aged mice enter into a vicious cycle as IL-1ß further activates the NF-κB/NLRP3 inflammasome link. The origin of NF-κB activation was related to the age-dependent Bmal1/Clock/RORα/Rev-Erbα loop disruption, which lowers NAD(+) levels, reducing the SIRT1 deacetylase ability to inactivate NF-κB. Consequently, NF-κB binding to DNA increases, raising the formation of proinflammatory mediators and inducing mitochondrial impairment. The cycle is then closed with the subsequent NLRP3 inflammasome activation. This paired contribution of the innate immune pathways serves as a catalyst to magnify the response to sepsis in aged compared with young mice. Melatonin administration blunted the septic response, reducing inflammation and oxidative stress, and enhancing mitochondrial function at the levels of nonseptic aged mice, but it did not counteract the age-related inflammation. Together, our results suggest that, although with different strengths, chronoinflammaging constitutes the biochemical substrate of aging and sepsis, and identifies the NLRP3 inflammasome as a new molecular target for melatonin, providing a rationale for its use in NLRP3-dependent diseases.


Subject(s)
Aging/metabolism , Carrier Proteins/biosynthesis , Gene Expression Regulation/drug effects , Inflammasomes/metabolism , Melatonin/pharmacology , NF-kappa B/metabolism , Sepsis/drug therapy , Aging/drug effects , Aging/pathology , Animals , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Sepsis/metabolism , Sepsis/pathology
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.
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
16.
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
17.
Cell Mol Life Sci ; 71(16): 2997-3025, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24554058

ABSTRACT

Endogenous melatonin is synthesized from tryptophan via 5-hydroxytryptamine. It is considered an indoleamine from a biochemical point of view because the melatonin molecule contains a substituted indolic ring with an amino group. The circadian production of melatonin by the pineal gland explains its chronobiotic influence on organismal activity, including the endocrine and non-endocrine rhythms. Other functions of melatonin, including its antioxidant and anti-inflammatory properties, its genomic effects, and its capacity to modulate mitochondrial homeostasis, are linked to the redox status of cells and tissues. With the aid of specific melatonin antibodies, the presence of melatonin has been detected in multiple extrapineal tissues including the brain, retina, lens, cochlea, Harderian gland, airway epithelium, skin, gastrointestinal tract, liver, kidney, thyroid, pancreas, thymus, spleen, immune system cells, carotid body, reproductive tract, and endothelial cells. In most of these tissues, the melatonin-synthesizing enzymes have been identified. Melatonin is present in essentially all biological fluids including cerebrospinal fluid, saliva, bile, synovial fluid, amniotic fluid, and breast milk. In several of these fluids, melatonin concentrations exceed those in the blood. The importance of the continual availability of melatonin at the cellular level is important for its physiological regulation of cell homeostasis, and may be relevant to its therapeutic applications. Because of this, it is essential to compile information related to its peripheral production and regulation of this ubiquitously acting indoleamine. Thus, this review emphasizes the presence of melatonin in extrapineal organs, tissues, and fluids of mammals including humans.


Subject(s)
Antioxidants/analysis , Antioxidants/metabolism , Central Nervous System Depressants/analysis , Central Nervous System Depressants/metabolism , Melatonin/analysis , Melatonin/metabolism , Animals , Antioxidants/adverse effects , Antioxidants/therapeutic use , Central Nervous System Depressants/adverse effects , Central Nervous System Depressants/therapeutic use , Cytoprotection/drug effects , Homeostasis/drug effects , Humans , Melatonin/adverse effects , Melatonin/therapeutic use , Oxidative Stress/drug effects , Receptors, Melatonin/metabolism
18.
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
19.
J Pineal Res ; 57(1): 1-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24920150

ABSTRACT

Melatonin is part of the evolutionary conserved highly functional network in vertebrates. It plays a central role in the adaptative behavior of the animal to the environment, including entrainment of daily and annual physiological rhythms, reproductive behavior, food intake, locomotor activity, growth, and breeding performance. In zebrafish, apart from its synchronizing capabilities, melatonin seems to have a major role in multiple physiological processes. Extensive knowledge of its genome and the identification of a series of genes with the same functions as those in humans, the relative ease of obtaining mutants, and the similarities between zebrafish and human pathologies make it an excellent experimental model organism of human diseases. Moreover, it is a common experimental species because of easy handling, breeding, and developmental control. Among other pathophysiologies, zebrafish are now used in studies of neurodegeneration and neurological diseases, endocrine diseases, behavior, muscular dystrophies, developmental alterations, circadian rhythms, and drugs screening. The purpose of this review was to update the current knowledge on the synthesis and biological functions of melatonin in zebrafish, keeping in mind its relevance not only in the physiology of the animal, but also in pathophysiological conditions.


Subject(s)
Melatonin/metabolism , Zebrafish/metabolism , Animals , Circadian Rhythm/physiology , Pineal Gland/metabolism
20.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167283, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38851305

ABSTRACT

Obesity represents a significant health challenge, intricately linked to conditions such as type II diabetes, metabolic syndrome, and hepatic steatosis. Several existing obesity treatments exhibit limited efficacy, undesirable side effects or a limited capability to maintain therapeutics effects in the long-term. Recently, modulation Coenzyme Q (CoQ) metabolism has emerged as a promising target for treatment of metabolic syndrome. This potential intervention could involve the modulation of endogenous CoQ biosynthesis by the use of analogs of the precursor of its biosynthesis, such as ß-resorcylic acid (ß-RA). Here, we show that oral supplementation with ß-RA, incorporated into the diet of diet-induced obese (DIO) mice, leads to substantial weight loss. The anti-obesity effects of ß-RA are partially elucidated through the normalization of mitochondrial CoQ metabolism in white adipose tissue (WAT). Additionally, we identify an HFN4α/LXR-dependent transcriptomic activation of the hepatic lipid metabolism that contributes to the anti-obesity effects of ß-RA. Consequently, ß-RA mitigates WAT hypertrophy, prevents hepatic steatosis, counteracts metabolic abnormalities in WAT and liver, and enhances glucose homeostasis by reducing the insulin/glucagon ratio and plasma levels of gastric inhibitory peptide (GIP). Moreover, pharmacokinetic evaluation of ß-RA supports its translational potential. Thus, ß-RA emerges as an efficient, safe, and translatable therapeutic option for the treatment and/or prevention of obesity, metabolic dysfunction-associated steatotic liver disease (MASLD).

SELECTION OF CITATIONS
SEARCH DETAIL