Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Redox Biol ; 55: 102403, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35863266

ABSTRACT

Defects in Coenzyme Q (CoQ) metabolism have been associated with primary mitochondrial disorders, neurodegenerative diseases and metabolic conditions. The consequences of CoQ deficiency have not been fully addressed, and effective treatment remains challenging. Here, we use mice with primary CoQ deficiency (Coq9R239X), and we demonstrate that CoQ deficiency profoundly alters the Q-junction, leading to extensive changes in the mitochondrial proteome and metabolism in the kidneys and, to a lesser extent, in the brain. CoQ deficiency also induces reactive gliosis, which mediates a neuroinflammatory response, both of which lead to an encephalopathic phenotype. Importantly, treatment with either vanillic acid (VA) or ß-resorcylic acid (ß-RA), two analogs of the natural precursor for CoQ biosynthesis, partially restores CoQ metabolism, particularly in the kidneys, and induces profound normalization of the mitochondrial proteome and metabolism, ultimately leading to reductions in gliosis, neuroinflammation and spongiosis and, consequently, reversing the phenotype. Together, these results provide key mechanistic insights into defects in CoQ metabolism and identify potential disease biomarkers. Furthermore, our findings clearly indicate that the use of analogs of the CoQ biosynthetic precursor is a promising alternative therapy for primary CoQ deficiency and has potential for use in the treatment of more common neurodegenerative and metabolic diseases that are associated with secondary CoQ deficiency.

2.
Int J Mol Sci ; 23(11)2022 Jun 04.
Article in English | MEDLINE | ID: mdl-35682977

ABSTRACT

Pompe disease (PD) is a rare disorder caused by mutations in the acid alpha-glucosidase (GAA) gene. Most gene therapies (GT) partially rely on the cross-correction of unmodified cells through the uptake of the GAA enzyme secreted by corrected cells. In the present study, we generated isogenic murine GAA-KO cell lines resembling severe mutations from Pompe patients. All of the generated GAA-KO cells lacked GAA activity and presented an increased autophagy and increased glycogen content by means of myotube differentiation as well as the downregulation of mannose 6-phosphate receptors (CI-MPRs), validating them as models for PD. Additionally, different chimeric murine GAA proteins (IFG, IFLG and 2G) were designed with the aim to improve their therapeutic activity. Phenotypic rescue analyses using lentiviral vectors point to IFG chimera as the best candidate in restoring GAA activity, normalising the autophagic marker p62 and surface levels of CI-MPRs. Interestingly, in vivo administration of liver-directed AAVs expressing the chimeras further confirmed the good behaviour of IFG, achieving cross-correction in heart tissue. In summary, we generated different isogenic murine muscle cell lines mimicking the severe PD phenotype, as well as validating their applicability as preclinical models in order to reduce animal experimentation.


Subject(s)
Dependovirus , Glycogen Storage Disease Type II , Animals , Cell Line , Dependovirus/genetics , Disease Models, Animal , Genetic Therapy , Genetic Vectors/genetics , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/therapy , Humans , Mice , Mice, Knockout , Muscle Cells/metabolism , Muscle, Skeletal/metabolism , Mutation , alpha-Glucosidases/metabolism
3.
Antioxidants (Basel) ; 10(11)2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34829558

ABSTRACT

Coenzyme Q (CoQ) is a vital lipophilic molecule that is endogenously synthesized in the mitochondria of each cell. The CoQ biosynthetic pathway is complex and not completely characterized, and it involves at least thirteen catalytic and regulatory proteins. Once it is synthesized, CoQ exerts a wide variety of mitochondrial and extramitochondrial functions thank to its redox capacity and its lipophilicity. Thus, low levels of CoQ cause diseases with heterogeneous clinical symptoms, which are not always understood. The decreased levels of CoQ may be primary caused by defects in the CoQ biosynthetic pathway or secondarily associated with other diseases. In both cases, the pathomechanisms are related to the CoQ functions, although further experimental evidence is required to establish this association. The conventional treatment for CoQ deficiencies is the high doses of oral CoQ10 supplementation, but this therapy is not effective for some specific clinical presentations, especially in those involving the nervous system. To better understand the CoQ biosynthetic pathway, the biological functions linked to CoQ and the pathomechanisms of CoQ deficiencies, and to improve the therapeutic outcomes of this syndrome, a variety of animal models have been generated and characterized in the last decade. In this review, we show all the animal models available, remarking on the most important outcomes that each model has provided. Finally, we also comment some gaps and future research directions related to CoQ metabolism and how the current and novel animal models may help in the development of future research studies.

4.
Biomedicines ; 9(10)2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34680574

ABSTRACT

Primary mitochondrial diseases are caused by mutations in mitochondrial or nuclear genes, leading to the abnormal function of specific mitochondrial pathways. Mitochondrial dysfunction is also a secondary event in more common pathophysiological conditions, such as obesity and metabolic syndrome. In both cases, the improvement and management of mitochondrial homeostasis remain challenging. Here, we show that beta-resorcylic acid (ß-RA), which is a natural phenolic compound, competed in vivo with 4-hydroxybenzoic acid, which is the natural precursor of coenzyme Q biosynthesis. This led to a decrease in demethoxyubiquinone, which is an intermediate metabolite of CoQ biosynthesis that is abnormally accumulated in Coq9R239X mice. As a consequence, ß-RA rescued the phenotype of Coq9R239X mice, which is a model of primary mitochondrial encephalopathy. Moreover, we observed that long-term treatment with ß-RA also reduced the size and content of the white adipose tissue (WAT) that is normally accumulated during aging in wild-type mice, leading to the prevention of hepatic steatosis and an increase in survival at the elderly stage of life. The reduction in WAT content was due to a decrease in adipogenesis, an adaptation of the mitochondrial proteome in the kidneys, and stimulation of glycolysis and acetyl-CoA metabolism. Therefore, our results demonstrate that ß-RA acted through different cellular mechanisms, with effects on mitochondrial metabolism; as such, it may be used for the treatment of primary coenzyme Q deficiency, overweight, and hepatic steatosis.

5.
Antioxidants (Basel) ; 10(4)2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33810539

ABSTRACT

Coenzyme Q10 (CoQ10) is classically viewed as an important endogenous antioxidant and key component of the mitochondrial respiratory chain. For this second function, CoQ molecules seem to be dynamically segmented in a pool attached and engulfed by the super-complexes I + III, and a free pool available for complex II or any other mitochondrial enzyme that uses CoQ as a cofactor. This CoQ-free pool is, therefore, used by enzymes that link the mitochondrial respiratory chain to other pathways, such as the pyrimidine de novo biosynthesis, fatty acid ß-oxidation and amino acid catabolism, glycine metabolism, proline, glyoxylate and arginine metabolism, and sulfide oxidation metabolism. Some of these mitochondrial pathways are also connected to metabolic pathways in other compartments of the cell and, consequently, CoQ could indirectly modulate metabolic pathways located outside the mitochondria. Thus, we review the most relevant findings in all these metabolic functions of CoQ and their relations with the pathomechanisms of some metabolic diseases, highlighting some future perspectives and potential therapeutic implications.

6.
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
7.
Nutrients ; 11(9)2019 Sep 14.
Article in English | MEDLINE | ID: mdl-31540029

ABSTRACT

Coenzyme Q (CoQ) is an essential endogenously synthesized molecule that links different metabolic pathways to mitochondrial energy production thanks to its location in the mitochondrial inner membrane and its redox capacity, which also provide it with the capability to work as an antioxidant. Although defects in CoQ biosynthesis in human and mouse models cause CoQ deficiency syndrome, some animals models with particular defects in the CoQ biosynthetic pathway have shown an increase in life span, a fact that has been attributed to the concept of mitohormesis. Paradoxically, CoQ levels decline in some tissues in human and rodents during aging and coenzyme Q10 (CoQ10) supplementation has shown benefits as an anti-aging agent, especially under certain conditions associated with increased oxidative stress. Also, CoQ10 has shown therapeutic benefits in aging-related disorders, particularly in cardiovascular and metabolic diseases. Thus, we discuss the paradox of health benefits due to a defect in the CoQ biosynthetic pathway or exogenous supplementation of CoQ10.


Subject(s)
Aging , Ataxia , Mitochondrial Diseases , Muscle Weakness , Ubiquinone/analogs & derivatives , Ubiquinone/deficiency , Adult , Animals , Antioxidants , Caenorhabditis elegans , Diet , Female , Hormesis/physiology , Humans , Male , Mice , Middle Aged , Mitochondria/physiology , Rats , Young Adult
8.
EBioMedicine ; 42: 511-523, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30898651

ABSTRACT

BACKGROUND: The vast majority of mitochondrial disorders have limited the clinical management to palliative care. Rapamycin has emerged as a potential therapeutic drug for mitochondrial diseases since it has shown therapeutic benefits in a few mouse models of mitochondrial disorders. However, the underlying therapeutic mechanism is unclear, the minimal effective dose needs to be defined and whether this therapy can be generally used is unknown. METHODS: We have evaluated whether low and high doses of rapamycin administration may result in therapeutic effects in a mouse model (Coq9R239X) of mitochondrial encephalopathy due to CoQ deficiency. The evaluation involved phenotypic, molecular, image (histopathology and MRI), metabolomics, transcriptomics and bioenergetics analyses. FINDINGS: Low dose of rapamycin induces metabolic changes in liver and transcriptomics modifications in midbrain. The high dose of rapamycin induces further changes in the transcriptomics profile in midbrain due to the general inhibition of mTORC1. However, neither low nor high dose of rapamycin were able to improve the mitochondrial bioenergetics, the brain injuries and the phenotypic characteristics of Coq9R239X mice, resulting in the lack of efficacy for increasing the survival. INTERPRETATION: These results may be due to the lack of microgliosis-derived neuroinflammation, the limitation to induce autophagy, or the need of a functional CoQ-junction. Therefore, the translation of rapamycin therapy into the clinic for patients with mitochondrial disorders requires, at least, the consideration of the particularities of each mitochondrial disease. FUND: Supported by the grants from "Fundación Isabel Gemio - Federación Española de Enfermedades Neuromusculares - Federación FEDER" (TSR-1), the NIH (P01HD080642) and the ERC (Stg-337327).


Subject(s)
Mitochondrial Diseases/drug therapy , Sirolimus/therapeutic use , Animals , Autophagy , Cell Respiration/drug effects , Cell Respiration/genetics , Disease Models, Animal , Gene Expression Profiling , Humans , Metabolomics/methods , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/etiology , Mitochondrial Encephalomyopathies/drug therapy , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Encephalomyopathies/metabolism , Phenotype , Sirolimus/administration & dosage , Sirolimus/adverse effects , Sirolimus/pharmacokinetics , Treatment Outcome , Ubiquinone/analogs & derivatives , Ubiquinone/genetics , Ubiquinone/metabolism
9.
EMBO Mol Med ; 11(1)2019 01.
Article in English | MEDLINE | ID: mdl-30482867

ABSTRACT

Coenzyme Q (CoQ) deficiency has been associated with primary defects in the CoQ biosynthetic pathway or to secondary events. In some cases, the exogenous CoQ supplementation has limited efficacy. In the Coq9R239X mouse model with fatal mitochondrial encephalopathy due to CoQ deficiency, we have tested the therapeutic potential of ß-resorcylic acid (ß-RA), a structural analog of the CoQ precursor 4-hydroxybenzoic acid and the anti-inflammatory salicylic acid. ß-RA noticeably rescued the phenotypic, morphological, and histopathological signs of the encephalopathy, leading to a significant increase in the survival. Those effects were due to the decrease of the levels of demethoxyubiquinone-9 (DMQ9) and the increase of mitochondrial bioenergetics in peripheral tissues. However, neither CoQ biosynthesis nor mitochondrial function changed in the brain after the therapy, suggesting that some endocrine interactions may induce the reduction of the astrogliosis, spongiosis, and the secondary down-regulation of astrocytes-related neuroinflammatory genes. Because the therapeutic outcomes of ß-RA administration were superior to those after CoQ10 supplementation, its use in the clinic should be considered in CoQ deficiencies.


Subject(s)
Hydroxybenzoates/administration & dosage , Mitochondrial Encephalomyopathies/drug therapy , Mitochondrial Encephalomyopathies/pathology , Neuroprotective Agents/administration & dosage , Ubiquinone/analogs & derivatives , Animals , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Energy Metabolism , Histocytochemistry , Mice , Salicylic Acid/administration & dosage , Survival Analysis , Treatment Outcome , Ubiquinone/analysis , Ubiquinone/deficiency , Ubiquinone/genetics , Ubiquinone/metabolism
10.
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
11.
PLoS One ; 12(8): e0183090, 2017.
Article in English | MEDLINE | ID: mdl-28800639

ABSTRACT

MPTP-mouse model constitutes a well-known model of neuroinflammation and mitochondrial failure occurring in Parkinson's disease (PD). Although it has been extensively reported that nitric oxide (NO●) plays a key role in the pathogenesis of PD, the relative roles of nitric oxide synthase isoforms iNOS and nNOS in the nigrostriatal pathway remains, however, unclear. Here, the participation of iNOS/nNOS isoforms in the mitochondrial dysfunction was analyzed in iNOS and nNOS deficient mice. Our results showed that MPTP increased iNOS activity in substantia nigra and striatum, whereas it sharply reduced complex I activity and mitochondrial bioenergetics in all strains. In the presence of MPTP, mice lacking iNOS showed similar restricted mitochondrial function than wild type or mice lacking nNOS. These results suggest that iNOS-dependent elevated nitric oxide, a major pathological hallmark of neuroinflammation in PD, does not contribute to mitochondrial impairment. Therefore, neuroinflammation and mitochondrial dysregulation seem to act in parallel in the MPTP model of PD. Melatonin administration, with well-reported neuroprotective properties, counteracted these effects, preventing from the drastic changes in mitochondrial oxygen consumption, increased NOS activity and prevented reduced locomotor activity induced by MPTP. The protective effects of melatonin on mitochondria are also independent of its anti-inflammatory properties, but both effects are required for an effective anti-parkinsonian activity of the indoleamine as reported in this study.


Subject(s)
Melatonin/pharmacology , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type I/genetics , Parkinson Disease, Secondary/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/deficiency , Nitric Oxide Synthase Type II/deficiency , Oxygen Consumption/drug effects , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/genetics , Parkinson Disease, Secondary/pathology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology
12.
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
13.
PLoS One ; 11(6): e0158344, 2016.
Article in English | MEDLINE | ID: mdl-27341668

ABSTRACT

Recent clinical trials have shown that in vivo and ex vivo gene therapy strategies can be an option for the treatment of several neurological disorders. Both strategies require efficient and safe vectors to 1) deliver the therapeutic gene directly into the CNS or 2) to genetically modify stem cells that will be used as Trojan horses for the systemic delivery of the therapeutic protein. A group of target diseases for these therapeutic strategies are mitochondrial encephalopathies due to mutations in nuclear DNA genes. In this study, we have developed a lentiviral vector (CCoq9WP) able to overexpress Coq9 mRNA and COQ9 protein in mouse embryonic fibroblasts (MEFs) and hematopoietic progenitor cells (HPCs) from Coq9R239X mice, an animal model of mitochondrial encephalopathy due to primary Coenzyme Q (CoQ) deficiency. Ectopic over-expression of Coq9 in both cell types restored the CoQ biosynthetic pathway and mitochondrial function, improving the fitness of the transduced cells. These results show the potential of the CCoq9WP lentiviral vector as a tool for gene therapy to treat mitochondrial encephalopathies.


Subject(s)
Fibroblasts/metabolism , Genetic Therapy , Hematopoietic Stem Cells/metabolism , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Proteins/genetics , Animals , Bone Marrow Transplantation , Disease Models, Animal , Gene Expression , Genetic Therapy/methods , Genetic Vectors/genetics , Lentivirus/genetics , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Encephalomyopathies/therapy , Mitochondrial Proteins/metabolism , Physical Fitness , Transduction, Genetic , Ubiquinone/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...