Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 158
Filtrer
1.
Biochim Biophys Acta Bioenerg ; 1865(4): 149492, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38960080

RÉSUMÉ

Mitochondrial DNA (mtDNA) mutations, including the m.3243A>G mutation that causes mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS), are associated with secondary coenzyme Q10 (CoQ10) deficiency. We previously demonstrated that PPARGC1A knockdown repressed the expression of PDSS2 and several COQ genes. In the present study, we compared the mitochondrial function, CoQ10 status, and levels of PDSS and COQ proteins and genes between mutant cybrids harboring the m.3243A>G mutation and wild-type cybrids. Decreased mitochondrial energy production, defective respiratory function, and reduced CoQ10 levels were observed in the mutant cybrids. The ubiquinol-10:ubiquinone-10 ratio was lower in the mutant cybrids, indicating blockage of the electron transfer upstream of CoQ, as evident from the reduced ratio upon rotenone treatment and increased ratio upon antimycin A treatment in 143B cells. The mutant cybrids exhibited downregulation of PDSS2 and several COQ genes and upregulation of COQ8A. In these cybrids, the levels of PDSS2, COQ3-a isoform, COQ4, and COQ9 were reduced, whereas those of COQ3-b and COQ8A were elevated. The mutant cybrids had repressed PPARGC1A expression, elevated ATP5A levels, and reduced levels of mtDNA-encoded proteins, nuclear DNA-encoded subunits of respiratory enzyme complexes, MNRR1, cytochrome c, and DHODH, but no change in TFAM, TOM20, and VDAC1 levels. Alterations in the CoQ10 level in MELAS may be associated with mitochondrial energy deficiency and abnormal gene regulation. The finding of a reduction in the ubiquinol-10:ubiquinone-10 ratio in the MELAS mutant cybrids differs from our previous discovery that cybrids harboring the m.8344A>G mutation exhibit a high ubiquinol-10:ubiquinone-10 ratio.

2.
Anim Microbiome ; 6(1): 40, 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39030597

RÉSUMÉ

Radiation enteritis is a frequently encountered issue for patients receiving radiotherapy and has a significant impact on cancer patients' quality of life. The gut microbiota plays a pivotal role in intestinal function, yet the impact of irradiation on gut microorganisms is not fully understood. This study explores the gastroprotective effect and gut microbiome-modulating potential of ubiquinol (Ubq), the reduced form of the powerful antioxidant CoQ-10. For this purpose, male albino rats were randomly assigned to four groups: Control, IRR (acute 7 Gy γ-radiation), Ubq_Post (Ubq for 7 days post-irradiation), and Ubq_Pre/Post (Ubq for 7 days pre and 7 days post-irradiation). The fecal microbiomes of all groups were profiled by 16S rRNA amplicon sequencing followed by bioinformatics and statistical analysis. Histopathological examination of intestinal tissue indicated severe damage in the irradiated group, which was mitigated by ubiquinol with enhanced regeneration, goblet cells, and intestinal alkaline phosphatase expression. Compared to the irradiated group, the Ubq-treated groups had a significant recovery of intestinal interleukin-1ß, caspase-3, nitric oxide metabolites, and thio-barbituric reactive substances to near-healthy levels. Ubq_Pre/Post group displayed elevated peroxisome proliferator-activated receptor (PPAR-γ) level, suggesting heightened benefits. Serum insulin reduction in irradiated rats improved post-Ubq treatment, with a possible anti-inflammatory effect on the pancreatic tissue. Fecal microbiota profiling revealed a dysbiosis state with a reduction of bacterial diversity post-irradiation, which was re-modulated in the Ubq treated groups to profiles that are indistinguishable from the control group. These findings underscore Ubq's gastroprotective effects against radiation-induced enteritis and its potential in restoring the gut microbiota's diversity and balance.

3.
Future Cardiol ; 20(4): 221-228, 2024 Mar 11.
Article de Anglais | MEDLINE | ID: mdl-39049769

RÉSUMÉ

Aim: We aim to analyze past literature to evaluate the efficacy of coenzyme Q10 (CoQ-10) in the population with heart failure (HF). Methods: A systematic literature search was conducted through MEDLINE (via PubMed) and Cochrane Library. The outcomes analyzed were a reduction in HF-related mortality, an improvement in exercise capacity, and the left ventricular ejection fraction (LVEF). Results: Among 16 studies, CoQ-10 significantly reduced HF-related mortality by 40% and improved exercise capacity in patients with HF, but demonstrated no significant difference in LVEF however, the potential of its efficacy on LVEF could not be ruled out. Conclusion: CoQ-10 significantly enhances exercise capacity and reduces HF-related mortality; however, its impact on patients with reduced LVEF requires further investigation.


[Box: see text].


Sujet(s)
Défaillance cardiaque , Ubiquinones , Humains , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/physiopathologie , Ubiquinones/analogues et dérivés , Ubiquinones/usage thérapeutique , Ubiquinones/pharmacologie , Débit systolique/physiologie , Tolérance à l'effort/physiologie , Tolérance à l'effort/effets des médicaments et des substances chimiques , Fonction ventriculaire gauche/physiologie , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Résultat thérapeutique
4.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38928470

RÉSUMÉ

Coenzyme Q10 (CoQ10) plays a key role in many aspects of cellular metabolism. For CoQ10 to function normally, continual interconversion between its oxidised (ubiquinone) and reduced (ubiquinol) forms is required. Given the central importance of this ubiquinone-ubiquinol redox cycle, this article reviews what is currently known about this process and the implications for clinical practice. In mitochondria, ubiquinone is reduced to ubiquinol by Complex I or II, Complex III (the Q cycle) re-oxidises ubiquinol to ubiquinone, and extra-mitochondrial oxidoreductase enzymes participate in the ubiquinone-ubiquinol redox cycle. In clinical terms, the outcome of deficiencies in various components associated with the ubiquinone-ubiquinol redox cycle is reviewed, with a particular focus on the potential clinical benefits of CoQ10 and selenium co-supplementation.


Sujet(s)
Oxydoréduction , Ubiquinones , Ubiquinones/analogues et dérivés , Ubiquinones/métabolisme , Ubiquinones/déficit , Humains , Mitochondries/métabolisme , Animaux , Sélénium/métabolisme , Ataxie , Faiblesse musculaire , Maladies mitochondriales
5.
Antioxidants (Basel) ; 13(4)2024 Apr 21.
Article de Anglais | MEDLINE | ID: mdl-38671943

RÉSUMÉ

Coenzyme Q (CoQ) is a lipidic compound that is widely distributed in nature, with crucial functions in metabolism, protection against oxidative damage and ferroptosis and other processes. CoQ biosynthesis is a conserved and complex pathway involving several proteins. COQ2 is a member of the UbiA family of transmembrane prenyltransferases that catalyzes the condensation of the head and tail precursors of CoQ, which is a key step in the process, because its product is the first intermediate that will be modified in the head by the next components of the synthesis process. Mutations in this protein have been linked to primary CoQ deficiency in humans, a rare disease predominantly affecting organs with a high energy demand. The reaction catalyzed by COQ2 and its mechanism are still unknown. Here, we aimed at clarifying the COQ2 reaction by exploring possible substrate binding sites using a strategy based on homology, comprising the identification of available ligand-bound homologs with solved structures in the Protein Data Bank (PDB) and their subsequent structural superposition in the AlphaFold predicted model for COQ2. The results highlight some residues located on the central cavity or the matrix loops that may be involved in substrate interaction, some of which are mutated in primary CoQ deficiency patients. Furthermore, we analyze the structural modifications introduced by the pathogenic mutations found in humans. These findings shed new light on the understanding of COQ2's function and, thus, CoQ's biosynthesis and the pathogenicity of primary CoQ deficiency.

6.
Biology (Basel) ; 13(4)2024 Mar 30.
Article de Anglais | MEDLINE | ID: mdl-38666843

RÉSUMÉ

Formoterol, a ß2-adrenergic receptor (ß2AR) agonist, shows promise in various diseases, but its effectiveness in Parkinson's disease (PD) is debated, with unclear regulation of mitochondrial homeostasis. This study employed a cell model featuring mitochondrial ubiquinol-cytochrome c reductase core protein 1 (UQCRC1) variants associated with familial parkinsonism, demonstrating mitochondrial dysfunction and dynamic imbalance, exploring the therapeutic effects and underlying mechanisms of formoterol. Results revealed that 24-h formoterol treatment enhanced cell proliferation, viability, and neuroprotection against oxidative stress. Mitochondrial function, encompassing DNA copy number, repatriation, and complex III-linked respiration, was comprehensively restored, along with the dynamic rebalance of fusion/fission events. Formoterol reduced extensive hypertubulation, in contrast to mitophagy, by significantly upregulating protein Drp-1, in contrast to fusion protein Mfn2, mitophagy-related protein Parkin. The upstream mechanism involved the restoration of ERK signaling and the inhibition of Akt overactivity, contingent on the activation of ß2-adrenergic receptors. Formoterol additionally aided in segregating healthy mitochondria for distribution and transport, therefore normalizing mitochondrial arrangement in mutant cells. This study provides preliminary evidence that formoterol offers neuroprotection, acting as a mitochondrial dynamic balance regulator, making it a promising therapeutic candidate for PD.

7.
Int J Mol Sci ; 25(4)2024 Feb 18.
Article de Anglais | MEDLINE | ID: mdl-38397087

RÉSUMÉ

It is well known that in the heart and kidney mitochondria, more than 95% of ATP production is supported by the ß-oxidation of long-chain fatty acids. However, the ß-oxidation of fatty acids by mitochondria has been studied much less than the substrates formed during the catabolism of carbohydrates and amino acids. In the last few decades, several discoveries have been made that are directly related to fatty acid oxidation. In this review, we made an attempt to re-evaluate the ß-oxidation of long-chain fatty acids from the perspectives of new discoveries. The single set of electron transporters of the cardiac mitochondrial respiratory chain is organized into three supercomplexes. Two of them contain complex I, a dimer of complex III, and two dimers of complex IV. The third, smaller supercomplex contains a dimer of complex III and two dimers of complex IV. We also considered other important discoveries. First, the enzymes of the ß-oxidation of fatty acids are physically associated with the respirasome. Second, the ß-oxidation of fatty acids creates the highest level of QH2 and reverses the flow of electrons from QH2 through complex II, reducing fumarate to succinate. Third, ß-oxidation is greatly stimulated in the presence of succinate. We argue that the respirasome is uniquely adapted for the ß-oxidation of fatty acids. The acyl-CoA dehydrogenase complex reduces the membrane's pool of ubiquinone to QH2, which is instantly oxidized by the smaller supercomplex, generating a high energization of mitochondria and reversing the electron flow through complex II, which reverses the electron flow through complex I, increasing the NADH/NAD+ ratio in the matrix. The mitochondrial nicotinamide nucleotide transhydrogenase catalyzes a hydride (H-, a proton plus two electrons) transfer across the inner mitochondrial membrane, reducing the cytosolic pool of NADP(H), thus providing the heart with ATP for muscle contraction and energy and reducing equivalents for the housekeeping processes.


Sujet(s)
Complexe III de la chaîne respiratoire , Acides gras , Acides gras/métabolisme , Complexe III de la chaîne respiratoire/métabolisme , Oxydoréduction , Mitochondries du myocarde/métabolisme , Membranes mitochondriales/métabolisme , Complexe IV de la chaîne respiratoire/métabolisme , Acide succinique/métabolisme , Succinates/métabolisme , Complexe I de la chaîne respiratoire/métabolisme , Adénosine triphosphate/métabolisme
8.
Int J Cardiol ; 401: 131817, 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38307422

RÉSUMÉ

BACKGROUND: High-altitude exposure changes the electrical conduction of the heart. However, reports on electrocardiogram (ECG) characteristics and potent prophylactic agents during high-altitude acclimatization and de-acclimatization are inadequate. This study aimed to investigate the effects of ubiquinol on electrophysiology after high-altitude hypoxia and reoxygenation. METHODS: The study was a prospective, randomized, double-blind, placebo-controlled trial. Forty-one participants were randomly divided into two groups receiving ubiquinol 200 mg daily or placebo orally 14 days before flying to high altitude (3900 m) until the end of the study. Cardiopulmonary exercise testing was performed at baseline (300 m), on the third day after reaching high altitude, and on the seventh day after returning to baseline. RESULTS: Acute high-altitude exposure prolonged resting ventricular repolarization, represented by increased corrected QT interval (455.9 ± 23.4 vs. 427.1 ± 19.1 ms, P < 0.001) and corrected Tpeak-Tend interval (155.5 ± 27.4 vs. 125.3 ± 21.1 ms, P < 0.001), which recovered after returning to low altitude. Ubiquinol supplementation shortened the hypoxia-induced extended Tpeak-Tend interval (-7.7 ms, [95% confidence interval (CI), -13.8 to -1.6], P = 0.014), Tpeak-Tend /QT interval (-0.014 [95% CI, -0.027 to -0.002], P = 0.028), and reserved maximal heart rate (11.9 bpm [95% CI, 3.2 to 20.6], P = 0.013) during exercise at high altitude. Furthermore, the decreased resting amplitude of the ST-segment in the V3 lead was correlated with decreased peak oxygen pulse (R = 0.713, P < 0.001) and maximum oxygen consumption (R = 0.595, P < 0.001). CONCLUSIONS: Our results illustrated the electrophysiology changes during high-altitude acclimatization and de-acclimatization. Similarly, ubiquinol supplementation shortened the prolonged Tpeak-Tend interval and reserved maximal heart rate during exercise at high altitude. REGISTRATION: URL: www.chictr.org.cn; Unique identifier: ChiCTR2200059900.


Sujet(s)
Altitude , Capacité cardiorespiratoire , Ubiquinones/analogues et dérivés , Humains , Études prospectives , Hypoxie , Acclimatation , Électrophysiologie
9.
Article de Anglais | MEDLINE | ID: mdl-38181883

RÉSUMÉ

AIM: Sjögren-Larsson syndrome (SLS) is a rare neurometabolic disorder that mainly affects brain, eye and skin and is caused by deficiency of fatty aldehyde dehydrogenase. Our recent finding of a profoundly disturbed brain tissue lipidome in SLS prompted us to search for similar biomarkers in plasma as no functional test in blood is available for SLS. METHODS AND RESULTS: We performed plasma lipidomics and used a newly developed bioinformatics tool to mine the untargeted part of the SLS plasma and brain lipidome to search for SLS biomarkers. Plasma lipidomics showed disturbed ether lipid metabolism in known lipid classes. Untargeted lipidomics of both plasma and brain (white and grey matter) uncovered two new endogenous lipid classes highly elevated in SLS. The first biomarker group were alkylphosphocholines/ethanolamines containing different lengths of alkyl-chains where some alkylphosphocholines were > 600-fold elevated in SLS plasma. The second group of biomarkers were a set of 5 features of unknown structure. Fragmentation studies suggested that they contain ubiquinol and phosphocholine and one feature was also found as a glucuronide conjugate in plasma. The plasma features were highly distinctive for SLS with levels >100-1000-fold the level in controls, if present at all. We speculate on the origin of the alkylphosphocholines/ethanolamines and the nature of the ubiquinol-containing metabolites. CONCLUSIONS: The metabolites identified in this study represent novel endogenous lipid classes thus far unknown in humans. They represent the first plasma metabolite SLS-biomarkers and may also yield more insight into SLS pathophysiology.


Sujet(s)
Syndrome de Sjögren-Larsson , Humains , Syndrome de Sjögren-Larsson/diagnostic , Syndrome de Sjögren-Larsson/métabolisme , Lipidomique , Peau/métabolisme , Éthanolamines , Lipides
10.
Life (Basel) ; 14(1)2024 Jan 17.
Article de Anglais | MEDLINE | ID: mdl-38255749

RÉSUMÉ

Coenzyme CoQ10 (CoQ10) is an endogenous lipid-soluble antioxidant that effectively protects lipids, proteins, and DNA from oxidation due to its ability to undergo redox transitions between oxidized and reduced forms. Various oxidative stress-associated infectious and somatic diseases have been observed to disrupt the balance of CoQ10 concentration in tissues. As a high molecular weight polar lipophilic compound, CoQ10 exhibits very limited oral bioavailability, which restrains its therapeutic potential. Nevertheless, numerous studies have confirmed the clinical efficacy of CoQ10 therapy through oral administration of high doses over extended time periods. Experimental studies have demonstrated that in emergency situations, intravenous administration of both oxidized and reduced-form CoQ10 leads to a rapid increase in its concentration in organ tissues, offering protection for organ tissues in ischemic conditions. This suggests that the cardio- and neuroprotective efficacy of intravenously administered CoQ10 forms could present new opportunities in treating acute ischemic conditions. Based on these findings, the review provides reasoning supporting further research and implementation of CoQ10 dosage forms for intravenous administration in emergency situations into clinical practice.

11.
Curr Cardiol Rep ; 25(12): 1759-1767, 2023 12.
Article de Anglais | MEDLINE | ID: mdl-37971634

RÉSUMÉ

PURPOSE OF REVIEW: According to the World Health Organization (WHO), cardiovascular disease is the leading cause of death worldwide. Heart failure has been defined as a global pandemic leading to millions of deaths. Recent research clearly approved the beneficial effect of Coenzyme Q10 supplementation in treatment and prevention of cardiovascular disease in patients with heart failure in clinical trials but did not distinguish between the oxidised form CoQ10 and reduced form CoQH2 of Coenzyme Q10. The aim of this study is to determine differences in medical application of CoQ10 and CoQH2 supplementation and evaluate the efficacy of CoQ10 and CoQH2 supplementation to prevent cardiovascular disease in patients with heart failure. RECENT FINDINGS: A PubMed search for the terms "ubiquinone" and "ubiquinol" was conducted, and 28 clinical trials were included. Our findings go along with the biochemical description of CoQ10 and CoQH2, recording cardiovascular benefits for CoQ10 and antioxidative and anti-inflammatory properties for CoQH2. Our main outcomes are the following: (I) CoQ10 supplementation reduced cardiovascular death in patients with heart failure. This is not reported for CoQH2. (II) Test concentrations leading to cardiovascular benefits are much lower in CoQ10 studies than in CoQH2 studies. (III) Positive long-term effects reducing cardiovascular mortality are only observed in CoQ10 studies. Based on the existing literature, the authors recommend CoQ10 instead of CoQH2 to treat and prevent cardiovascular disease in patients with heart failure.


Sujet(s)
Maladies cardiovasculaires , Défaillance cardiaque , Humains , Ubiquinones/pharmacologie , Ubiquinones/usage thérapeutique , Maladies cardiovasculaires/traitement médicamenteux , Maladies cardiovasculaires/prévention et contrôle , Compléments alimentaires , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/prévention et contrôle
12.
Pharmaceutics ; 15(10)2023 Oct 20.
Article de Anglais | MEDLINE | ID: mdl-37896258

RÉSUMÉ

Coenzyme Q10 (CoQ10) exists in two forms, an oxidized form and a reduced form. Ubiquinol is the fully reduced form of CoQ10. Compared to the oxidized form, ubiquinol has a much higher biological absorption and better therapeutic effect. However, ubiquinol has an important stability problem which hampers its storage and formulation. It can be easily transformed into its oxidized form-ubiquinone-even at low temperature. In this work, we designed, synthesized, and characterized a new cocrystal of ubiquinol with vitamin B3 nicotinamide (UQ-NC). Compared to the marketed ubiquinol form, the cocrystal exhibited an excellent stability, improved dissolution properties, and higher bioavailability. The cocrystal remained stable for a long period, even when stored under stressed conditions. In the dissolution experiments, the cocrystal generated 12.6 (in SIF) and 38.3 (in SGF) times greater maximum ubiquinol concentrations above that of the marketed form. In addition, in the PK studies, compared to the marketed form, the cocrystal exhibited a 2.2 times greater maximum total coenzyme Q10 concentration and a 4.5 times greater AUC than that of the marketed form.

13.
Front Cardiovasc Med ; 10: 1129144, 2023.
Article de Anglais | MEDLINE | ID: mdl-37560117

RÉSUMÉ

Cardiorespiratory function influences exercise capacity and is an important determinant of high-altitude adaptation. Some studies have investigated the characteristics of changes in cardiorespiratory fitness during high-altitude acclimatization. However, studies on changes in cardiorespiratory fitness during high-altitude de-acclimatization are still lacking and have not yet been elucidated. Furthermore, few drugs have been studied to improve cardiorespiratory function during both processes. The Shigatse CARdiorespiratory Fitness (SCARF) study is a single-center, randomized, double-blind, placebo-control clinical trial to explore the effects of ubiquinol on cardiorespiratory fitness during high-altitude acclimatization and de-acclimatization in healthy adults. Participants will be randomly assigned 1:1 to ubiquinol 200 mg daily or a placebo for 14 days before departure until the end of data collection after return in 7 days. Cardiorespiratory fitness is the primary outcome, while acute mountain sickness and high-altitude de-acclimatization symptoms are secondary endpoints. In addition, laboratory measurements, including routine blood tests and serological measurements, will be performed. To the best of our knowledge, the SCARF study will be the first to reveal the changes in the cardiorespiratory fitness characteristics during high-altitude acclimatization and de-acclimatization. Furthermore, the results of this study will contribute to exploring whether ubiquinol supplementation could be beneficial for endurance exercise capacity at different altitudes and help improve adaptation to acute hypoxia and de-acclimatization. Clinical Trial Registration: This study has been registered in the Chinese Clinical Trial Register (www.chictr.org.cn) as ChiCTR2200059900 and ChiCTR2200066328.

14.
J Chiropr Med ; 22(3): 222-229, 2023 Sep.
Article de Anglais | MEDLINE | ID: mdl-37644997

RÉSUMÉ

Objective: The purpose of this case report was to describe a multimodal approach for the treatment of premenstrual syndrome (PMS). Clinical Features: A 36-year-old nulliparous woman presented to a free clinic for veterans and their spouses. She received a PMS diagnosis at age 18. She was previously prescribed hormonal birth control and nonsteroidal anti-inflammatory drugs, which minimally affected her condition. She stopped using conventional medicine therapies at age 27. Laboratory results showed that her progesterone was below 0.5 ng/mL. Her symptom score was 50 out of 60 on the Treatment Strategies for PMS assessment tool. During her menses, she experienced low back pain and stiffness, bloating, swelling, weight gain, breast tenderness, swelling, and pain, and she felt overwhelmed and stressed. Intervention and Outcome: Traditional Chinese medicine acupuncture was administered in conjunction with 100 mg of coenzyme Q10 (ubiquinol) and a B-100 complex once a day and 400 mg of magnesium citrate, 1000 mg of flaxseed oil (Linum usitatissimum), and 1000 mg of turmeric (Curcuma longa) twice a day. Five days before the onset of her menstrual period, she was to ingest a B-100 complex twice a day and 400 mg of magnesium citrate, 1000 mg of flaxseed oil, and 1000 mg of turmeric 3 times a day. Mindfulness meditation was encouraged twice a day for 10 minutes to reduce stress. After 12 treatments over 3 months, her symptom score decreased to 18 out of 60 and remained below 20 for an additional 32 weeks. Conclusion: This patient with PMS symptoms positively responded to a multimodal approach using traditional Chinese medicine-style acupuncture, dietary supplements, and mindfulness meditation.

15.
Int Immunopharmacol ; 123: 110647, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-37499399

RÉSUMÉ

Radiation has been applied in cancer treatment to eradicate tumors and displayed great therapeutic benefits for humans. However, it is associated with negative impacts on normal cells, not only cancer cells. Irradiation can trigger cell death through several mechanisms, such as apoptosis, necrosis, and autophagy. This study aimed to investigate the radioprotective efficacy of ubiquinol against radiation-induced splenic tissue injury in animals and the related involved mechanisms. Animals were classified into four groups: group 1 (normal untreated rats) received vehicle 5 % Tween 80; group 2 received 7 Gy γ-radiation; group 3 received 10 mg/Kg oral ubiquinol post-irradiation; and group 4 received 10 mg/Kg oral ubiquinol before and after (pre/post-) irradiation. Ubiquinol restored the spleen histoarchitecture, associated with improved immunohistochemical quantification of B and T lymphocyte markers and ameliorated hematological alterations induced by irradiation. Such effects may be due to an enhanced antioxidant pathway through stimulation of p62, Nrf2, and GSH, associated with reduced Keap1 and MDA. Moreover, ubiquinol decreased mTOR, thus enhanced autophagy markers viz. LC3-II. Furthermore, ubiquinol showed an antiapoptotic effect by enhancing Bcl-2 and reducing caspase-3 and Bax. Consequently, ubiquinol exerts a splenic-protective effect against irradiation via enhancing antioxidant, autophagic, and survival pathways.


Sujet(s)
Antioxydants , Rate , Humains , Rats , Animaux , Antioxydants/pharmacologie , Protéine-1 de type kelch associée à ECH/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Apoptose , Autophagie
16.
Antioxidants (Basel) ; 12(6)2023 May 31.
Article de Anglais | MEDLINE | ID: mdl-37371923

RÉSUMÉ

The benefits of physical exercise on health are diminished when it is non-planned, strenuous, or vigorous, which causes an increase in oxygen consumption and production of free radicals, particularly serious at the muscular level. Ubiquinol could help achieve an antioxidant, anti-inflammatory, and ergogenic effect. The aim of this study is to evaluate whether a supplementation of ubiquinol during a short period could have a positive effect on muscle aggression, physical performance, and fatigue perception in non-elite athletes after high intensity circuit weight training. One hundred healthy and well-trained men, (firemen of the Fire Department of Granada) were enrolled in a placebo-controlled, double-blinded, and randomized study, and separated into two groups: the placebo group (PG, n = 50); and the ubiquinol group (UG, n = 50), supplemented with an oral dose. Before and after the intervention, data related to the number of repetitions, muscle strength, and perceived exertion, as well as blood samples were collected. An increase was observed in the UG regarding average load and repetitions, revealing an improvement in muscle performance. Ubiquinol supplementation also reduced muscle damage markers, showing a protective effect on muscle fibers. Therefore, this study provides evidence that ubiquinol supplementation improves muscle performance and prevents muscle damage after strenuous exercise in a population of well-trained individuals who are not elite athletes.

17.
Front Bioeng Biotechnol ; 11: 1178536, 2023.
Article de Anglais | MEDLINE | ID: mdl-37168609

RÉSUMÉ

Background: The toxic gas carbon monoxide (CO) is abundantly present in synthesis gas (syngas) and certain industrial waste gases that can serve as feedstocks for the biological production of industrially significant chemicals and fuels. For efficient bacterial growth to occur, and to increase productivity and titres, a high resistance to the gas is required. The aerobic bacterium Cupriavidus necator H16 can grow on CO2 + H2, although it cannot utilise CO as a source of carbon and energy. This study aimed to increase its CO resistance through adaptive laboratory evolution. Results: To increase the tolerance of C. necator to CO, the organism was continually subcultured in the presence of CO both heterotrophically and autotrophically. Ten individual cultures were evolved heterotrophically with fructose in this manner and eventually displayed a clear growth advantage over the wild type strain. Next-generation sequencing revealed several mutations, including a single point mutation upstream of a cytochrome bd ubiquinol oxidase operon (cydA2B2), which was present in all evolved isolates. When a subset of these mutations was engineered into the parental H16 strain, only the cydA2B2 upstream mutation enabled faster growth in the presence of CO. Expression analysis, mutation, overexpression and complementation suggested that cydA2B2 transcription is upregulated in the evolved isolates, resulting in increased CO tolerance under heterotrophic but not autotrophic conditions. However, through subculturing on a syngas-like mixture with increasing CO concentrations, C. necator could also be evolved to tolerate high CO concentrations under autotrophic conditions. A mutation in the gene for the soluble [NiFe]-hydrogenase subunit hoxH was identified in the evolved isolates. When the resulting amino acid change was engineered into the parental strain, autotrophic CO resistance was conferred. A strain constitutively expressing cydA2B2 and the mutated hoxH gene exhibited high CO tolerance under both heterotrophic and autotrophic conditions. Conclusion: C. necator was evolved to tolerate high concentrations of CO, a phenomenon which was dependent on the terminal respiratory cytochrome bd ubiquinol oxidase when grown heterotrophically and the soluble [NiFe]-hydrogenase when grown autotrophically. A strain exhibiting high tolerance under both conditions was created and presents a promising chassis for syngas-based bioproduction processes.

18.
J Fungi (Basel) ; 9(5)2023 May 13.
Article de Anglais | MEDLINE | ID: mdl-37233281

RÉSUMÉ

Alternative oxidase is a terminal oxidase in the branched mitochondrial electron transport chain of most fungi including Aspergillus niger (subgenus Circumdati, section Nigri). A second, paralogous aox gene (aoxB) is extant in some A. niger isolates but also present in two divergent species of the subgenus Nidulantes-A. calidoustus and A. implicatus-as well as in Penicillium swiecickii. Black aspergilli are cosmopolitan opportunistic fungi that can cause diverse mycoses and acute aspergillosis in immunocompromised individuals. Amongst the approximately 75 genome-sequenced A. niger strains, aoxB features considerable sequence variation. Five mutations were identified that rationally affect transcription or function or terminally modify the gene product. One mutant allele that occurs in CBS 513.88 and A. niger neotype strain CBS 554.65 involves a chromosomal deletion that removes exon 1 and intron 1 from aoxB. Another aoxB allele results from retrotransposon integration. Three other alleles result from point mutations: a missense mutation of the start codon, a frameshift, and a nonsense mutation. A. niger strain ATCC 1015 has a full-length aoxB gene. The A. niger sensu stricto complex can thus be subdivided into six taxa according to extant aoxB allele, which may facilitate rapid and accurate identification of individual species.

19.
EClinicalMedicine ; 59: 101920, 2023 May.
Article de Anglais | MEDLINE | ID: mdl-37256098

RÉSUMÉ

Background: Functionally impaired variants of COQ2, encoding an enzyme in biosynthesis of coenzyme Q10 (CoQ10), were found in familial multiple system atrophy (MSA) and V393A in COQ2 is associated with sporadic MSA. Furthermore, reduced levels of CoQ10 have been demonstrated in MSA patients. Methods: This study was a multicentre, randomised, double-blinded, placebo-controlled phase 2 trial. Patients with MSA were randomly assigned (1:1) to either ubiquinol (1500 mg/day) or placebo. The primary efficacy outcome was the change in the unified multiple system atrophy rating scale (UMSARS) part 2 at 48 weeks. Efficacy was assessed in all patients who completed at least one efficacy assessment (full analysis set). Safety analyses included patients who completed at least one dose of investigational drug. This trial is registered with UMIN-CTR (UMIN000031771), where the drug name of MSA-01 was used to designate ubiquinol. Findings: Between June 26, 2018, and May 27, 2019, 139 patients were enrolled and randomly assigned to the ubiquinol group (n = 69) or the placebo group (n = 70). A total of 131 patients were included in the full analysis set (63 in the ubiquinol group; 68 in the placebo group). This study met the primary efficacy outcome (least square mean difference in UMSARS part 2 score (-1.7 [95% CI, -3.2 to -0.2]; P = 0.023)). The ubiquinol group also showed better secondary efficacy outcomes (Barthel index, Scale for the Assessment and Rating of Ataxia, and time required to walk 10 m). Rates of adverse events potentially related to the investigational drug were comparable between ubiquinol (n = 15 [23.8%]) and placebo (n = 21 [30.9%]). Interpretation: High-dose ubiquinol was well-tolerated and led to a significantly smaller decline of UMSARS part 2 score compared with placebo. Funding: Japan Agency for Medical Research and Development.

20.
J Clin Biochem Nutr ; 72(2): 101-106, 2023 Mar.
Article de Anglais | MEDLINE | ID: mdl-36936872

RÉSUMÉ

Coenzyme Q10 (CoQ10) is an important lipid-soluble antioxidant and an essential component of the mitochondria. The oral bioavailability of the reduced form of CoQ10, ubiquinol-10, has been reported to be greater than that of the oxidized form of CoQ10, ubiquinone-10, in some studies. In contrast, it has also been highlighted that the oral bioavailability of ubiquinol-10 is not superior to that of ubiquinone-10 because ubiquinol-10 may be oxidized during digestion. In fact, it has not been shown which form of CoQ10 exists in the process from oral intake to absorption in the gastrointestinal tract. In this study, the amounts of ubiquinol-10 and ubiquinone-10 were measured in the gastrointestinal content and small intestine tissue after oral administration of ubiquinol-10 or ubiquinone-10 to C57BL/6J mice. The form of CoQ10 detected in the gastrointestinal content and small intestine tissue was almost the same as that when administered orally. The results of our study suggested that the orally administered ubiquinol-10 and ubiquinone-10 mostly reached the small intestine without oxidizing to ubiquinone-10 and reducing to ubiquinol-10, and both were absorbed by the small intestine tissue in almost their original forms.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE