Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
Add more filters

Publication year range
1.
Bol Asoc Med P R ; 107(2): 60-6, 2015.
Article in Spanish | MEDLINE | ID: mdl-26434086

ABSTRACT

Human development and its physiology depends on a number of complex biochemical body processes, many of which are interactive and codependent. The speed and the degree in which many physiological reactions are completed depend on enzyme activity, which in turn depends on the bioavailability of co-factors and micronutrients such as vitamins and minerals. To achieve a healthy physiological state, organism need that biochemical reactions occur in a controlled and specific way at a particular speed and level or grade fully completed. To achieve this, is required an optimal metabolic balance. Factors such as, a particular genetic composition, inadequate dietary consumption patterns, traumas, diseases, toxins and environmental stress all of these factors rising demands for nutrients in order to obtain optimal metabolic balance. Metabolic correction is a biochemical and physiological concept that explains how improvements in cellular biochemistry of an organism can help the body achieve metabolic and physiological optimization. We summarize the contribution of several pioneers in understanding the role of micronutrients in health management. The concept of metabolic correction is becoming a significant term due to the presence of genetic variants that affect the speed of reactions of enzymes, causing metabolic alterations that enhance or promote the state/development of multiple diseases. Decline in the nutritional value of the food we eat, the increase in demand for certain nutrients caused by normal development, diseases and medications induce, usually, nutrients consumption. These nutritional deficiencies and insufficiencies are causing massive economic costs due to increased morbidity and mortality in our society. In summary, metabolic correction improves the enzymatic function, which favors the physiological normal functions, thus, contributing to improving health and the welfare of the human being. The purpose of this paper is to describe and introduce the concept of optimal metabolic correction as a functional cost-effective mechanism against disease, in addition, to contribute to diseases prevention and regeneration of the body and health.


Subject(s)
Micronutrients/physiology , Primary Prevention/methods , Avitaminosis/complications , Avitaminosis/therapy , Coenzymes/deficiency , Coenzymes/physiology , Coenzymes/therapeutic use , DNA Damage , Dietary Supplements , Energy Metabolism , Enzymes/physiology , Feeding Behavior , Humans , Malnutrition/complications , Malnutrition/therapy , Micronutrients/deficiency , Micronutrients/therapeutic use , Minerals/therapeutic use , Models, Biological , Nutritional Requirements , Precision Medicine , United States , Vitamins/therapeutic use
2.
Hum Mol Genet ; 20(13): 2628-41, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21527427

ABSTRACT

The discovery of a pharmacological treatment for phenylketonuria (PKU) raised new questions about function and dysfunction of phenylalanine hydroxylase (PAH), the enzyme deficient in this disease. To investigate the interdependence of the genotype, the metabolic state (phenylalanine substrate) and treatment (BH(4) cofactor) in the context of enzyme function in vitro and in vivo, we (i) used a fluorescence-based method for fast enzyme kinetic analyses at an expanded range of phenylalanine and BH(4) concentrations, (ii) depicted PAH function as activity landscapes, (iii) retraced the analyses in eukaryotic cells, and (iv) translated this into the human system by analyzing the outcome of oral BH(4) loading tests. PAH activity landscapes uncovered the optimal working range of recombinant wild-type PAH and provided new insights into PAH kinetics. They demonstrated how mutations might alter enzyme function in the space of varying substrate and cofactor concentrations. Experiments in eukaryotic cells revealed that the availability of the active PAH enzyme depends on the phenylalanine-to-BH(4) ratio. Finally, evaluation of data from BH(4) loading tests indicated that the patient's genotype influences the impact of the metabolic state on drug response. The results allowed for visualization and a better understanding of PAH function in the physiological and pathological state as well as in the therapeutic context of cofactor treatment. Moreover, our data underscore the need for more personalized procedures to safely identify and treat patients with BH(4)-responsive PAH deficiency.


Subject(s)
Biopterins/analogs & derivatives , Coenzymes/therapeutic use , Genotype , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Phenylalanine/metabolism , Phenylketonurias , Biopterins/pharmacology , Biopterins/therapeutic use , Coenzymes/pharmacology , Enzyme Activation/drug effects , HEK293 Cells , Humans , Kinetics , Molecular Chaperones/metabolism , Mutation/genetics , Phenylalanine Hydroxylase/deficiency , Phenylketonurias/drug therapy , Phenylketonurias/enzymology , Phenylketonurias/genetics
4.
Harefuah ; 151(6): 362-3, 377, 376, 2012 Jun.
Article in Hebrew | MEDLINE | ID: mdl-22991868

ABSTRACT

Healthcare systems worldwide are dealing with the uncertainty characterizing new and expensive health technoLogies, particularly aspects involving drug effectiveness and the extent and doses required for utilization. Reducing this uncertainty can be achieved mainly by using either coverage with evidence development methods or risk-sharing schemes (RSS). In 2011, the first phenylketonuria (PKU) risk-sharing scheme was set up in Israel, through the public funding health services updating process. This was done in order to ensure that people with PKU could access PKU sole treatment--sapropterin dihydrochloride, Kuvan. The apparent effectiveness of the treatment, on one hand, and the uncertainty regarding the number of patients and average treatment dosage, on the other hand, dictated the RRS. This scheme determined a ceiling number of tablets to be funded by the insurer, above this ceiling the manufacturer would finance Kuvan. Furthermore, it was agreed that after 3 years Kuvan would be brought to the public committee for updating reimbursement decisions. It is inevitable that risk sharing and conditional coverage agreements will become a common practice in the reimbursement process in the future. This will allow competent authorities and pharmaceutical companies to build clinical experience and other required data with medicines which might normally not be eLigible for reimbursement. Before it becomes the common practice in Israel, the RSS for Kuvan, process and outcomes, should be monitored and analyzed by the Ministry of Health, to ensure patients access to treatment, the effective collection of the research data and the effective interaction between Israel's four health funds and the manufacturer.


Subject(s)
Biopterins/analogs & derivatives , Medication Therapy Management/organization & administration , Phenylketonurias , Biopterins/economics , Biopterins/therapeutic use , Coenzymes/economics , Coenzymes/therapeutic use , Cost Sharing/methods , Drug Costs/trends , Humans , Israel , Phenylketonurias/drug therapy , Phenylketonurias/economics , Reimbursement Mechanisms/trends , Risk Sharing, Financial
5.
Front Endocrinol (Lausanne) ; 13: 810242, 2022.
Article in English | MEDLINE | ID: mdl-35265037

ABSTRACT

Purpose: Male infertility is a global public health issue recognized by the WHO. Recently, antioxidants are increasingly used to treat idiopathic male infertility. However, the lack of available evidence has led to the inability to rank the effects of antioxidants on the sperm quality parameters and pregnancy rate of infertile men. This network meta-analysis studied the effects of different antioxidants on the sperm quality and pregnancy rate of idiopathic male infertility. Methods: We searched PubMed, Embase, Web of Science, and Cochrane Library databases for randomized controlled trials (RCTs). The weighted mean difference (WMD) and odds ratio (OR) were applied for the comparison of continuous and dichotomous variables, respectively, with 95% CIs. The outcomes were sperm motility, sperm concentration, sperm morphology, and pregnancy rate. Results: A total of 23 RCTs with 1,917 patients and 10 kids of antioxidants were included. l-Carnitine, l-carnitine+l-acetylcarnitine, coenzyme-Q10, ω-3 fatty acid, and selenium were more efficacious than placebo in sperm quality parameters. l-Carnitine was ranked first in sperm motility and sperm morphology (WMD 6.52% [95% CI: 2.55% to 10.05%], WMD 4.96% [0.20% to 9.73%]). ω-3 fatty acid was ranked first in sperm concentration (WMD 9.89 × 106/ml, [95% CI: 7.01 to 12.77 × 106/ml]). In terms of pregnancy rate, there was no significant effect as compared with placebo. Conclusions: l-Carnitine was ranked first in sperm motility and sperm morphology. ω-3 fatty acid was ranked first in sperm concentration. Coenzyme-Q10 had better effective treatment on sperm motility and concentration. Furthermore, high-quality RCTs with adequate sample sizes should be conducted to compare the outcomes of different antioxidants.


Subject(s)
Fatty Acids, Omega-3 , Infertility, Male , Antioxidants/pharmacology , Carnitine , Coenzymes/pharmacology , Coenzymes/therapeutic use , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Female , Humans , Infertility, Male/drug therapy , Male , Network Meta-Analysis , Pregnancy , Pregnancy Rate , Randomized Controlled Trials as Topic , Spermatozoa
6.
J Biol Chem ; 285(40): 30686-97, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20667834

ABSTRACT

Protein misfolding with loss-of-function of the enzyme phenylalanine hydroxylase (PAH) is the molecular basis of phenylketonuria in many individuals carrying missense mutations in the PAH gene. PAH is complexly regulated by its substrate L-Phenylalanine and its natural cofactor 6R-L-erythro-5,6,7,8-tetrahydrobiopterin (BH(4)). Sapropterin dihydrochloride, the synthetic form of BH(4), was recently approved as the first pharmacological chaperone to correct the loss-of-function phenotype. However, current knowledge about enzyme function and regulation in the therapeutic setting is scarce. This illustrates the need for comprehensive analyses of steady state kinetics and allostery beyond single residual enzyme activity determinations to retrace the structural impact of missense mutations on the phenylalanine hydroxylating system. Current standard PAH activity assays are either indirect (NADH) or discontinuous due to substrate and product separation before detection. We developed an automated fluorescence-based continuous real-time PAH activity assay that proved to be faster and more efficient but as precise and accurate as standard methods. Wild-type PAH kinetic analyses using the new assay revealed cooperativity of activated PAH toward BH(4), a previously unknown finding. Analyses of structurally preactivated variants substantiated BH(4)-dependent cooperativity of the activated enzyme that does not rely on the presence of l-Phenylalanine but is determined by activating conformational rearrangements. These findings may have implications for an individualized therapy, as they support the hypothesis that the patient's metabolic state has a more significant effect on the interplay of the drug and the conformation and function of the target protein than currently appreciated.


Subject(s)
Biopterins/analogs & derivatives , Coenzymes/chemistry , Phenylalanine Hydroxylase/chemistry , Phenylalanine/chemistry , Allosteric Regulation/genetics , Biopterins/chemistry , Biopterins/metabolism , Biopterins/therapeutic use , Coenzymes/metabolism , Coenzymes/therapeutic use , Enzyme Activation/genetics , Fluorescence , Humans , Kinetics , Mutation, Missense , Phenylalanine/genetics , Phenylalanine/metabolism , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Phenylketonurias/drug therapy , Phenylketonurias/enzymology , Phenylketonurias/genetics
7.
Reprod Toxicol ; 101: 50-62, 2021 04.
Article in English | MEDLINE | ID: mdl-33548410

ABSTRACT

Evidences have shown that alterations in testicular dehydrogenase and ionic-ATPase activities have important implications in spermatogenesis and sperm capacitation, a penultimate biochemical change required for fertilization. Previous studies have revealed that taurine and coenzyme-Q10 (COQ-10), which are synergistic testicle-active bioflavonoids, with proven gonadotropin-enhancing properties reduce testicular damage in rats. Hence, this study investigated the effects of taurine and COQ-10 or their combination alone, and in the preventive and reversal of chlorpromazine-induced inhibition of testicular dehydrogenase enzymes, electrogenic pumps, sperm capacitation and acrosomal-reaction in male Wister rats. In the drug-treatment alone or preventive-protocol, rats received oral treatment of saline (10 mL/kg), taurine (150 mg/kg/day), COQ-10 (10 mg/kg/day) or both alone repeatedly for 56 days, or in combination with chlorpromazine (30 mg/kg/p.o./day) from days 29-56. In the reversal-protocol, the animals received chlorpromazine for 56 days prior to saline, taurine, COQ-10 or the combination from days 29-56. Thereafter, spermatogenesis (sperm count, viability, motility and morphology), testicular dehydrogenase [3beta-hydroxysteroid dehydrogenase (3ß-HSD), 17beta-hydroxysteroid dehydrogenase (17ß-HSD), glucose-6-phosphate dehydrogenase (G6PDH), lactate dehydrogenase-X (LDH-X)], ATPase (Na+/K+, Ca2+, Mg2+, H+) activities, sperm capacitation and acrosomal reaction were evaluated. Taurine and COQ-10 or their combination increased spermatogenesis, testicular 3ß-HSD, 17ß-HSD, G6PDH and LDH-X enzymes of naïve and chlorpromazine-treated rats. Both taurine and COQ-10 increased Na+/K+, Ca2+, Mg2+ and H+-ATPase activities. Also, taurine and COQ-10 or their combination prevented and reversed chlorpromazine-induced inhibition of sperm capacitation and acrosomal-reaction. The study showed that taurine and COQ-10 prevent and reverse chlorpromazine-induced inhibition of spermatogenesis, epididymal sperm capacitation and acrosomal reaction in rats through increased testicular dehydrogenases and electrogenic pump activities.


Subject(s)
Antipsychotic Agents/toxicity , Chlorpromazine/toxicity , Coenzymes/therapeutic use , Flavonoids/therapeutic use , Protective Agents/therapeutic use , Taurine/therapeutic use , Testis/drug effects , 17-Hydroxysteroid Dehydrogenases/metabolism , Adenosine Triphosphatases/metabolism , Animals , Coenzymes/pharmacology , Drug Synergism , Flavonoids/pharmacology , Glucosephosphate Dehydrogenase/metabolism , Isoenzymes/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Protective Agents/pharmacology , Rats , Rats, Wistar , Sperm Count , Sperm Motility , Spermatozoa/drug effects , Spermatozoa/physiology , Taurine/pharmacology , Testis/metabolism
8.
Nutrients ; 11(7)2019 Jul 12.
Article in English | MEDLINE | ID: mdl-31336926

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is caused by the imbalance between lipid deposition and lipid removal from the liver, and its global prevalence continues to increase dramatically. NAFLD encompasses a spectrum of pathological conditions including simple steatosis and non-alcoholic steatohepatitis (NASH), which can progress to cirrhosis and liver cancer. Even though there is a multi-disciplinary effort for development of a treatment strategy for NAFLD, there is not an approved effective medication available. Single or combined metabolic cofactors can be supplemented to boost the metabolic processes altered in NAFLD. Here, we review the dosage and usage of metabolic cofactors including l-carnitine, Nicotinamide riboside (NR), l-serine, and N-acetyl-l-cysteine (NAC) in human clinical studies to improve the altered biological functions associated with different human diseases. We also discuss the potential use of these substances in treatment of NAFLD and other metabolic diseases including neurodegenerative and cardiovascular diseases of which pathogenesis is linked to mitochondrial dysfunction.


Subject(s)
Coenzymes/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , Cardiovascular Diseases/drug therapy , Humans , Neurodegenerative Diseases/drug therapy
9.
Arterioscler Thromb Vasc Biol ; 27(6): 1334-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17413035

ABSTRACT

OBJECTIVE: 6R-5,6,7,8-tetrahydro-L-biopterin (6R-BH4) is a cofactor for endothelial nitric oxide synthase but also has antioxidant properties. Its stereo-isomer 6S-5,6,7,8-tetrahydro-L-biopterin (6S-BH4) and structurally similar pterin 6R,S-5,6,7,8-tetrahydro-D-neopterin (NH4) are also antioxidants but have no cofactor function. When endothelial nitric oxide synthase is 6R-BH4-deplete, it synthesizes superoxide rather than nitric oxide. Reduced nitric oxide bioavailability by interaction with reactive oxygen species is implicated in endothelial dysfunction (ED). 6R-BH4 corrects ED in animal models of ischemia reperfusion injury (IRI) and in patients with cardiovascular risks. It is uncertain whether the effect of exogenous 6R-BH4 on ED is through its cofactor or antioxidant action. METHODS AND RESULTS: In healthy volunteers, forearm blood flow was measured by venous occlusion plethysmography during intra-arterial infusion of the endothelium-dependent vasodilator acetylcholine, or the endothelium-independent vasodilator glyceryl trinitrate, before and after IRI. IRI reduced plasma total antioxidant status (P=0.03) and impaired vasodilatation to acetylcholine (P=0.01), but not to glyceryl trinitrate (P=0.3). Intra-arterial infusion of 6R-BH4, 6S-BH4 and NH4 at approximately equimolar concentrations prevented IRI. CONCLUSION: IRI causes ED associated with increased oxidative stress that is prevented by 6R-BH4, 6S-BH4, and NH4, an effect mediated perhaps by an antioxidant rather than cofactor function. Regardless of mechanism, 6R-BH4, 6S-BH4, or NH4 may reduce tissue injury during clinical IRI syndromes.


Subject(s)
Antioxidants/therapeutic use , Biopterins/analogs & derivatives , Coenzymes/therapeutic use , Endothelium, Vascular/drug effects , Forearm/blood supply , Reperfusion Injury/prevention & control , Vasodilation/drug effects , Acetylcholine/pharmacology , Adult , Antioxidants/chemistry , Antioxidants/pharmacology , Biopterins/blood , Biopterins/chemistry , Biopterins/pharmacology , Biopterins/therapeutic use , Coenzymes/blood , Coenzymes/chemistry , Coenzymes/pharmacology , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Female , Humans , Male , Nitric Oxide/blood , Nitric Oxide Synthase/metabolism , Oxidative Stress/drug effects , Regional Blood Flow/drug effects , Reperfusion Injury/metabolism , Reperfusion Injury/physiopathology , Stereoisomerism , Time Factors , Vasodilator Agents/pharmacology
10.
Brain ; 130(Pt 8): 2037-44, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17412732

ABSTRACT

Coenzyme Q10 (CoQ10) deficiency is an autosomal recessive disorder with heterogenous phenotypic manifestations and genetic background. We describe seven patients from five independent families with an isolated myopathic phenotype of CoQ10 deficiency. The clinical, histological and biochemical presentation of our patients was very homogenous. All patients presented with exercise intolerance, fatigue, proximal myopathy and high serum CK. Muscle histology showed lipid accumulation and subtle signs of mitochondrial myopathy. Biochemical measurement of muscle homogenates showed severely decreased activities of respiratory chain complexes I and II + III, while complex IV (COX) was moderately decreased. CoQ10 was significantly decreased in the skeletal muscle of all patients. Tandem mass spectrometry detected multiple acyl-CoA deficiency, leading to the analysis of the electron-transferring-flavoprotein dehydrogenase (ETFDH) gene, previously shown to result in another metabolic disorder, glutaric aciduria type II (GAII). All of our patients carried autosomal recessive mutations in ETFDH, suggesting that ETFDH deficiency leads to a secondary CoQ10 deficiency. Our results indicate that the late-onset form of GAII and the myopathic form of CoQ10 deficiency are allelic diseases. Since this condition is treatable, correct diagnosis is of the utmost importance and should be considered both in children and in adults. We suggest to give patients both CoQ10 and riboflavin supplementation, especially for long-term treatment.


Subject(s)
Electron-Transferring Flavoproteins/genetics , Iron-Sulfur Proteins/genetics , Muscular Diseases/genetics , Mutation , Oxidoreductases Acting on CH-NH Group Donors/genetics , Ubiquinone/analogs & derivatives , Adolescent , Adult , Biopsy , Child , Coenzymes/deficiency , Coenzymes/therapeutic use , Female , Follow-Up Studies , Humans , Male , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscular Diseases/drug therapy , Muscular Diseases/enzymology , Riboflavin/therapeutic use , Ubiquinone/deficiency , Ubiquinone/therapeutic use
11.
Eur J Cancer ; 43(7): 1134-40, 2007 May.
Article in English | MEDLINE | ID: mdl-17383867

ABSTRACT

Anthracycline-induced cardiotoxicity after treatment for childhood cancer is a considerable and serious problem. In this review, important insight into the current state of the evidence on the use of different cardioprotective agents, different anthracycline analogues, and different anthracycline infusion durations to reduce or prevent cardiotoxicity in children treated with anthracyclines is provided. It has become clear that, at the present time, there is not enough reliable evidence for many aspects of the prevention of anthracycline-induced cardiotoxicity in children. More high quality research is necessary. Suggestions for future research have been presented. As the results of these new studies become available, it will hopefully be possible to develop evidence-based recommendations for preventing anthracycline-induced cardiotoxicity in children. Until then, we can only advise care providers to carefully monitor the cardiac function of children treated with anthracyclines. With regard to the use of the cardioprotectant dexrazoxane, it might be justified to use dexrazoxane in children if the risk of cardiac damage is expected to be high. However, for each individual patient, care providers should weigh the cardioprotective effect of dexrazoxane against the possible risk of adverse effects including a lower response rate. We recommend its use in the context of well-designed studies.


Subject(s)
Anthracyclines/adverse effects , Antineoplastic Agents/adverse effects , Cardiotonic Agents/therapeutic use , Heart Diseases/chemically induced , Child , Coenzymes/therapeutic use , Heart Diseases/prevention & control , Humans , Randomized Controlled Trials as Topic , Razoxane/therapeutic use , Risk Factors , Ubiquinone/analogs & derivatives , Ubiquinone/therapeutic use
12.
Am J Cardiol ; 100(9): 1400-3, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17950797

ABSTRACT

Myalgia is the most frequently reported adverse side effect associated with statin therapy and often necessitates reduction in dose, or the cessation of therapy, compromising cardiovascular risk management. One postulated mechanism for statin-related myalgia is mitochondrial dysfunction through the depletion of coenzyme Q(10), a key component of the mitochondrial electron transport chain. This pilot study evaluated the effect of coenzyme Q(10) supplementation on statin tolerance and myalgia in patients with previous statin-related myalgia. Forty-four patients were randomized to coenzyme Q(10) (200 mg/day) or placebo for 12 weeks in combination with upward dose titration of simvastatin from 10 mg/day, doubling every 4 weeks if tolerated to a maximum of 40 mg/day. Patients experiencing significant myalgia reduced their statin dose or discontinued treatment. Myalgia was assessed using a visual analogue scale. There was no difference between combined therapy and statin alone in the myalgia score change (median 6.0 [interquartile range 2.1 to 8.8] vs 2.3 [0 to 12.8], p = 0.63), in the number of patients tolerating simvastatin 40 mg/day (16 of 22 [73%] with coenzyme Q(10) vs 13 of 22 [59%] with placebo, p = 0.34), or in the number of patients remaining on therapy (16 of 22 [73%] with coenzyme Q(10) vs 18 of 22 [82%] with placebo, p = 0.47). In conclusion, coenzyme Q(10) supplementation did not improve statin tolerance or myalgia, although further studies are warranted.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscular Diseases/chemically induced , Simvastatin/adverse effects , Ubiquinone/analogs & derivatives , Vitamins/therapeutic use , Coenzymes/blood , Coenzymes/therapeutic use , Double-Blind Method , Drug Tolerance , Female , Humans , Male , Middle Aged , Pilot Projects , Ubiquinone/blood , Ubiquinone/therapeutic use
13.
Am J Cardiol ; 99(10): 1409-12, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17493470

ABSTRACT

Treatment of hypercholesterolemia with statins (3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors) is effective in the primary and secondary prevention of cardiovascular disease. However, statin use is often associated with a variety of muscle-related symptoms or myopathies. Myopathy may be related in part to statin inhibition of the endogenous synthesis of coenzyme Q10, an essential cofactor for mitochondrial energy production. The aim of this study is to determine whether coenzyme Q10 supplementation would reduce the degree of muscle pain associated with statin treatment. Patients with myopathic symptoms were randomly assigned in a double-blinded protocol to treatment with coenzyme Q10 (100 mg/day, n = 18) or vitamin E (400 IU/day, n = 14) for 30 days. Muscle pain and pain interference with daily activities were assessed before and after treatment. After a 30-day intervention, pain severity decreased by 40% (p <0.001) and pain interference with daily activities decreased by 38% (p <0.02) in the group treated with coenzyme Q10. In contrast, no changes in pain severity (+9%, p = NS) or pain interference with daily activities (-11%, p = NS) was observed in the group treated with vitamin E. In conclusion, results suggest that coenzyme Q10 supplementation may decrease muscle pain associated with statin treatment. Thus, coenzyme Q10 supplementation may offer an alternative to stopping treatment with these vital drugs.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscular Diseases/chemically induced , Muscular Diseases/drug therapy , Ubiquinone/analogs & derivatives , Vitamins/pharmacology , Activities of Daily Living , Aged , Biomarkers/blood , Cholesterol, LDL/blood , Cholesterol, LDL/drug effects , Coenzymes/drug effects , Coenzymes/pharmacology , Coenzymes/therapeutic use , Creatine Kinase/blood , Creatine Kinase/drug effects , Double-Blind Method , Female , Humans , Hypercholesterolemia/drug therapy , Male , Middle Aged , Muscular Diseases/physiopathology , Pain/chemically induced , Pain/drug therapy , Pain/physiopathology , Pain Measurement , Patient Compliance , Severity of Illness Index , Surveys and Questionnaires , Treatment Outcome , Triglycerides/blood , Ubiquinone/drug effects , Ubiquinone/pharmacology , Ubiquinone/therapeutic use , Vitamin E/therapeutic use , Vitamins/therapeutic use
14.
Mitochondrion ; 7 Suppl: S168-74, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17482884

ABSTRACT

Statins are drugs of known and undisputed efficacy in the treatment of hypercholesterolemia, usually well tolerated by most patients. In some cases treatment with statins produces skeletal muscle complaints, and/or mild serum CK elevation; the incidence of rhabdomyolysis is very low. As a result of the common biosynthetic pathway Coenzyme Q (ubiquinone) and dolichol levels are also affected, to a certain degree, by the treatment with these HMG-CoA reductase inhibitors. Plasma levels of CoQ10 are lowered in the course of statin treatment. This could be related to the fact that statins lower plasma LDL levels, and CoQ10 is mainly transported by LDL, but a decrease is also found in platelets and in lymphocytes of statin treated patients, therefore it could truly depend on inhibition of CoQ10 synthesis. There are also some indications that statin treatment affects muscle ubiquinone levels, although it is not yet clear to which extent this depends on some effect on mitochondrial biogenesis. Some papers indicate that CoQ10 depletion during statin therapy might be associated with subclinical cardiomyopathy and this situation is reversed upon CoQ10 treatment. We can reasonably hypothesize that in some conditions where other CoQ10 depleting situations exist treatment with statins may seriously impair plasma and possible tissue levels of coenzyme Q10. While waiting for a large scale clinical trial where patients treated with statins are also monitored for their CoQ10 status, with a group also being given CoQ10, physicians should be aware of this drug-nutrient interaction and be vigilant to the possibility that statin drugs may, in some cases, impair skeletal muscle and myocardial bioenergetics.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Ubiquinone/analogs & derivatives , Animals , Biochemistry/methods , Biosynthetic Pathways , Clinical Trials as Topic , Coenzymes/physiology , Coenzymes/therapeutic use , Cricetinae , Dogs , Heart Diseases/drug therapy , Heart Diseases/metabolism , Humans , Hyperlipidemias/drug therapy , Models, Biological , Rats , Ubiquinone/metabolism , Ubiquinone/physiology , Ubiquinone/therapeutic use , Vitamins/therapeutic use
15.
Mitochondrion ; 7 Suppl: S136-45, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17485245

ABSTRACT

The evidence supporting a treatment benefit for coenzyme Q10 (CoQ10) in primary mitochondrial disease (mitochondrial disease) whilst positive is limited. Mitochondrial disease in this context is defined as genetic disease causing an impairment in mitochondrial oxidative phosphorylation (OXPHOS). There are no treatment trials achieving the highest Level I evidence designation. Reasons for this include the relative rarity of mitochondrial disease, the heterogeneity of mitochondrial disease, the natural cofactor status and easy 'over the counter availability' of CoQ10 all of which make funding for the necessary large blinded clinical trials unlikely. At this time the best evidence for efficacy comes from controlled trials in common cardiovascular and neurodegenerative diseases with mitochondrial and OXPHOS dysfunction the etiology of which is most likely multifactorial with environmental factors playing on a background of genetic predisposition. There remain questions about dosing, bioavailability, tissue penetration and intracellular distribution of orally administered CoQ10, a compound which is endogenously produced within the mitochondria of all cells. In some mitochondrial diseases and other commoner disorders such as cardiac disease and Parkinson's disease low mitochondrial or tissue levels of CoQ10 have been demonstrated providing an obvious rationale for supplementation. This paper discusses the current state of the evidence supporting the use of CoQ10 in mitochondrial disease.


Subject(s)
Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/metabolism , Oxygen/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism , Ubiquinone/therapeutic use , Animals , Antioxidants , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Coenzymes/chemistry , Coenzymes/therapeutic use , Evidence-Based Medicine , Guinea Pigs , Humans , Mitochondria/metabolism , Models, Biological , Oxidative Phosphorylation , Phosphorylation , Rats , Ubiquinone/chemistry
16.
Mitochondrion ; 7 Suppl: S154-67, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17485243

ABSTRACT

In this review we summarise the current state of knowledge of the therapeutic efficacy and mechanisms of action of CoQ(10) in cardiovascular disease. Our conclusions are: 1. There is promising evidence of a beneficial effect of CoQ(10) when given alone or in addition to standard therapies in hypertension and in heart failure, but less extensive evidence in ischemic heart disease. 2. Large scale multi-centre prospective randomised trials are indicated in all these areas but there are difficulties in funding such trials. 3. Presently, due to the notable absence of clinically significant side effects and likely therapeutic benefit, CoQ(10) can be considered a safe adjunct to standard therapies in cardiovascular disease.


Subject(s)
Cardiovascular Diseases/drug therapy , Ubiquinone/analogs & derivatives , Adenosine Triphosphate/chemistry , Anthracyclines/metabolism , Antioxidants/metabolism , Clinical Trials as Topic , Coenzymes/metabolism , Coenzymes/physiology , Coenzymes/therapeutic use , Diet , Heart Failure/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Hypertension/drug therapy , Ischemia/pathology , Mitochondria/metabolism , Models, Biological , Ubiquinone/chemistry , Ubiquinone/metabolism , Ubiquinone/physiology , Ubiquinone/therapeutic use
17.
Mitochondrion ; 7 Suppl: S127-35, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17485244

ABSTRACT

Since the identification of the genetic mutation causing Friedreich's ataxia (FRDA) our understanding of the mechanisms underlying disease pathogenesis have improved markedly. The genetic abnormality results in the deficiency of frataxin, a protein targeted to the mitochondrion. There is extensive evidence that mitochondrial respiratory chain dysfunction, oxidative damage and iron accumulation play significant roles in the disease mechanism. There remains considerable debate as to the normal function of frataxin, but it is likely to be involved in mitochondrial iron handling, antioxidant regulation, and/or iron sulphur centre regulation. Therapeutic avenues for patients with FRDA are beginning to be explored in particular targeting antioxidant protection, enhancement of mitochondrial oxidative phosphorylation, iron chelation and more recently increasing FRDA transcription. The use of quinone therapy has been the most extensively studied to date with clear benefits demonstrated using evaluations of both disease biomarkers and clinical symptoms, and this is the topic that will be covered in this review.


Subject(s)
Friedreich Ataxia/diagnosis , Friedreich Ataxia/genetics , Ubiquinone/analogs & derivatives , Vitamin E/therapeutic use , Animals , Ataxia/metabolism , Benzoquinones/therapeutic use , Coenzymes/genetics , Coenzymes/metabolism , Coenzymes/therapeutic use , Disease Models, Animal , Humans , Iron/metabolism , Mutation , Neurodegenerative Diseases/metabolism , Oxidative Stress , Oxygen/metabolism , Quinones/metabolism , Time Factors , Ubiquinone/genetics , Ubiquinone/metabolism , Ubiquinone/therapeutic use , Vitamin E/metabolism
18.
Mitochondrion ; 7 Suppl: S175-80, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17442628

ABSTRACT

Mevalonic aciduria (MVA) and phenylketonuria (PKU) are inborn errors of metabolism caused by deficiencies in the enzymes mevalonate kinase and phenylalanine 4-hydroxylase, respectively. Despite numerous studies the factors responsible for the pathogenicity of these disorders remain to be fully characterised. In common with MVA, a deficit in coenzyme Q10 (CoQ10) concentration has been implicated in the pathophysiology of PKU. In MVA the decrease in CoQ10 concentration may be attributed to a deficiency in mevalonate kinase, an enzyme common to both CoQ10 and cholesterol synthesis. However, although dietary sources of cholesterol cannot be excluded, the low/normal cholesterol levels in MVA patients suggests that some other factor may also be contributing to the decrease in CoQ10.The main factor associated with the low CoQ10 level of PKU patients is purported to be the elevated phenylalanine level. Phenylalanine has been shown to inhibit the activities of both 3-hydroxy-3-methylglutaryl-CoA reductase and mevalonate-5-pyrophosphate decarboxylase, enzymes common to both cholesterol and CoQ10 biosynthesis. Although evidence of a lowered plasma/serum CoQ10 level has been reported in MVA and PKU, few studies have assessed the intracellular CoQ10 concentration of patients. Plasma/serum CoQ10 is influenced by dietary intake as well as its lipoprotein content and therefore may be limited as a means of assessing intracellular CoQ10 concentration. Whether the pathogenesis of MVA and PKU are related to a loss of CoQ10 has yet to be established and further studies are required to assess the intracellular CoQ10 concentration of patients before this relationship can be confirmed or refuted.


Subject(s)
Mevalonic Acid/metabolism , Phenylketonurias/drug therapy , Ubiquinone/analogs & derivatives , Amino Acid Metabolism, Inborn Errors/drug therapy , Animals , Antioxidants/metabolism , Coenzymes/metabolism , Coenzymes/physiology , Coenzymes/therapeutic use , Humans , Hydroxymethylglutaryl CoA Reductases/metabolism , Metabolic Networks and Pathways , Mitochondrial Diseases/drug therapy , Models, Biological , Oxidative Stress , Phenylalanine/metabolism , Ubiquinone/metabolism , Ubiquinone/physiology , Ubiquinone/therapeutic use
19.
Nutr Rev ; 65(6 Pt 1): 286-93, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17605305

ABSTRACT

Cardiovascular disease (CVD) is the leading cause of morbidity and mortality in the Western world. Oxidative stress appears to play a pivotal role in atherosclerosis. Coenzyme Q10 (CoQ10), one of the most important antioxidants, is synthesized de novo by every cell in the body. Its biosynthesis decreases with age and its deficit in tissues is associated with degenerative changes of aging, thus implicating a possible therapeutic role of CoQl0 in human diseases. There is evidence to support the therapeutic value of CoQ10 as an adjunct to standard medical therapy in congestive heart failure. However, much further research is required, especially in the use of state-of-the-art techniques to assess functional outcomes in patients with congestive heart failure.


Subject(s)
Antioxidants/physiology , Cardiac Output, Low/drug therapy , Heart Failure/prevention & control , Ubiquinone/analogs & derivatives , Vitamins/physiology , Aging/physiology , Antioxidants/therapeutic use , Coenzymes/physiology , Coenzymes/therapeutic use , Drug Interactions , Humans , Oxidative Stress/drug effects , Ubiquinone/physiology , Ubiquinone/therapeutic use , Vitamins/therapeutic use
20.
Mol Biotechnol ; 37(1): 31-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17914161

ABSTRACT

For a number of years, coenzyme Q (CoQ10 in humans) was known for its key role in mitochondrial bioenergetics; later studies demonstrated its presence in other subcellular fractions and in plasma, and extensively investigated its antioxidant role. These two functions constitute the basis on which research supporting the clinical use of CoQ10 is founded. Also at the inner mitochondrial membrane level, coenzyme Q is recognized as an obligatory co-factor for the function of uncoupling proteins and a modulator of the transition pore. Furthermore, recent data reveal that CoQ10 affects expression of genes involved in human cell signalling, metabolism, and transport and some of the effects of exogenously administered CoQ10 may be due to this property. Coenzyme Q is the only lipid soluble antioxidant synthesized endogenously. In its reduced form, CoQH2, ubiquinol, inhibits protein and DNA oxidation but it is the effect on lipid peroxidation that has been most deeply studied. Ubiquinol inhibits the peroxidation of cell membrane lipids and also that of lipoprotein lipids present in the circulation. Dietary supplementation with CoQ10 results in increased levels of ubiquinol-10 within circulating lipoproteins and increased resistance of human low-density lipoproteins to the initiation of lipid peroxidation. Moreover, CoQ10 has a direct anti-atherogenic effect, which has been demonstrated in apolipoprotein E-deficient mice fed with a high-fat diet. In this model, supplementation with CoQ10 at pharmacological doses was capable of decreasing the absolute concentration of lipid hydroperoxides in atherosclerotic lesions and of minimizing the size of atherosclerotic lesions in the whole aorta. Whether these protective effects are only due to the antioxidant properties of coenzyme Q remains to be established; recent data point out that CoQ10 could have a direct effect on endothelial function. In patients with stable moderate CHF, oral CoQ10 supplementation was shown to ameliorate cardiac contractility and endothelial dysfunction. Recent data from our laboratory showed a strong correlation between endothelium bound extra cellular SOD (ecSOD) and flow-dependent endothelial-mediated dilation, a functional parameter commonly used as a biomarker of vascular function. The study also highlighted that supplementation with CoQ10 that significantly affects endothelium-bound ecSOD activity. Furthermore, we showed a significant correlation between increase in endothelial bound ecSOD activity and improvement in FMD after CoQ10 supplementation. The effect was more pronounced in patients with low basal values of ecSOD. Finally, we summarize the findings, also from our laboratory, on the implications of CoQ10 in seminal fluid integrity and sperm cell motility.


Subject(s)
Antioxidants/metabolism , Antioxidants/pharmacology , Atherosclerosis/prevention & control , Infertility, Male/enzymology , Ubiquinone/analogs & derivatives , Animals , Antioxidants/therapeutic use , Atherosclerosis/drug therapy , Coenzymes/metabolism , Coenzymes/pharmacology , Coenzymes/therapeutic use , Endothelium, Vascular/enzymology , Energy Metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Infertility, Male/drug therapy , Lipid Metabolism , Lipids/blood , Male , Mice , Mitochondria/enzymology , Superoxide Dismutase/metabolism , Ubiquinone/metabolism , Ubiquinone/pharmacology , Ubiquinone/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL