Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 194
Filter
Add more filters

Publication year range
1.
Bioorg Chem ; 105: 104329, 2020 12.
Article in English | MEDLINE | ID: mdl-33068813

ABSTRACT

CoQ10 and Vitamin E are used in medicinal applications, but they are both lipophilic molecules and the poor solubility in aqueous media results in an inefficient administration, poor bioavailability and potential toxicity. A mixed conjugate Ubiquinol-Polyethylene glycol-Vitamin E was synthesized and characterized to improve the bioavailability of CoQ10 and Vitamin E. The synthesized mixed PEG conjugate was characterized by 1H NMR spectroscopy and MALDI spectrometry. The in vitro release of the conjugate was measured at various pH conditions and in human plasma and the evaluation of free CoQ10 and Vitamin E were also conducted. The obtained results demonstrated that more CoQ10 and Vitamin E were released from PEG conjugate at pH 7.4 and in plasma within the 24 h. The antioxidant activity evaluation was carried out by DPPH assay. Our results indicated that the chemical modification after esterification with PEG of the two drugs Ubiquinol and Vitamin E doesn't significantly affected their antioxidant potential.


Subject(s)
Antioxidants/chemistry , Drug Carriers/chemistry , Polyethylene Glycols/chemistry , Ubiquinone/analogs & derivatives , Vitamin E/chemistry , Antioxidants/pharmacology , Biological Availability , Drug Compounding , Drug Liberation , Drug Therapy, Combination , Humans , Solubility , Succinic Acid/chemistry , Ubiquinone/chemistry , Ubiquinone/pharmacokinetics , Vitamin E/pharmacokinetics
2.
Skin Pharmacol Physiol ; 33(6): 293-299, 2020.
Article in English | MEDLINE | ID: mdl-33212439

ABSTRACT

INTRODUCTION: Coenzyme Q10 (CoQ10) has been widely used in topical and cosmeceutical products due to its cutaneous antioxidant and energizer effects. CoQ10 is found in a higher concentration in the epidermis compared to dermis. The epidermal level of CoQ10 can be reduced due to several factors such as skin UV irradiation and photoaging. Various dermal nano-formulations have been investigated to overcome the skin barrier and enhance the poor penetration of CoQ10. The nanocarriers are designed to target and concentrate the CoQ10 in the viable epidermis. Most of these studies, however, failed to show the depth and extent of penetration of CoQ10 from the various carrier systems. OBJECTIVE: The distribution of CoQ10 across the various skin layers has to be shown using skin slices representing the different skin layers. METHODS: To realize this objective, a sensitive and selective HPLC method was developed and validated for the quantification of CoQ10 in the different skin slices. The method applicability to skin penetration (using excised human skin) as well as stability studies was investigated using CoQ10-loaded lecithin-based microemulsion (ME) and hydrophilic cream formulations. RESULTS: It could be shown that the highest concentration of CoQ10 in the viable epidermis, the target skin layer for CoQ10, was observed after application of the CoQ10 in the hydrophilic cream. This cream contains 10% of 2-ethylhexyl laurate which works obviously as a penetration enhancer for CoQ10. In contrast, the penetration of CoQ10 was lower from the ME. Just in the deeper dermis, a certain amount of CoQ10 could be detected. CONCLUSIONS: The HPLC method quantified the trace quantities of the CoQ10 distributed across the various skin layers and, hence, can be used to investigate the skin penetration of CoQ10 from various dermal standard and nano-formulations.


Subject(s)
Skin/metabolism , Ubiquinone/analogs & derivatives , Administration, Cutaneous , Drug Compounding/methods , Emulsions/administration & dosage , Emulsions/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Ointments , Permeability , Skin Absorption/drug effects , Ubiquinone/administration & dosage , Ubiquinone/chemistry , Ubiquinone/pharmacokinetics , Vitamins/administration & dosage , Vitamins/pharmacokinetics
3.
Int J Mol Sci ; 21(22)2020 Nov 10.
Article in English | MEDLINE | ID: mdl-33182646

ABSTRACT

The aim of this review is to shed light over the most recent advances in Coenzyme Q10 (CoQ10) applications as well as to provide detailed information about the functions of this versatile molecule, which have proven to be of great interest in the medical field. Traditionally, CoQ10 clinical use was based on its antioxidant properties; however, a wide range of highly interesting alternative functions have recently been discovered. In this line, CoQ10 has shown pain-alleviating properties in fibromyalgia patients, a membrane-stabilizing function, immune system enhancing ability, or a fundamental role for insulin sensitivity, apart from potentially beneficial properties for familial hypercholesterolemia patients. In brief, it shows a remarkable amount of functions in addition to those yet to be discovered. Despite its multiple therapeutic applications, CoQ10 is not commonly prescribed as a drug because of its low oral bioavailability, which compromises its efficacy. Hence, several formulations have been developed to face such inconvenience. These were initially designed as lipid nanoparticles for CoQ10 encapsulation and distribution through biological membranes and eventually evolved towards chemical modifications of the molecule to decrease its hydrophobicity. Some of the most promising formulations will also be discussed in this review.


Subject(s)
Ubiquinone/analogs & derivatives , Administration, Oral , Antioxidants/administration & dosage , Antioxidants/pharmacokinetics , Antioxidants/therapeutic use , Biological Availability , Drug Compounding/methods , Drug Delivery Systems , Humans , Liposomes , Solubility , Ubiquinone/administration & dosage , Ubiquinone/pharmacokinetics , Ubiquinone/therapeutic use
4.
Compr Rev Food Sci Food Saf ; 19(2): 574-594, 2020 03.
Article in English | MEDLINE | ID: mdl-33325173

ABSTRACT

World population growth and aging are posing unprecedented challenges in sustaining the health of 9.1 billion people that will be occupying the planet by 2050. Although noncommunicable diseases such as cardiovascular and neurodegenerative diseases, cancer, and diabetes are among the top 10 global causes of death, they can be prevented by risk factor reduction, early detection, and adequate treatment. Since a healthy diet along with dietary supplementation could play an important role to reduce morbidity and cut off its associated health care costs, research in the food and nutrition area is required to find solutions to global challenges affecting health. As a result of the healthy living trend, dietary supplements category is growing fast, leading to an urgent need for dietitians, physicians, and policy makers to broaden the scientific evidence on the efficacy and safety of a wide range of active ingredients. Coenzyme Q10 (CoQ10), as the third most consumed dietary supplement, and as a potential candidate for the treatment of various noncommunicable diseases that are among the global top 10 causes of death, has gained interest over years. Scientific evidence regarding mainly CoQ10 efficacy and safety, as well as formulation challenges, is addressed in this review.


Subject(s)
Ubiquinone/analogs & derivatives , Antioxidants , Dietary Supplements , Humans , Ubiquinone/pharmacokinetics , Ubiquinone/pharmacology , Ubiquinone/therapeutic use
5.
Biochem Biophys Res Commun ; 512(3): 611-615, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30914201

ABSTRACT

Coenzyme Q10 (CoQ10) plays a key role not only as an essential electron carrier in the mitochondrial electron transport chain, but also as an antioxidant to protect cells from oxidative stress. CoQ10 supplementation is expected to be effective for a variety of diseases. The predominant forms of CoQ10 are the ubiquinol-10 (reduced form) and ubiquinone-10 (oxidized form). Both forms of CoQ10 supplements are commercially available, however, their kinetic difference is still unclear. In order to conduct in vivo analysis of the kinetics of ubiquinol-10 and ubiquinone-10, we succeeded in synthesizing 11C-labeled ubiquinol-10 ([11C]UQL) and ubiquinone-10 ([11C]UQN), respectively. In the present study, we aimed to investigate the kinetics of [11C]UQL and [11C]UQN, both of which were administered via the tail vein of 8-week-old male Sprague-Dawley rats. Whole-body positron emission tomography (PET) imaging was performed to follow the time course of accumulation in the liver, spleen, brain, and other organs. Then, at the two typical time points at 20 or 90 min after injection, we conducted the biodistribution study. Various organs/tissues and blood were collected, weighed and counted with a gamma counter. Percent injected dose per gram of tissue (%ID/g) was calculated as the indicator of the accumulation of each compound. As the results, at both time points, %ID/g of [11C]UQL in the cerebrum, cerebellum, white adipose tissue, muscle, kidney, and testis were higher (P < 0.05) than that of [11C]UQN: at 90-min time point, %ID/g of [11C]UQL in the brown adipose tissue was higher (P < 0.05) than that of [11C]UQN: on the contrary, %ID/g of [11C]UQL in the spleen was lower (P < 0.05) than that of [11C]UQN at 90 min. In a separate study of the metabolite analysis in the plasma, UQL injected into the tail vein of rats was almost unchanged during the PET scanning time, but UQN was gradually converted to the reduced form UQL. Therefore, the uptake values of UQL into the tissues and organs were rather accurate but those of UQN might be the sum of UQN uptake and partly converted UQL uptake. These studies suggested that the accumulation level of administered CoQ10 differs depending on its redox state, and that CoQ10 redox state could be crucial for optimization of the effective supplementation.


Subject(s)
Antioxidants/pharmacokinetics , Ubiquinone/analogs & derivatives , Animals , Dietary Supplements/analysis , Male , Oxidation-Reduction , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley , Tissue Distribution , Ubiquinone/pharmacokinetics
6.
J Hum Genet ; 64(4): 297-304, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30659264

ABSTRACT

COQ4 mutations have recently been shown to cause a broad spectrum of mitochondrial disorders in association with CoQ10 deficiency. Herein, we report the clinical phenotype, in silico and biochemical analyses, and intervention for a novel c.370 G > A (p.G124S) COQ4 mutation in a Chinese family. This mutation is exclusively present in the East Asian population (allele frequency of ~0.001). The homozygous mutation caused CoQ10 deficiency-associated Leigh syndrome with an onset at 1-2 months of age, presenting as respiratory distress, lactic acidosis, dystonia, seizures, failure to thrive, and detectable lesions in the midbrain and basal ganglia. No renal impairment was involved. The levels of CoQ10 and mitochondrial respiratory chain complex (C) II + III activity were clearly lower in cultured fibroblasts derived from the patient than in those from unaffected carriers; the decreased CII + III activity could be increased by CoQ10 treatment. Follow-up studies suggested that our patient benefitted from the oral supplementation of CoQ10, which allowed her to maintain a relatively stable health status. Based on the genetic testing, preimplantation and prenatal diagnoses were performed, confirming that the next offspring of this family was unaffected. Our cases expand the phenotypic spectrum of COQ4 mutations and the genotypic spectrum of Leigh syndrome.


Subject(s)
Ataxia/genetics , Genetic Testing , Leigh Disease/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Muscle Weakness/genetics , Ubiquinone/deficiency , Asian People/genetics , Ataxia/complications , Child, Preschool , Computer Simulation , Female , Fibroblasts/metabolism , Heterozygote , Homozygote , Humans , Infant , Leigh Disease/complications , Leigh Disease/physiopathology , Male , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/complications , Muscle Weakness/complications , Mutation , Phenotype , Ubiquinone/genetics , Ubiquinone/pharmacokinetics
7.
Toxicol Appl Pharmacol ; 370: 78-92, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30836114

ABSTRACT

Mitochondrial dysfunctions induced by oxidative stress could play a pivotal role in the development of testicular damage and degeneration, leading to impaired fertility in adulthood. MitoQ as mitochondria-targeted antioxidant has been used in many diseases for a long time, but its therapeutic effects on testicular injury 'have not been reported yet. Here, we examined the protective action mechanism of MitoQ on testicular injury from oxidative stress induced by triptolide (TP). Mice were orally administrated with MitoQ (1.3, 2.6 and 5 .2mg/kg, respectively) in a TP-induced model of testicular damage for 14 days. And then testis injuries were comprehensively evaluated in terms of morphological changes, spermatogenesis assessment, blood-testis barrier (BTB) integrity, and apoptosis. The results demonstrated MitoQ effectively increased testicular weight, maintained the integrity of BTB, protected microstructure of testicular tissue and sperm morphology by inhibition of oxidative stress. Further mechanism studies revealed that MitoQ markedly activates the Keap1-Nrf2 antioxidative defense system characterized by increasing the expression of Nrf2 and its target genes HO-1 and NQO1. Meanwhile, MitoQ upregulated the expression of mitochondrial dynamics proteins Mfn2 and Drp-1and exerted a protective effect on mitochondria. On this basis, the results from pharmacokinetic study indicated that the MitoQ could enter into testis tissues after oral administration in despite of the low absolute bioavailability, which provided the material basis for MitoQ in the treatment of testicular damage. More importantly, MitoQ reached mitochondria quickly and had an outstanding feature of mitochondria targeting in Sertoli cells. Therefore, these results provide information for the application of MitoQ against testicular injury diseases.


Subject(s)
Kelch-Like ECH-Associated Protein 1/metabolism , Mitochondria/drug effects , NF-E2-Related Factor 2/metabolism , Organophosphorus Compounds/administration & dosage , Signal Transduction/drug effects , Testicular Diseases/prevention & control , Ubiquinone/analogs & derivatives , Animals , Antioxidants/pharmacology , Biological Availability , Blood-Testis Barrier/drug effects , Gene Expression/drug effects , Male , Mice , Mitochondria/physiology , NF-E2-Related Factor 2/genetics , Organophosphorus Compounds/pharmacokinetics , Oxidation-Reduction , Sertoli Cells/ultrastructure , Signal Transduction/physiology , Spermatogenesis/drug effects , Testicular Diseases/pathology , Testicular Diseases/physiopathology , Ubiquinone/administration & dosage , Ubiquinone/pharmacokinetics
8.
Drug Dev Ind Pharm ; 45(9): 1451-1458, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31216907

ABSTRACT

Objective: The aim of this study was to develop a coenzyme Q10 nanoemulsion cream, characterize and to determine the influence of omega fatty acids on the delivery of coenzyme Q10 across model skin membrane via ex vivo and in silico techniques. Methods: Coenzyme Q10 nanoemulsion creams were prepared using natural edible oils such as linseed, evening primrose, and olive oil. Their mechanical features and ability to deliver CoQ10 across rat skin were characterized. Computational docking analysis was performed for in silico evaluation of CoQ10 and omega fatty acid interactions. Results: Linseed, evening primrose, and olive oils each produced nano-sized emulsion creams (343.93-409.86 nm) and exhibited excellent rheological features. The computerized docking studies showed favorable interactions between CoQ10 and omega fatty acids that could improve skin permeation. The three edible-oil nanoemulsion creams displayed higher ex vivo skin permeation and drug flux compared to the liquid-paraffin control cream. The linseed oil formulation displayed the highest skin permeation (3.97 ± 0.91 mg/cm2) and drug flux (0.19 ± 0.05 mg/cm2/h). Conclusion: CoQ10 loaded-linseed oil nanoemulsion cream displayed the highest skin permeation. The highest permeation showed by linseed oil nanoemulsion cream may be due to the presence of omega-3, -6, and -9 fatty acids which might serve as permeation enhancers. This indicated that the edible oil nanoemulsion creams have potential as drug vehicles that enhance CoQ10 delivery across skin.


Subject(s)
Drug Carriers/chemistry , Fatty Acids, Unsaturated/chemistry , Skin Cream/pharmacokinetics , Ubiquinone/analogs & derivatives , Administration, Cutaneous , Animals , Computer Simulation , Drug Compounding , Emulsions , Nanoparticles/chemistry , Permeability , Rats , Skin/drug effects , Skin/metabolism , Skin Absorption , Skin Cream/administration & dosage , Ubiquinone/administration & dosage , Ubiquinone/pharmacokinetics
9.
Biochim Biophys Acta Biomembr ; 1860(5): 1205-1215, 2018 May.
Article in English | MEDLINE | ID: mdl-29470946

ABSTRACT

Ubiquinone-10 (Q10) plays a pivotal role as electron-carrier in the mitochondrial respiratory chain, and is also well known for its powerful antioxidant properties. Recent findings suggest moreover that Q10 could have an important membrane stabilizing function. In line with this, we showed in a previous study that Q10 decreases the permeability to carboxyfluorescein (CF) and increases the mechanical strength of 1-palmitoyl-2-oleyl-sn-glycero-phosphocholine (POPC) membranes. In the current study we report on the effects exerted by Q10 in membranes having a more complex lipid composition designed to mimic that of the inner mitochondrial membrane (IMM). Results from DPH fluorescence anisotropy and permeability measurements, as well as investigations probing the interaction of liposomes with silica surfaces, corroborate a membrane stabilizing effect of Q10 also in the IMM-mimicking membranes. Comparative investigations examining the effect of Q10 and the polyisoprenoid alcohol solanesol on the IMM model and on membranes composed of individual IMM components suggest, moreover, that Q10 improves the membrane barrier properties via different mechanisms depending on the lipid composition of the membrane. Thus, whereas Q10's inhibitory effect on CF release from pure POPC membranes appears to be directly and solely related to Q10's lipid ordering and condensing effect, a mechanism linked to Q10's ability to amplify intrinsic curvature elastic stress dominates in case of membranes containing high proportions of palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine (POPE).


Subject(s)
Lipid Bilayers/chemistry , Mitochondrial Membranes/chemistry , Mitochondrial Membranes/drug effects , Ubiquinone/pharmacology , Adsorption , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Cell Membrane Permeability/drug effects , Lipid Bilayers/metabolism , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Mitochondrial Membranes/metabolism , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/pharmacokinetics , Terpenes/chemistry , Terpenes/pharmacology , Ubiquinone/pharmacokinetics
10.
Drug Dev Ind Pharm ; 44(2): 289-295, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29082762

ABSTRACT

OBJECTIVE: In this study, solid dispersion (SD) for oral delivery of a poorly water-soluble drug, coenzyme Q10 was developed by supercritical fluid technology and characterized in vitro and in vivo. METHODS: Dissolution was used to optimize the formulations of CoQ10-SD. The physicochemical properties of SD were investigated by differential scanning calorimetry (DSC), powder X-ray diffraction (PXRD), and scanning electron microscopy (SEM). The supercritical fluid chromatography-electrospray ionization tandem mass spectrometry (SFC-ESI-MS/MS) was used for the in vivo study. RESULTS: The results of DSC and PXRD indicated that the drug in SD was in amorphous state. In vitro drug release, the dissolution of coenzyme Q10 in solid dispersion improved to 78.8% compared with commercial tablets of 0.16%. The area under c-t curve (AUC0-72h) and mean maximum concentrations (Cmax) of CoQ10-SD were 2.43-fold and 3.0-fold, respectively higher than that of commercial tablets in rats, confirming improved bioavailability. CONCLUSION: Supercritical fluid technology was successfully used for the preparation and analysis of CoQ10-SD for the first time and significantly improved the dissolution and bioavailability of coenzyme Q10.


Subject(s)
Silicon Dioxide/chemistry , Technology, Pharmaceutical/methods , Ubiquinone/analogs & derivatives , Administration, Oral , Animals , Area Under Curve , Calorimetry, Differential Scanning , Chromatography, Supercritical Fluid , Drug Liberation , Half-Life , Male , Metabolic Clearance Rate , Microscopy, Electron, Scanning , Powder Diffraction , Pressure , Rats , Solubility , Tandem Mass Spectrometry , Temperature , Time Factors , Ubiquinone/administration & dosage , Ubiquinone/pharmacokinetics
11.
Pharmazie ; 72(12): 714-720, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29441955

ABSTRACT

Methoxy polyethylene glycol conjugated with coenzyme Q10 (mPEG)-CoQ10 and analog adducts with amino acids as spacers were synthesized as a new drug delivery systems for CoQ10. Alanine and branched chain amino acids (valine, leucine and isoleucine) were conjugated to mPEG by an amide linkage and to CoQ10 by an ester bond. Recently, branched chain amino acids (BCAAs), which are released along with CoQ10, have received increasing attention as 'anti-fatigue' elements. FT-IR and 1H NMR spectroscopic analysis were useful to characterize the synthesized conjugates. Studies in vitro, in buffer solutions at different pH and in the presence of esterase were conducted. The hydrolysis studies showed a specific cleavage dependent on the pH of the medium and by the presence of proteolytic enzymes. The results showed the improvement of the pharmacokinetic properties of CoQ10. The antioxidant activity of the synthesized conjugates was also evaluated by DPPH assay.


Subject(s)
Antioxidants/administration & dosage , Drug Delivery Systems , Polyethylene Glycols/chemistry , Ubiquinone/analogs & derivatives , Amino Acids/chemistry , Antioxidants/chemistry , Antioxidants/pharmacokinetics , Drug Liberation , Esterases/metabolism , Hydrogen-Ion Concentration , Hydrolysis , Proton Magnetic Resonance Spectroscopy , Spectroscopy, Fourier Transform Infrared , Ubiquinone/administration & dosage , Ubiquinone/chemistry , Ubiquinone/pharmacokinetics
12.
AAPS PharmSciTech ; 18(5): 1657-1672, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27677262

ABSTRACT

The aim of our investigation is to develop and characterize self-nanoemulsifying drug delivery systems (SNEDDS) of CoQ10 to improve its water solubility, dissolution rate, and bioavailability, and then evaluate its biochemical and physiological effect on liver cirrhosis in rats compared with CoQ10 powder. SNEDDS are isotropic and thermodynamically stable mixture of oil, surfactant, co-surfactant, and drug that form an oil/water nanoemulsion when added to aqueous phases with soft agitation. Upon administration, self-nanoemulsifying system becomes in contact with gastrointestinal fluid and forms o/w nanoemulsion by the aid of gastrointestinal motility. When the nanoemulsion is formed in the gastrointestinal tract, it presents the drug in a solubilized form inside small nano-sized droplets that provide a large surface area for enhancing the drug release and absorption. Solubility of CoQ10 in various oils, surfactants, and co-surfactants were studied to identify the components of SNEDDS; pseudo-ternary phase diagrams were plotted to identify the efficient self-emulsifying regions. CoQ10-loaded SNEDDS were prepared using isopropyl myristate as oil; Cremophor El, Labrasol, or Tween80 as surfactant; and Transcutol as co-surfactant. The amount of CoQ10 in each vehicle was 3%. The formulations that passed thermostability evaluation test were assessed for particle size analysis, morphological characterization, refractive index, zeta potential, viscosity, electroconductivity, drug release profile, as well as ex vivo permeability. Pharmacokinetics and hepatoprotective efficiency of the optimized SNEDDS of CoQ10 compared with CoQ10 suspension were performed. Results showed that all optimized formulae have the ability to form a good and stable nanoemulsion when diluted with water; the mean droplet size of all formulae was in the nanometric range (11.7-13.5 nm) with optimum polydispersity index values (0.2-0.21). All formulae showed negative zeta potential (-11.3 to -17.2), and maximum drug loading efficiency. One hundred percent of CoQ10 was released from most formulae within 30 min. One hundred percent of CoQ10 was permeated from all formulae through 10 h. The pharmacokinetic study in rabbits revealed a significant increase in bioavailability of CoQ10 SNEDDS to 2.1-fold compared with CoQ10 suspension after oral administration. Comparative effect of the optimized formulae on acute liver injury compared with CoQ10 powder was also studied; it was found that all the liver biochemical markers as alanine transferase (ALT), aspartate amino transferase (AST), alkaline phosphatase (ALP), total protein (TP), and albumin were significantly improved at p < 0.05. Also, histochemical and histopthological studies confirm the biochemical results. Our results suggest the potential use of SNEDDS to increase the solubility of liphophilic drug as poorly water-soluble CoQ10 and improve its oral absorption, so it can be more efficient to improve liver damage compared to CoQ10 powder. These results demonstrated that CoQ10 SNEDDS inhibited thioacetamide (TAA)-induced liver fibrosis mainly through suppression of collagen production.


Subject(s)
Drug Delivery Systems , Liver Cirrhosis, Experimental/drug therapy , Ubiquinone/analogs & derivatives , Administration, Oral , Animals , Biological Availability , Drug Liberation , Emulsions/chemistry , Male , Rabbits , Rats , Solubility , Ubiquinone/chemistry , Ubiquinone/pharmacokinetics , Ubiquinone/therapeutic use
13.
AAPS PharmSciTech ; 17(3): 757-66, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26340950

ABSTRACT

To enhance the stability of coenzyme Q10 (CoQ10), Kolliphor® HS 15 (HS15) was employed as a carrier to build up a stable CoQ10-loaded micelle delivery system. The impact of micellar compositions, the preparation condition, and the preparation method on size characteristics, the solubilization efficiency, and micellar stability were investigated. The optimal preparation conditions were 1:6, 4, 0.2%, 118°C, and 25 min for CoQ10/HS15 mass ratio, pH value, the concentration of glucose, and the sterilization conditions. Upon these conditions, the particle size, polydispersity index (PDI), zeta potential, the entrapment efficiency, drug loading, and the critical micelle concentration (CMC) of CoQ10-loaded micelles were 19.76 nm, 0.112, -3.405 mV, 99.39%, 13.77%, and 5.623 × 10(-4) g/mL, respectively. Differential scanning calorimetry (DSC) analysis collectively corroborated that CoQ10 was entrapped into the micelles in amorphous form. The release pattern of drug was analyzed and proved to follow the first order. Additionally, the samples were exposed to the temperatures of 30°C for 6 months with more significant impact on their stabilities as compared to 4 and 25°C based on particle size and PDI. Under constant humidity with light protection long-term (25 ± 2°C, relative humidity (RH) 60 ± 10%, 18 months) conditions, there was no variation except minor changes of CoQ10 content of the samples. The shelf life of the micellar samples could be predicted as 24 months based on the stability results. Consequently, the CoQ10-loaded micelles showed excellent stabilities below 25°C as a potential drug candidate for further clinical applications.


Subject(s)
Drug Delivery Systems/methods , Micelles , Polyethylene Glycols/administration & dosage , Stearates/administration & dosage , Ubiquinone/analogs & derivatives , Animals , Chemistry, Pharmaceutical , Drug Stability , Erythrocytes/drug effects , Erythrocytes/physiology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Rabbits , Stearates/chemistry , Stearates/pharmacokinetics , Ubiquinone/administration & dosage , Ubiquinone/chemistry , Ubiquinone/pharmacokinetics
14.
Bull Exp Biol Med ; 161(5): 654-656, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27709380

ABSTRACT

The dynamics of coenzyme Q10 concentration in the blood plasma, liver, and brain of passive and active rats was studied on the model of metabolic stress. This parameter was shown to differ in rats with various patterns of behavior. Dietary consumption of coenzyme Q10 in doses of 10 and 100 mg/kg body weight was followed by changes in its content in experimental animals.


Subject(s)
Stress, Physiological , Ubiquinone/analogs & derivatives , Administration, Oral , Animals , Brain/metabolism , Dietary Supplements , Food Deprivation , Liver/metabolism , Male , Motor Activity , Organ Specificity , Rats, Wistar , Ubiquinone/administration & dosage , Ubiquinone/pharmacokinetics
15.
J Bioenerg Biomembr ; 47(1-2): 111-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25262284

ABSTRACT

Ubiquinone, commonly called coenzyme Q10 (CoQ), is a lipophilic electron carrier and endogenous antioxidant found in all cellular membranes. In the mitochondrial inner membrane it transfers electrons to complex III of the electron transport chain. The short chain CoQ analogue idebenone is in clinical trials for a number of diseases that exhibit a mitochondrial etiology. Nevertheless, evidence that idebenone ameliorates neurological symptoms in human disease is inconsistent. Although championed as an antioxidant, idebenone can also act as a pro-oxidant by forming an unstable semiquinone at complex I. The antioxidant function of idebenone is critically dependent on two-electron reduction to idebenol without the creation of unstable intermediates. Recently, cytoplasmic NAD(P)H: quinone oxidoreductase 1 (NQO1) was identified as a major enzyme catalyzing idebenone reduction. While reduction allows idebenone to act as an antioxidant, evidence also suggests that NQO1 enables idebenone to shuttle reducing equivalents from cytoplasmic NAD(P)H to mitochondrial complex III, bypassing any upstream damage to the electron transport chain. In this mini-review we discuss how idebenone can influence mitochondrial function within the context of cytoprotection. Importantly, in the brain NQO1 is expressed primarily by glia rather than neurons. As NQO1 is an inducible enzyme regulated by oxidative stress and the nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway, optimizing NQO1 expression in appropriate cell types within a specific disease context may be key to delivering on idebenone's therapeutic potential.


Subject(s)
Antioxidants , Electron Transport Complex III/metabolism , Electron Transport Complex I/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neuroprotective Agents , Oxidants , Ubiquinone/analogs & derivatives , Animals , Antioxidants/pharmacokinetics , Antioxidants/pharmacology , Humans , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Oxidants/pharmacokinetics , Oxidants/pharmacology , Oxidation-Reduction/drug effects , Ubiquinone/pharmacokinetics , Ubiquinone/pharmacology
16.
BMC Nephrol ; 16: 183, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26531095

ABSTRACT

BACKGROUND: Coenzyme Q10 (CoQ10) supplementation improves mitochondrial coupling of respiration to oxidative phosphorylation, decreases superoxide production in endothelial cells, and may improve functional cardiac capacity in patients with congestive heart failure. There are no studies evaluating the safety, tolerability and efficacy of varying doses of CoQ10 in chronic hemodialysis patients, a population subject to increased oxidative stress. METHODS: We performed a dose escalation study to test the hypothesis that CoQ10 therapy is safe, well-tolerated, and improves biomarkers of oxidative stress in patients receiving hemodialysis therapy. Plasma concentrations of F2-isoprostanes and isofurans were measured to assess systemic oxidative stress and plasma CoQ10 concentrations were measured to determine dose, concentration and response relationships. RESULTS: Fifteen of the 20 subjects completed the entire dose escalation sequence. Mean CoQ10 levels increased in a linear fashion from 704 ± 286 ng/mL at baseline to 4033 ± 1637 ng/mL, and plasma isofuran concentrations decreased from 141 ± 67.5 pg/mL at baseline to 72.2 ± 37.5 pg/mL at the completion of the study (P = 0.003 vs. baseline and P < 0.001 for the effect of dose escalation on isofurans). Plasma F2-isoprostane concentrations did not change during the study. CONCLUSIONS: CoQ10 supplementation at doses as high as 1800 mg per day was safe in all subjects and well-tolerated in most. Short-term daily CoQ10 supplementation decreased plasma isofuran concentrations in a dose dependent manner. CoQ10 supplementation may improve mitochondrial function and decrease oxidative stress in patients receiving hemodialysis. TRIAL REGISTRATION: This clinical trial was registered on clinicaltrials.gov [NCT00908297] on May 21, 2009.


Subject(s)
Dietary Supplements , Kidney Failure, Chronic/therapy , Oxidative Stress/drug effects , Renal Dialysis/methods , Ubiquinone/analogs & derivatives , Administration, Oral , Adolescent , Adult , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Drug Tolerance , Female , Humans , Male , Maximum Allowable Concentration , Middle Aged , Ubiquinone/administration & dosage , Ubiquinone/adverse effects , Ubiquinone/pharmacokinetics , United States , Young Adult
17.
Neuro Endocrinol Lett ; 36(4): 337-47, 2015.
Article in English | MEDLINE | ID: mdl-26454489

ABSTRACT

BACKGROUND: Many Vitamins and minerals for dietary supplements lack a standard scientific and regulatory definition that accurately reflects the bioavailabilities in humans. Especially the bioavailability of natural compounds in complex mixtures, where the different ingredients may interfere with each other, is unknown. METHODS: To learn more about the bioavailability of the ingredients in the complex compound LaVita® we examined blood levels of subjects, who ingested the multivitamin and trace element composition for 6 month continuously. Blood samples for the analysis of the ingredients were taken before, during, and after administration. RESULTS: Our data indicated a significant increase of most ingredients after 3 month, and additional three months, except for Vitamin (B9 Folic acid). The semivitamins Q10 and carnitine increased in the first 3 month (both p<0.001). While carnitine dropped during the second term, Q10 levels increased further slowly. After three months a significant increase was observed for iron (serum p=0.039; blood cells p=0.025), Selenium (serum p=0.048; cells p=0.006), and chromium (serum p=0.029). Zinc - known to interfere with the iron resorption - increased slowly in the first term of 3 months, but was raised significantly after 6 months (serum and blood cells, each p<0.001). The Copper/Zink ratio dropped accordingly (p<0.001). CONCLUSION: We conclude that resorption interference between specific ingredients, and after resorption redistribution of specific ingredients to various tissue compartments precludes a linear increase of the respective serum parameters. We observed no deleterious resorption competitions for individual compounds. No parameter reached critical levels. We conclude that the test substance (LaVita®) is a sufficiently safe composite for long term consumption.


Subject(s)
Dietary Supplements/standards , Trace Elements/pharmacokinetics , Vitamins/pharmacokinetics , Adult , Biological Availability , Carnitine/pharmacokinetics , Chromium/pharmacokinetics , Female , Folic Acid/pharmacokinetics , Healthy Volunteers , Humans , Iron/pharmacokinetics , Male , Middle Aged , Trace Elements/blood , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacokinetics , Vitamins/blood , Zinc/pharmacokinetics
18.
J Microencapsul ; 32(2): 157-65, 2015.
Article in English | MEDLINE | ID: mdl-25329530

ABSTRACT

This article describes the physicochemical properties of chitosan-coated liposomes containing skin-protecting agents, coenzyme Q10 and alpha-lipoic acid (CCAL). CCAL had a spherical shell-core structure and liposomes inverted the surface charge from negative to positive after coating with chitosan. Compared with the uncoated liposome, CCAL had higher zeta potential, larger droplet size and long-term stability. Fourier transform infrared spectroscopy (FTIR) study showed that the driving force for chitosan coating the liposomes was enhanced via hydrogen bonding and ionic bond force between the chitosan and the alpha-lipoic acid. While the encapsulation efficiency (EE) of alpha-lipoic acid also increased by interacting with the chitosan shell. In vitro antioxidant activity study showed an excellent hydroxyl radical scavenging activity of CCAL. In vitro release study displayed a sustained drug release, and in vitro penetration studies promoted the accumulation of drugs in rabbit skin.


Subject(s)
Chitosan , Coated Materials, Biocompatible , Free Radical Scavengers , Skin Absorption , Thioctic Acid , Ubiquinone/analogs & derivatives , Administration, Cutaneous , Animals , Chitosan/chemistry , Chitosan/pharmacokinetics , Chitosan/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacokinetics , Coated Materials, Biocompatible/pharmacology , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacokinetics , Free Radical Scavengers/pharmacology , Liposomes , Rabbits , Thioctic Acid/chemistry , Thioctic Acid/pharmacokinetics , Thioctic Acid/pharmacology , Ubiquinone/chemistry , Ubiquinone/pharmacokinetics , Ubiquinone/pharmacology
19.
J Nutr ; 144(7): 1030-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24759932

ABSTRACT

HDL and apolipoprotein A1 (apoA1) concentrations inversely correlate with risk of death from ischemic heart disease; however, the role of apoA1 in the myocardial response to ischemia has not been well defined. To test whether apoA1, the primary HDL apolipoprotein, has an acute anti-inflammatory role in ischemic heart disease, we induced myocardial infarction via direct left anterior descending coronary artery ligation in apoA1 null (apoA1(-/-)) and apoA1 heterozygous (apoA1(+/-)) mice. We observed that apoA1(+/-) and apoA1(-/-) mice had a 52% and 125% increase in infarct size as a percentage of area at risk, respectively, compared with wild-type (WT) C57BL/6 mice. Mitochondrial oxidation contributes to tissue damage in ischemia-reperfusion injury. A substantial defect was present at baseline in the electron transport chain of cardiac myocytes from apoA1(-/-) mice localized to the coenzyme Q (CoQ) pool with impaired electron transfer (67% decrease) from complex II to complex III. Administration of coenzyme Q10 (CoQ10) to apoA1 null mice normalized the cardiac mitochondrial CoQ pool and reduced infarct size to that observed in WT mice. CoQ10 administration did not significantly alter infarct size in WT mice. These data identify CoQ pool content leading to impaired mitochondrial function as major contributors to infarct size in the setting of low HDL/apoA1. These data suggest a previously unappreciated mechanism for myocardial stunning, cardiac dysfunction, and muscle pain associated with low HDL and low apoA1 concentrations that can be corrected by CoQ10 supplementation and suggest populations of patients that may benefit particularly from CoQ10 supplementation.


Subject(s)
Antioxidants/metabolism , Apolipoprotein A-I/metabolism , Disease Models, Animal , Mitochondria, Heart/metabolism , Myocardial Infarction/therapy , Myocardium/metabolism , Ubiquinone/analogs & derivatives , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacokinetics , Antioxidants/therapeutic use , Apolipoprotein A-I/blood , Apolipoprotein A-I/genetics , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/metabolism , Cardiotonic Agents/pharmacokinetics , Cardiotonic Agents/therapeutic use , Dietary Supplements , Electron Transport/drug effects , Electron Transport Complex II/chemistry , Electron Transport Complex II/metabolism , Electron Transport Complex III/chemistry , Electron Transport Complex III/metabolism , Heart/drug effects , Hypoalphalipoproteinemias/physiopathology , Injections, Intraperitoneal , Intestinal Absorption , Male , Mice , Mice, Knockout , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/enzymology , Myocardium/pathology , Tissue Distribution , Ubiquinone/administration & dosage , Ubiquinone/metabolism , Ubiquinone/pharmacokinetics , Ubiquinone/therapeutic use
20.
Mol Pharm ; 11(5): 1435-49, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24742138

ABSTRACT

The present investigation reports implications of the lipase digestibility of lyotropic liquid crystalline nanoparticles (LCNPs) on the oral bioavailability, in vivo antioxidant potential, and in vitro-in vivo relationship (IVIVR) of CoQ10 loaded LCNPs prepared from glyceryl monooleate (GLCQ) and phytantriol (PLCQ). Exhaustive optimization of the process variables was carried out, and optimized lyophilized formulations were found to have particle sizes of 140.45 ± 5.47 nm and 238.42 ± 8.35 nm and a polydispersity index (PDI) of 0.15 ± 0.01 and 0.22 ± 0.03 for GLCQ and PLCQ, respectively. The entrapment efficiency at 10% theoretical loading was found to be >90% in both the cases. The morphological characteristics of the developed formulations were assessed using high resolution transmission electron microscopy and small-angle X-ray scattering analysis, which showed hexagonal (HII) structure. The developed formulations were also found to be stable in simulated gastrointestinal fluids for the stipulated period of time. The in vitro drug release studies revealed a bimodal sustained release drug profile with Higuchi type release kinetics as the best fit release model for both the formulations. The best fit release models were found to be of the Hixson Crowell type in the case of GLCQ when carried out in lipase rich media, suggestive of matrix erosion and subsequent formation of secondary structures, which was further corroborated by carrier degradation studies. Furthermore, 9.1- and 10.67-fold increase in Caco-2 cell uptake was observed in the case of GLCQ and PLCQ, respectively, attributed to the formation of the virtual channel pathway as a probable absorption mechanism. Consequently, 7.09- and 8.67-fold increase in oral bioavailability was observed in the case of GLCQ and PLCQ, respectively. The IVIVR was also established with r(2) values in the order of 0.996 and 0.999 for GLCQ and PLCQ, respectively, in contrast to that of 0.484 for free CoQ10. Finally, in vivo prophylactic antioxidant efficacy against the STZ-treated rats using various markers such as GSH, LDH, SOD, MDA, glucose level, and body weight showed significantly higher antioxidant activity of CoQ10-LCNPs as compared to that of free CoQ10. In a nutshell, the developed formulation strategy poses great potential in improving the oral bioavailability of difficult-to-deliver drugs such as CoQ10.


Subject(s)
Nanoparticles/chemistry , Ubiquinone/analogs & derivatives , Animals , Antioxidants , Caco-2 Cells , Chemistry, Pharmaceutical/methods , Female , Humans , Microscopy, Electron, Transmission , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Scattering, Small Angle , Ubiquinone/chemistry , Ubiquinone/pharmacokinetics , Ubiquinone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL