Next Article in Journal
Association between Dysphagia and Frailty in Older Adults: A Systematic Review and Meta-Analysis
Next Article in Special Issue
Muscle Recovery and Nutrition
Previous Article in Journal
Impact of a Digital Lifestyle Intervention on Diabetes Self-Management: A Pilot Study
Previous Article in Special Issue
Krill-Oil-Dependent Increases in HS-Omega-3 Index, Plasma Choline and Antioxidant Capacity in Well-Conditioned Power Training Athletes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Coenzyme Q10 Supplementation and Its Impact on Exercise and Sport Performance in Humans: A Recovery or a Performance-Enhancing Molecule?

by
Franchek Drobnic
1,*,
Mª Antonia Lizarraga
2,
Alberto Caballero-García
3 and
Alfredo Cordova
4
1
Medical Services Shanghai Shenhua FC, Shanghai 201315, China
2
Medical Services FC Barcelona, 08014 Barcelona, Spain
3
Department of Anatomy and Radiology, Faculty of Health Sciences, GIR: “Physical Exercise and Aging”, Campus Universitario “Los Pajaritos”, University of Valladolid, 42004 Soria, Spain
4
Department of Biochemistry, Molecular Biology and Physiology, Faculty of Health Sciences, GIR: “Physical Exercise and Aging”, Campus Universitario “Los Pajaritos”, University of Valladolid, 42004 Soria, Spain
*
Author to whom correspondence should be addressed.
Nutrients 2022, 14(9), 1811; https://doi.org/10.3390/nu14091811
Submission received: 6 April 2022 / Revised: 22 April 2022 / Accepted: 22 April 2022 / Published: 26 April 2022
(This article belongs to the Special Issue Muscle Recovery and Nutrition)

Abstract

:
Evidence exists to suggest that ROS induce muscular injury with a subsequent decrease in physical performance. Supplementation with certain antioxidants is important for physically active individuals to hasten recovery from fatigue and to prevent exercise damage. The use of nutritional supplements associated with exercise, with the aim of improving health, optimizing training or improving sports performance, is a scientific concern that not only drives many research projects but also generates great expectations in the field of their application in pathology. Since its discovery in the 1970s, coenzyme Q10 (CoQ10) has been one of the most controversial molecules. The interest in determining its true value as a bioenergetic supplement in muscle contraction, antioxidant or in the inflammatory process as a muscle protector in relation to exercise has been studied at different population levels of age, level of physical fitness or sporting aptitude, using different methodologies of effort and with the contribution of data corresponding to very diverse variables. Overall, in the papers reviewed, although the data are inconclusive, they suggest that CoQ10 supplementation may be an interesting molecule in health or disease in individuals without a pathological deficiency and when used for optimising exercise performance. Considering the results observed in the literature, and as a conclusion of this systematic review, we could say that it is an interesting molecule in sports performance. However, clear approaches should be considered when conducting future research.

Graphical Abstract

1. Introduction

The use of performance-enhancing substances is current but not new to mankind. Substances to increase performance, reduce fatigue, facilitate recovery and even modify the will have been present and used since ancient times in the form of stimulants, defatigating or anabolic agents [1]. Therefore, a performance improvement is the combination of increased efficiency and effectiveness and is conceived as the achievement of the desired goal. In the field of sport, an improvement in performance can be considered as achieving the objectives of the chosen sport because to the skills acquired or the elements provided to improve it [2].
Associated with this training there is a wide range of support where nutritional supplementation has its place, either directly or indirectly. We can consider a nutritional supplement to be that extra contribution to the usual diet in order to complement it, to meet the requirements produced by an excess of us or because the characteristics of the substance provided offer qualities that improve or facilitate the sporting activity or work. The objective of its use, whether that substance improves health or the physical or sporting action itself, is to achieve optimum performance within the sphere of health. In a more practical and concrete way, the definition of supplement could be defined as “a substance that is purposefully ingested in addition to the habitually-consumed diet with the aim of achieving a specific health and/or performance benefit” [3].
Some supplements certainly have strong evidence bases to reflect a direct impact on athletic performance through the augmentation of various rate-limiting processes. However, other supplements may have an indirect impact on performance via their ability to support the training process, through their influence on factors such as inflammatory modulation, oxidative stress and signaling pathways for adaptation or their ability to support repetitive performance by restoring homeostasis between two exercise bouts. [4].
The reactive oxygen species (ROS) are a family of molecules that are continuously generated and consumed in all living organisms as a consequence of aerobic life [5]. The biological impact of ROS depends not only on their quantities but also on their chemical nature, (sub)cellular and tissue location and the rates of their formation and degradation. ROS represent key modulators of cellular physiology triggering adaptive responses when produced in limited amounts but are detrimental when they are produced in excess, leading to oxidative stress and cellular dysfunction [6]. Enhanced metabolic rate together with a rise in temperature and a decrease in cellular pH could accelerate ROS production [7], particularly in mitochondria, where constitutively 1–2% of oxygen is converted into superoxide anion [8]. As a result, the exercising muscle produces much larger amounts of ROS compared to the muscle at rest [9].
Skeletal muscle has a high capacity to adapt to various demands. Interestingly, the functions of skeletal muscle and their neighboring cells are altered by oxidative stress modulation [10]. Macrophages are a major source of reactive oxygen species (ROS) and reactive nitrogen species (RNS) in inflammation [11]. Inflammatory macrophages release glutathionylated peroxiredoxin-2, which acts as a “danger signal” to trigger the production of tumour necrosis factor alpha [12]. ROS exert major biological effects as purely deleterious molecules causing damage to DNA, proteins and lipids, notably during exercise [13]. Evidence exists to suggest that ROS induce muscular injury with a subsequent decrease in physical performance [12,13]. Supplementation with certain antioxidants is important for physically active individuals to hasten recovery from fatigue and prevent exercise damage [14].
There are fundamental molecules in the organism that we have been studying for years as nutritional supplementation and which, for various reasons, have not yet defined their true role in the possible improvement of sports performance such as CoQ10. The CoQ10 molecule is a fat-soluble, vitamin-like, ubiquitous compound, with a key role in cellular bioenergetics, because act as a cofactor in the mitochondrial respiratory chain to supply energy for cells [15,16].
CoQ10 participates in redox reactions within the electron transport chain at the mitochondrial level facilitating the production of adenosine triphosphate (ATP) [15] to a mobile redox agent shuttling electrons and protons in the electron transport chain. The CoQ10 have more functions that its role in the mitochondria. These including lipophilic antioxidant effect that protects the DNA [17], the phospholipids and the mitochondrial membrane proteins against the lipid peroxidation [18,19]. CoQ10 also help to regenerates vitamins C and E and reduces inflammatory markers [15,20].
CoQ10 acts as an antioxidant in both the mitochondria and lipid membranes by scavenging reactive oxygen species (ROS), either directly or in conjunction with a-tocopherol [15,21,22]. This antioxidant activity appears only with the reduced form (ubiquinol). The oxidized form (ubiquinone) is readily reduced to ubiquinol enzymically after dietary uptake [23].
The research on the impact of Ubiquinone, and its reduced form Ubiquinol, as sup-plementation in exercise in humans began in the late 1980s. More recently labdeveloped Mitoquinone [24,25,26,27]. Unfortunately, there is no consistency in the studies due to the diversity of functions of CoQ10 in the body that can be applied to the improvement of physical performance. There are a wide dispersion of study models allowing the coexistence of different dose, patterns of administration, vehicle or diet to enhance bioavailability, subjects considered present diverse physical capacities or sports experience, are of various ages, and undertake different types of stressing exercise if it is done, etc.
Given the methodological diversity, it is difficult to consider uniformity of studies, which could increase the scientific evidence. Greater uniformity can be achieved if results are evaluated without considering methodologies. For this reason, it is not possible to elaborate an ideal review to elucidate the effect of Q10 on sports performance. Nor would it be feasible in studies on its use in clinical therapeutics, ageing and disease [27,28,29,30,31]. However, there are previous reviews that provide interesting data on the subject, some of them recent [32], and some of them extraordinary to understand the basis for the usefulness of CoQ10 as an ergogenic substance and its limitations. Previously, Malm et al. [33] and Sarmiento et al. [34] conducted studies of great interest for the same purpose.
The current work that we present reviews, in a holistic and at the same time pragmatic way, the usefulness of CoQ10 in sports performance. The main objective of this review was to critically evaluate the effectiveness of CoQ10 supplementation on physical performance. Also, analyse its effects as inflammatory and oxidative factors, in a physically active population and specially in athletes.

2. Methods

2.1. Search Strategy

The literature search was conducted according to the guidelines for meta-analysis of systematic reviews (PRISMA). [35,36,37]. The studies were evaluated according to variables related to the type and size of the population studied, the administration regimen of the product, its bioavailability, the existence or not of a placebo and/or control group, the tests used to assess performance improvement and the appropriateness of the method used. Of course, all in relation to our objective, when Ubiquinone [38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79], Ubiquinol [80,81,82,83,84,85,86,87,88,89,90] or Mitoquinone [80,91,92,93] was administered.
A structured search was carried out in the databases SCOPUS, Medline (PubMed), and Web of Science (WOS), which includes other databases such as BCI, BIOSIS, CCC, DIIDW, INSPEC, KJD, MEDLINE, RSCI, SCIELO, all of which are high-quality databases which guarantee good bibliographic support. The search was performed from the first available date, according to the characteristics of each database until February 2022 (time of last update).

2.2. Selection of Articles: Inclusion and Exclusion Criteria

For the articles obtained in the search, the following inclusion criteria were applied to select studies: articles depicting a well-designed experiment that included the ingestion of a dose of CoQ10, or a CoQ10 product, before and/or during exercise in humans. It was considered appropriate to use the descriptors “Ubiquinone”, “Coenzyme Q10”, “Ubiquinol”, “Q10”, “Mitoquinone” combined with “Exercise”, “Athlete”, “Performance”, Sport” and “Physical Exertion”. The limit “Humans” was applied.
The suitability of the articles was determined using the GRADE [94] and the level of evidence criterion [95]. All analysed articles demonstrated a moderate or high scientific quality, and those with a degree of evidence that can be classified from 2 to 2++ were selected. With the same concept as the Sarmiento et al. review [34], no restrictions on the participants’ gender, age or fitness level were established initially. Inclusion criteria were then applied, which were: randomised, double-blind, controlled, parallel design studies assessing plasma CoQ10. Likewise, the studies would be about the assessment of the inflammatory and oxidative effects, and aspects related to muscle injury or its impact on physical performance.
On the other hand, and because of the objective of the substance administration, in studies where the evaluation of its impact on physical and sporting performance had to be assessed, it should be carried out with athletes or subjects who were trained and comfortable with performing intense exercise. Finally, the measurement of the CoQ10 in muscle-by-muscle biopsy was also considered an important and necessary aspect, as it is the target site of the functionality of the molecule under study. The “Full search strategy” is presented below.
Study quality was assessed according to the PEDro scale (https://pedro.org.au/english/resources/PEDro-scale/) (accessed on 4 September 2021). The PEDro scale has strong reliability and validity [96,97]. The scale consists of a list of 11 items; for each individual criterion of scientific methodology, studies receive a score of 1 when the criterion is clearly met or 0 when that criterion is not adequately met. The first item (eligibility criteria) does not receive a score because it is related to external validity and, therefore, does not reflect the quality dimensions assessed by the PEDro scale. The PEDro score for each study is shown in Table 1.
Thus, the total scores range from 0 to 10. The criteria included in the Scale are:
  • Eligibility criteria were specified (no score).
  • Subjects were randomly allocated to groups.
  • Allocation was concealed.
  • The groups were similar at baseline regarding the most important prognostic indicators.
  • There was blinding of all subjects.
  • There was blinding of all therapists who administered the therapy.
  • There was blinding of all assessors who measured at least one key outcome.
  • Measures of at least one key outcome were obtained from more than 85% of the subjects initially allocated to groups.
  • All subjects for whom outcome measures were available received the treatment or control condition as allocated or, where this was not the case, data for at least one key outcome was analysed by “intention to treat”.
  • The results of between-group statistical comparisons are reported for at least one key outcome.
  • The study provides both point measures and measures of variability for at least one key outcome.

3. Results

Of the articles viewed, 59 were related to the select descriptors, and only 9 met all the inclusion/exclusion criteria (Figure 1). Four, out of the 59, were duplicated in any sense, Drobnic [47] and Paredes-Fuentes [98], Tauler [69] and Ferrer [70], the two studies by Ciocoi-Pop [44], and Diaz-Castro [83,84]. The data for the studies that meet all the proposed inclusion criteria are listed together with those that only lack a muscle biopsy in Table 2 and Table 3. The remaining studies are listed in Table 4, Table 5, Table 6, Table 7, Table 8 and Table 9. Only Svensson’s 1999 study [68] fulfilled all criteria. These authors suplemented the subjects with 120 mg/day of Ubiquinone. In the results they observed an increase in plasma CoQ10 levels after administration. However, it did not increase in muscle. Also, was not observed improvement in the oxidative pattern after physical assessment tests. Of the other eight studies selected, four are with Ubiquinone and the other four with Ubiquinol. All observed elevation of baseline plasma levels after treatment. Of the nine studies, only six assessed the impact on physical performance, none on sports performance. Two, Suzuki [90] and Kon [86], found a positive impact, while Svensson [68] Kizaki [85], Braun [41] and Weston [72] did not. Interestingly, there is a positive response on the inflammatory [83,89] and antioxidant pattern to the stress [41,56,86,89], bone remodelling [85] and the metabolic [57] response.
The inclusion criteria administered in the review are indeed strict. It is true that some of them could be omitted, e.g., that the study can be cross-over, that studies with a control group and not only placebo are included, or that it is not determined whether or not the tested molecule increases in tissues. If the discussion focuses not only on the objective but considers a correct methodological structure for assessing the effect on performance, then there would be various deselected studies that certainly deserve to be considered. For this reason, Table 4, Table 5, Table 6, Table 7, Table 8 and Table 9 include the rest of the studies screened and some of them are discussed.
In the tables the dosage administered is indicated in mg per day (mg/d) and in those studies where it was offered together with vitamin C, E or another substance, it is indicated with a symbol (+) next to the molecule administered. The number of subjects included in the study is shown in the “Total/CoQ10” column, where those who took the molecule in the study are indicated in reference to the total number of subjects in the study. The crossover studies show the same number on both sides of the slash bar. The quality of physical fitness is shown in the column “Type of subjects”. To homogenize the groups, although there is no consistent definition in the study methodologies in this respect, it has been considered in a somewhat arbitrary way but determined by the existing indications where available, the quality of the tests performed and the data of the subjects, in five different groups “High level”, “Well trained subjects”, “Moderately trained subjects”, “Physically active subjects”, “No active subjects”, “Patients”. The difference between the groups “Moderately trained subjects” and “Physically active subjects” depends on the quality of the indications in the studies. The first ones used to practice sport or have a regular weekly training schedule. However, the second are only, as the concept indicates, “physically active subjects” and they cannot be considered sedentary or specifically, “no active”. The physical tests are indicated in the tables when it was performed. From the 55 studies, in 22 were performed a graded maximal exercise test, and in 8, specific anaerobic tests. Other exercise stress methods, either sport-specific, with field or swimming pool tests, or custom-made laboratory tests, categorized as maximal, sub-maximal, evaluating energy metabolism, degree of fatigue, or the provocation of muscle injury, were also performed within 30 of these other studies. Also, there were 4 studies with no specific stress test. It is interesting that for the Ubiquinol and Mitoquinone none of the exercise tests were similar.
Table 10 shows the data of the studies related to the sports level of the subject with the impact on physical and sports performance variables, inflammatory and oxidative patterns, muscle injury or other aspects of health, in a simple form, indicating whether it has a positive effect, no effect or a negative effect. As it is shown, there are more studies aimed at sporting people of a certain level, which in turn are those with the most positive results (39 vs. 12, 74.5%), with the data in relation to the oxidative and inflammatory response being the most attractive. No such benefit was observed in moderately active subjects, nor in non-active subjects (11 vs. 4; 73.3%). In a general observation of all subjects, there is a beneficial trend for all the evaluated objectives, sports performance, physical performance, inflammatory response pattern, oxidative and other health-related domains. On the other side, when age categories, Table 11, are evaluated, there is a trend of beneficial effect for all groups, with the most prominent positive effect on the oxidative pattern and the different health evaluations. The most evaluated group is <30 years old (63.8%), and the lowest is >50 years old (12.1%).

4. Discussion

Recent publications on CoQ10 supplementation suggest that it may be an interesting molecule in health [27,28] and for optimising exercise performance [32]. However, in the field of physical activity, there is a great diversity of work with different orientations, which sometimes makes it difficult to situate this work in the context of sports performance. Our discussion could provide a practical criterion to be taken into account by professionals wishing to assess this molecule (CoQ10) in the context of sport.

4.1. Dosage and Biodisponibility

Coenzyme Q10 is a water-insoluble substance with limited lipid solubility and a relatively high molecular weight, so it is also poorly absorbed in the gastrointestinal tract [16], but its reduced form (Ubiquinol) is 6–10 times more bioavailable than Ubiquinone or oxidised CoQ10 [100]. Its absorption and uptake pathways begin with emulsification and micelle formation with the fatty components of food, which is facilitated by pancreatic and biliary secretions [101]. Likewise, the efficiency of absorption also depends on the dose administered [102], the bioavailability [103,104] the diet [105] and the stability of the final product [106]. That is the reason why strict vegetarian diets may be deficient to maintain the CoQ10 level [107]. Not only because of the lower contribution of CoQ10, but also because the diet modifies its bioavailability. In the studies screened in this review, the dose of Ubiquinone administered is quite heterogeneous, ranging from 30 to 300 mg/day, with lower doses being more frequent in the older studies. With regard to Ubiquinol, the studies indicate doses ranging from 200 to 300 mg/day, with a single exception of 600 mg/day. In general, the studies analysed show a fivefold increase in plasma levels when Ubiquinol is administered at a dose of 250 mg/d and a twofold increase in plasma levels when Ubiquinone is administered at a dose of 150 mg/d.
The CoQ10 content in the body varies in different organelles, tissues and species [108]: in plasma, 0.46–1.78 μmol/L or 101–265 μmol CoQ10/mol Chol [109], and in muscle 140–580 nmol/g protein [110]. In football players, CoQ10 levels have been linked to sports performance [99]. In this regard, Bonetti et al. [40] indicate that structured exercise and training maintain and improve plasma levels over the long term. However, it should be noted that excessive physical work, illness and oxidative stress can lead to a reduction in plasma CoQ10 levels [51,73,98,111].
Throughout the analysis of the literature data used in this review, we found that the mean plasma CoQ10 level in high-training subjects was 0.97 μg/mL, compared to 0.83 μg/mL in moderately trained subjects or 0.93 μg/mL in sedentary subjects. Only two articles found higher levels, Tauler et al. [69]: 3 μg/mL, and Kizaki et al. [85]: 1.7 μg/mL. Laaksonen et al. [58] reported values of 0.9 μg/mL in young athletes and 1.3 μg/mL in older athletes. However, at the muscle level, the young athletes had a higher concentration of CoQ10, 118 μg/mL compared to 92 μg/mL in the older subjects. This discrepancy in the population of a certain age has already been assessed in plasma, irrespective of differences in formulations and excipients, doses administered and treatment time [112].
However, it also warns us that an increase in plasma does not necessarily correlate with an increase in muscle tissue. It is possible that sufficient treatment time or higher dosage may be required, as it occurs in animal models where it is shown that chronic ingestion of relatively large doses of CoQ10 in the diet is able to increase the CoQ10 concentrations, especially in the mitochondrial fractions of the heart [113]. It is therefore worth remembering that the use of the same dose of Ubiquinone, or Ubiquinol, taken orally by athletes with different body compositions, leads to different intakes of CoQ10 per kg of body weight [32]. On the other side, the change in muscle and plasma levels in relation to the CoQ10 administration is inconsistent. In this way some authors [45,58,68,78] have shown an increase of Q10 in plasma that it was not reflected in the muscle. In contrast, Laaksonen et al. [58] in the young athletes and Drobnic et al. [47] do observe an increase in concentration, although not significant, before and after supplementation. This discrepancy may be due to other methodological aspect, and to the CoQ10 regimen administered. Muscle biopsy is usually performed by needle biopsy. Although fine-needle aspiration is a well-referenced model, the samples are relatively small (15–30 mg Cooke, 40–80 mg Svensson, 50–100 mg Zhou), which are, in our case intended to illustrate the entire composition of the muscle mass of a high level or veteran athlete. This type of sample is undoubtedly excellent for discriminating and diagnosing pathology but limits the repeatability of assessments when quantifying molecules, so a larger muscle sample is much appreciated by the laboratory pathologist. In the last study discussed [47,98], a muscle biopsy sample of 200–250 mg was obtained from the vastus lateralis muscle using the ENCOR ULTRA® 10 G biopsy system under ultrasound guidance. This is a simple and practical method, although not cheap, but it provides a high-quality homogeneous sample, very suitable for the study of muscle tissue, and allows portions to be retained for various subsequent analyses and pooling if a multicentre project is considered.

4.2. Antioxidant Activity

The use of CoQ10 for its antioxidant effect has been widely described in different pathologies [30,114,115,116,117] and in exercise [34,46,65,115,118]. The most important and relevant of its actions is the potent antioxidant capacity of its coexisting redox forms (ubiquinone, semiubiquinone and ubiquinol), which act on the mitochondrial and cell membrane [115,116,117,118]. These antioxidant properties of CoQ10 also help to recycle other antioxidants such as vitamin C and vitamin E, which act on free radicals or oxidants, reducing and neutralising compounds [116,117,118].
It is estimated that the increase in the production of free radicals and other ROS by exercise [119] accounts for 1% to 5% of the oxygen consumed during respiration [120]. ROS have been suggested to be the main reason for exercise-induced alterations in the redox balance of muscle, promoting oxidative injury and muscle fatigue [121], in addition to the damage to DNA and muscle structure [122], all of which impairs athletic performance. In addition, CoQ10 also regulates eNOS function in different cell membranes. Therefore, the depletion of CoQ10 may promote uncoupling of eNOS, making it an additional source of ROS, shifting the nitro-redox balance towards oxidation [123] that can be helpful at different ages [124].
As can be seen in Table 10, which shows the effects of Ubiquinone and Ubiquinol supplementation on different variables as a function of physical condition, antioxidant capacity in relation to exercise is rated as much more beneficial than the others. It is evident in both athletes and sedentary subjects irrespective of their age. This suggests that their antioxidant activity is real and may be aimed to prevent the excess of ROS favour the tissue and bioenergetic recovery of the cells.

4.3. Muscular Injury and Inflammatory Process

High intensity, strenuous and/or long-duration exercise, especially eccentric contraction exercise, can induce muscle damage (EIMD). This damage is derived from the mechanical stress caused and the inflammatory response [125,126,127,128,129]. This inflammatory response also leads to the release of ROS and cytokines [130,131] promoting the activation of gene transcription factors [132,133]. During the inflammatory process, there is increased leakage of muscle proteins into the circulation (creatine kinase-CK, myoglobin-Mb, aspartate transaminase-AST) [134,135,136,137,138]. The CK and Mb are indirect markers of muscle damage, moreover, ROS produced by neutrophils contribute to muscle damage and circulating neutrophils increase after exercise, leading to muscle damage [127,138].
The effect of CoQ10 supplementation on exercise-induced muscle damage CK level is controversial. It decreases in rats [139], but the results in the lab for humans are very diverse, offering similar [64,65,79], lower [42,48,49,86,90] or even higher [60] concentrations. In the case of the field tests in runners, nor Kaikkonen et al. [58] nor Armanfar et al. [39]) did observe any change on CK activity after a marathon or a 3000 m test, respectively.
Of all these biomarkers, CK is the most commonly used to assess muscle damage [140,141,142,143,144]. The CK enters the blood through the increase in membrane permeability that occurs after exercise. In addition, plasma CK amounts are conditioned by many exercise-related factors, such as intensity, duration, training status of the individual or exercise experience [141,142]. The amount of muscle mass and CK content within the muscle fibre, which is dependent on individual genetics and gender, must also be considered [143,144,145]. Moreover, muscle damage shows an enormous inter-individual variability that depends on additional factors that could exert an additional influence [143,146,147,148,149,150,151] so its use should not be used as a gold standard parameter for assessing the extent of muscle injury due to exercise, as its use is controversial for research [152] or monitoring training [153] but it can be useful as a complementary value associated with other more specific determinations.
The impact of CoQ10 in the prevention of muscle injury should lie mainly in its antioxidant and anti-inflammatory action, its protective action on mitochondria and DNA and its modifying effect on gene expression [154]. In addition, CoQ10 stabilises the phospholipid structure of cell membranes and protects skeletal muscle cells [155]. The CoQ10 supplementation may therefore reduce exercise-induced muscle injury by increasing the concentration of CoQ10 in muscle cell membranes and stabilising the cell membrane.
Other authors [156] have reported that CoQ10 treatment attenuates the oxidative activity of neutrophils. Thus, as can be seen in this review, the studies analysed show a beneficial tendency for the administration of CoQ10 to modify the inflammatory response, cytokines and transfer factors [39,46,47,84,89].

4.4. Sport and Exercise Performance

The bioenergetic approach to sport performance is one of many different ways to assess the success of exercise performance [157]. The determination of the maximal oxygen uptake (VO2max), anaerobic threshold, lactate concentration, presence of CK, oxidation-reduction metabolism molecules, perception of effort, heart rate evolution or fatigue index are some of the other variables frequently studied in research on the supplementation of ergogenic aids.
The evaluation of the VO2max is undoubtedly one of the fundamental variables when it comes to observing the effect of a exercise-enhancing substance. However, oxygen consumption can be limited by several factors with all the systems involved: respiratory, haematological, circulatory and mitochondrial metabolism [158]. On the other side, it is the energy cost that really “makes the difference” [159]. Coenzyme Q10, has some place in that environment, and therefore must be considered, as it is certainly in ATP regeneration and anaerobic threshold conflict that the useful administration of a compound is likely to have an influence. The VO2max will determine exercise time to exhaustion, so we may see great variability, which should be taken into account when considering a study on the effect of ergogenic aids. In this regard, McLellan et al. [160] have calculated a range of variability from 2.8 to 31.4% for five submaximal tests in cyclists during exercise to exhaustion. Also, Pereira et al. [161] have observed that the intraindividual variation of running economy cannot be underestimated, mostly when a substance used to enhance the success is under evaluation.
The ability to maintain a high percentage of maximal oxygen uptake offers the possibility of achieving high exercise intensity [162]. Although the gas exchange threshold (GET) is a good predictor of exercise performance, it does not reflect the specific intensity of the athlete’s competition [163]. Alternatively, other variables such as critical power (CP) (for cycling) or critical velocity (for running) have emerged as more viable surrogates and are considered to be more consistent in high-intensity exercise [164]. Indeed, CP represents the highest oxygen volume at which lactate and blood oxygen levels can stabilize [165]. Because of this, CP has been found to be a robust parameter representative of a fatigue threshold, located approximately midway between GET and VO2max., which demarcates strong from severe intensity domains [162].
Only in two studies, out of the nine selected in this review, is there some evidence of improved physical performance attributable to CoQ10 supplementation. With respect to the 55 initially screened, most of them assess variables related to maximal work or exercise levels reaching fatigue. However, it is not easy to justify a possible VO2max improvement effect when this depends on many factors as it has exposed [158] other than those provided by CoQ10 supplementation, especially in subjects whose mitochondria are not primed to be significantly more efficient without an adequate period of associated stimulus (exercise). Mitochondrial capacity appears to be much higher than that assessed by this parameter [165]. Individuals with higher mitochondrial density enjoy a higher margin of functional mitochondrial capacity, which perhaps explains why is greater the effect of COQ10 when administered to them. It should be remembered that mitochondrial capacity may be underexpressed in individuals who, although not physically active at the start of the studies, were very active in their youth and have a pool of latent mitochondria that have not disappeared [166]. That would be also the case for athletes who have taken anabolic steroids when young [167]. The increased mitochondrial responsiveness can be activated with an appropriate physical and nutritional stimulus. It is quite possible that the change in performance would be different in individuals who never played sport intensively in their youth than those who did. In any case, the results would be homogenised across studies if mitochondrial density were determined.

4.5. Other Action of Coenzyme Q10 Associated to Exercise

Of the selected studies, two explore the exercise-associated effect of CoQ10 on glucose metabolism and bone remodelling [56,84]. Interestingly, as CoQ10 is a ubiquitous molecule at the metabolic level, it is expected to have positive effects by optimising the response of exercise-related systems [42,71,79,87].
There are many other actions or possible implications of this molecule in relation to exercise and sport [32]: its impact on the nervous system and muscle diseases [168], stabilizing red blood cells (to increase the resistance to oxidative stress) [169], more fluidity properties [170], optimizing endothelial dysfunction [171,172], and even modifying muscle composition [173]. All of which are extremely interesting and which, once studied in-depth, could determine the true impact of CoQ10 on each of them.
Furthermore, CoQ10 supplementation may work synergistically with other molecules that help to prevent or restore tissue after the stress produced by the exercise. This is the case of creatine in the energy metabolism and tissue restoration [174,175,176], the omega-3 fatty acids in their antioxidant and cellular modulating activity [177,178,179], or the curcumin for its specific antiinflammatory and antioxidant activity [180,181,182].

5. Recommendations for Future Research

One of the major limitations is that there is a diversity of exercise modalities in different sports disciplines. Additionally, the sample size of the studies is not homogeneous. Moreover, some of the studies do not specify gender and there is also the problem of doses and times of use of CoQ10 supplementation.
Therefore, we believe that given the differences between the studies evaluated, we can offer a reference of methodological guidelines with a certain justification to facilitate a more precise analysis of those that have been developed with scientific guarantees in the future (Table 12).
The rationale for each of the recommendations is the subject of specific comments in the discussion of this review, such as the desire that it should not be a crossover study or that the exercise variables to be determined before and after supplementation should be well-defined. Others are basic, such as using placebos, or suggesting that the study be double-blind or randomised [183,184]. However, there are others that are set out in the table and which we extend below.
There are four types of populations that are markedly different in the possible action of CoQ10 in relation to physical performance: the sedentary, the moderately physically active and the very active [185,186], which in turn correspond to different age groups. Functionality, learning, memory and mitochondrial density are likely to behave differently after exposure to the supplement. The proposed table shows four age ranges, which are somewhat arbitrary and could be adjusted to human age periods [187], but since these correspond more to concepts of mental maturity and senility, it is considered more interesting to associate them with aspects of physical maturation and exposure to pathology [188] rather than purely psychological or mental aspects.
The administered dose of both the oxidised and reduced forms must be adequate to ensure that bioavailability is correct. A dose is proposed that, based on the latest studies with Ubiquinol, shows an adequate increase in plasma levels. Moreover, the period of administration must be given a minimum of time to justify that the functional results correspond to the presence of the element studied in the place where it functions, the mitochondria [101,108]. This thinking justifies the determination of levels not only in plasma, which we consider mandatory, but also recommends determination in muscle tissue to associate it with the mitochondrial characteristics of interest to us. In addition, it is recommended that at least one biopsy be taken from the vastus lateralis quadriceps if it is decided to do several. The different concentration in humans is not fully defined but it is presumable that it occurs as in rodents and its concentration is different depending on the muscle examined [188,189]. In turn, plasma determination should not be done when the treatment has been administered at a time when plasma availability is high due to absorption. It is therefore justified that at least 24 h should elapse, considering the bioavailability of the substance [16].
It is also necessary to consider aspects related to the idiosyncrasies of the subject in relation to the group studied, such as the diet maintained, excessive exercise, or the sedentary lifestyle of adults who practised high-intensity sport in their youth, as explained in the discussion.

6. Conclusions

From the evaluation of the various studies reviewed, and considering the methodological difficulties that exist between them, it can be concluded that the use of Coenzyme Q10 seems to offer a good profile in the control of an oxidative pattern with a certain anti-inflammatory activity at the cellular level in response to exercise in the various populations studied, in addition to some properties that should be studied in greater depth. It can therefore be seen as a protective and recuperative substance rather than an ergogenic substance in itself. Coenzyme Q10 is a promising molecule in terms of its qualities and safety profile, which is why it is considered necessary to carry out studies with a methodology that is more oriented towards the profile of the substance under study, with practical criteria that could favour the performance of more complex analyses between the various scientific groups involved.

Author Contributions

Conceptualization, F.D. and A.C.; methodology, F.D.; resources, data curation F.D. and A.C.; writing—original draft preparation, F.D., A.C., A.C.-G. and MAL.; writing—review and editing F.D., A.C., A.C.-G., M.A.L.; funding A.C.-G.; project administration A.C. and A.C.-G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Caja Rural Soria.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Caja Rural Soria.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kamieński, Ł. Shooting Up: A Short History of Drugs and War; Oxford University Press: New York, NY, USA, 2016; 381p. [Google Scholar]
  2. Newell, A.; Rosembloom, P. Mechanisms of skill acquisition and the law of practice. In Cognitive Skills and Their Acquisition; Anderson, J., Ed.; Psychology Press: Hillsdale, NJ, USA, 1981; pp. 1–55. Available online: https://www.researchgate.net/publication/243783833_Mechanisms_of_skill_acquisition_and_the_law_of_practice (accessed on 5 April 2022).
  3. Maughan, R.J.; Burke, L.M.; Dvorak, J.; Larson-Meyer, D.E.; Peeling, P.; Phillips, S.M.; Rawson, E.S.; Walsh, N.P.; Garthe, I.; Geyer, H.; et al. IOC Consensus Statement: Dietary Supplements and the High-Performance Athlete. Int. J. Sport Nutr. Exerc. Metab. 2018, 28, 104–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Peeling, P.; Binnie, M.J.; Goods, P.S.R.; Sim, M.; Burke, L.M. Evidence-Based Supplements for the Enhancement of Athletic Performance. Int. J. Sport Nutr. Exerc. Metab. 2018, 28, 178–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Dickinson, B.C.; Chang, C.J. Chemistry and biology of reactive oxygen species in signaling or stress responses. Nat. Chem. Biol. 2011, 7, 504–511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Sies, H. Oxidative Stress: Introductory Remarks; Oxidative Stress Academic Press: London, UK, 1985; pp. 1–8. [Google Scholar]
  7. Yavari, A.; Javadi, M.; Mirmiran, P.; Bahadoran, Z. Exercise-induced oxidative stress and dietary antioxidants. Asian J. Sports Med. 2015, 6, 24898. [Google Scholar] [CrossRef] [Green Version]
  8. Boveris, A.; Chance, B. The mitochondrial generation of hydrogen peroxide. General properties and effect of hyperbaric oxygen. Biochem. J. 1973, 134, 707–716. [Google Scholar] [CrossRef]
  9. Chance, B.; Williams, G.R. The Respiratory Chain and Oxidative Phosphorylation. In Advances in Enzymology—And Related Areas of Molecular Biology; Nord, F.F., Ed.; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2006; pp. 65–134. [Google Scholar]
  10. Brüne, B.; Dehne, N.; Grossmann, N.; Jung, M.; Namgaladze, D.; Schmid, T.; von Knethen, A.; Weigert, A. Redox control of inflammation in macrophages. Antioxid. Redox Signal. 2013, 19, 595–637. [Google Scholar] [CrossRef] [Green Version]
  11. Bogdan, C.; Röllinghoff, M.; Diefenbach, A. Reactive oxygen and reactive nitrogen intermediates in innate and specific immunity. Curr. Opin. Immunol. 2000, 12, 64–76. [Google Scholar] [CrossRef]
  12. Salzano, S.; Checconi, P.; Hanschmann, E.M.; Lillig, C.H.; Bowler, L.D.; Chan, P.; Vaudry, D.; Mengozzi, M.; Coppo, L.; Sacre, S.; et al. Linkage of inflammation and oxidative stress via release of glutathionylated peroxiredoxin-2, which acts as a danger signal. Proc. Natl. Acad. Sci. USA 2014, 111, 12157–12162. [Google Scholar] [CrossRef] [Green Version]
  13. Fernández-Lázaro, D.; Mielgo-Ayuso, J.; Seco Calvo, J.; Córdova Martínez, A.; Caballero García, A.; Fernandez-Lazaro, C.I. Modulation of Exercise-Induced Muscle Damage, Inflammation, and Oxidative Markers by Curcumin Supplementation in a Physically Active Population: A Systematic Review. Nutrients 2020, 12, 501. [Google Scholar] [CrossRef] [Green Version]
  14. Banerjee, A.K.; Mandal, A.; Chanda, D.; Chakraborti, S. Oxidant, antioxidant and physical exercise. Mol. Cell Biochem. 2003, 253, 307–312. [Google Scholar] [CrossRef]
  15. Ernster, L.; Dallner, G. Biochemical, physiological and medical aspects of ubiquinone function. Biochim. Biophys. Acta 1995, 1271, 195–204. [Google Scholar] [CrossRef] [Green Version]
  16. Bhagavan, H.N.; Chopra, R.K. Plasma coenzyme Q10 response to oral ingestion of coenzyme Q10 formulations. Mitochondrion 2007, 7, S78–S88. [Google Scholar] [CrossRef]
  17. Williamson, J.; Davison, G. Targeted Antioxidants in Exercise-Induced Mitochondrial Oxidative Stress: Emphasis on DNA Damage. Antioxidants 2020, 9, 1142. [Google Scholar] [CrossRef]
  18. Forsmark-Andrée, P.; Ernster, L. Evidence for a protective effect of endogenous ubiquinol against oxidative damage to mitochondrial protein and DNA during lipid peroxidation. Mol. Asp. Med. 1994, 15, s73–s81. [Google Scholar] [CrossRef]
  19. López-Lluch, G.; Rodríguez-Aguilera, J.C.; Santos-Ocaña, C.; Navas, P. Is coenzyme Q a key factor in aging? Mech. Ageing Dev. 2010, 131, 225–235. [Google Scholar] [CrossRef]
  20. Crane, F.L.; Sun, I.L.; Sun, E.E. The essential functions of coenzyme Q. Clin. Investig. 1993, 71 (Suppl. S8), S55–S59. [Google Scholar] [CrossRef] [PubMed]
  21. Kagan, V.; Serbinova, E.; Packer, L. Antioxidant effects of ubiquinones in microsomes and mitochondria are mediated by tocopherol recycling. Biochem. Biophys. Res. Commun. 1990, 169, 851–857. [Google Scholar] [CrossRef]
  22. Lass, A.; Sohal, R.S. Electron transport-linked ubiquinone-dependent recycling of alpha-tocopherol inhibits autooxidation of mitochondrial membranes. Arch. Biochem. Biophys. 1998, 352, 229–236. [Google Scholar] [CrossRef]
  23. Mohr, D.; Bowry, V.W.; Stocker, R. Dietary supplementation with coenzyme Q10 results in increased levels of ubiquinol-10 within circulating lipoproteins and increased resistance of human low-density lipoprotein to the initiation of lipid peroxidation. Biochim. Biophys. Acta 1992, 1126, 247–254. [Google Scholar] [CrossRef]
  24. Smith, R.A.J.; Porteous, C.M.; Gane, A.M.; Murphy, M.P. Delivery of bioactive molecules to mitochondria in vivo. Proc. Natl. Acad. Sci. USA 2003, 100, 5407–5412. [Google Scholar] [CrossRef] [Green Version]
  25. Murphy, M.P.; Smith, R.A.J. Targeting Antioxidants to Mitochondria by Conjugation to Lipophilic Cations. Annu. Rev. Pharmacol. Toxicol. 2007, 47, 629–656. [Google Scholar] [CrossRef] [PubMed]
  26. Gane, E.J.; Weilert, F.; Orr, D.W.; Keogh, G.F.; Gibson, M.; Lockhart, M.M.; Frampton, C.M.; Taylor, K.M.; Smith, R.A.; Murphy, M.P. The mitochondria-targeted anti-oxidant mitoquinone decreases liver damage in a phase ii study of hepatitis c patients. Liver. Int. 2010, 30, 1019–1026. [Google Scholar] [CrossRef] [PubMed]
  27. Cirilli, I.; Damiani, E.; Dludla, P.V.; Hargreaves, I.; Marcheggiani, F.; Millichap, L.E.; Orlando, P.; Silvestri, S.; Tiano, L. Role of Coen-zyme Q10 in Health and Disease: An Update on the Last 10 Years (2010–2020). Antioxidants 2021, 10, 1325. [Google Scholar] [CrossRef] [PubMed]
  28. Testai, L.; Martelli, A.; Flori, L.; Cicero, A.; Colletti, A. Coenzyme Q10: Clinical Applications beyond Cardiovascular Diseases. Nutrients 2021, 13, 1697. [Google Scholar] [CrossRef] [PubMed]
  29. Sharma, A.; Fonarow, G.C.; Butler, J.; Ezekowitz, J.A.; Felker, G.M. Coenzyme Q10 and Heart Failure: A State-of-the-Art Review. Circ. Hear. Fail. 2016, 9, e002639. [Google Scholar] [CrossRef]
  30. Garrido-Maraver, J.; Cordero, M.D.; Oropesa-Avila, M.; Vega, A.F.; de la Mata, M.; Pavon, A.D.; Alcocer-Gomez, E.; Calero, C.P.; Paz, M.V.; Alanis, M.; et al. Clinical applications of coenzyme Q10. Front. Biosci. 2014, 19, 619–633. [Google Scholar] [CrossRef] [Green Version]
  31. Zozina, V.I.; Covantev, S.; Goroshko, O.A.; Krasnykh, L.M.; Kukes, V.G. Coenzyme Q10 in Cardiovascular and Metabolic Diseases: Current State of the Problem. Curr. Cardiol. Rev. 2018, 14, 164–174. [Google Scholar] [CrossRef]
  32. Siebrecht, S.; Chan, D.Y.L.; Rosenfeld, F.; Lin, K.W. Coenzyme Q10 and ubiquinol for physical performance. In Coenzyme Q10: From Fact to Fiction; Hargreaves, I., Ed.; Nova Science Pub Inc.: London, UK, 2015. [Google Scholar]
  33. Malm, C.; Svensson, M. Effects of Ubiquinone-10 Supplementation on Physical Performance in Humans. In Coenzyme Q: Molecular Mechanisms in Health and Disease, 1st ed.; Kagan, V.E., Quinn, P.J., Eds.; CRC Press: Boca Raton, FL, USA, 2000. [Google Scholar] [CrossRef]
  34. Sarmiento, A.; Díaz-Castro, J.; Pulido-Moran, M.; Kajarabille, N.; Guisado, R.; Ochoa, J.J. Coenzyme Q10 Supplementation and Exercise in Healthy Humans: A Systematic Review. Curr. Drug Metab. 2016, 17, 345–358. [Google Scholar] [CrossRef]
  35. Cumpston, M.; Li, T.; Page, M.J.; Chandler, J.; Welch, V.A.; Higgins, J.P.; Thomas, J. Updated guidance for trusted systematic reviews: A new edition of the Cochrane Handbook for Systematic Reviews of Interventions. Cochrane Database Syst. Rev. 2019, 10, D142. [Google Scholar] [CrossRef] [Green Version]
  36. Higgins, J.P.; Thomas, J.; Cumpston, M.; Li, T.; Page, M.J.; Welch, V.A. Cochrane Handbook for Systematic Reviews of Interventions, 2nd ed.; John Wiley & Sons, Inc.: Chichester, UK, 2019. [Google Scholar]
  37. Hutton, B.; Catalá-López, F.; Moher, D. La extensión de la declaración PRISMA para revisiones sistemáticas que incorporan metaanálisis en red: PRISMA-NMA [The PRISMA statement extension for systematic reviews incorporating network meta-analysis: PRISMA-NMA]. Med. Clin. 2016, 147, 262–266. [Google Scholar] [CrossRef]
  38. Amadio, E.; Palermo, R.; Peloni, G.; Littarru, G.P. Effect of CoQ10 administration on V02max and diastolic function in athletes. In Biomedical and Clinical Aspects of Coenzyme Q; Folkers, K., Littarru, G.P., Yamagami, T., Eds.; Elsevier Science Publishers: Amsterdam, The Netherlands, 1991; pp. 525–533. [Google Scholar]
  39. Armanfar, M.; Jafari, A.; Dehghan, G.R.; Abdizadeh, L. Effect of coenzyme Q10 supplementation on exercise-induced response of inflammatory indicators and blood lactate in male runners. Med. J. Islam. Repub. Iran 2015, 29, 202. [Google Scholar] [PubMed]
  40. Bonetti, A.; Solito, F.; Carmosino, G.; Bargossi, A.M.; Fiorella, P.L. Effect of ubidecarenone oral treatment on aerobic power in mid-dle-aged trained subjects. J. Sports Med. Phys. Fit. 2000, 40, 51–57. [Google Scholar]
  41. Braun, B.; Clarkson, P.M.; Freedson, P.S.; Kohl, R.L. Effects of Coenzyme Q10 Supplementation on Exercise Performance, VO2max, and Lipid Peroxidation in Trained Cyclists. Int. J. Sport Nutr. 1991, 1, 353–365. [Google Scholar] [CrossRef]
  42. Cerioli, G.; Tirelli, F.; Musiani, L. Effect of Coenzyme Q10 on the metabolic Response to work. In Biomedical and Clinical Aspects of Coenzyme Q; Folkers, K., Littarru, G.P., Yamagami, T., Eds.; Elsevier Science Publishers: Amsterdam, The Netherlands, 1991; pp. 521–524. [Google Scholar]
  43. Cinquegrana, G.; Meccariello, P.; Spinelli, L.; Romano, M.; Sotgiu, P.; Cristiano, C.; Ferrara, M.; Giardino, L. Effetti del coenzima Q10 sulla tolleranza all’esercizio fisico e sulla performance cardiaca in soggetti normali non allenati [Effects of coenzyme Q10 on physical exercise tolerance and cardiac performance in normal untrained subjects]. Clin. Ter. 1987, 123, 15–20. [Google Scholar]
  44. Ciocoi-Pop, R.; Tache, S.; Bondor, C. The effect of Coenzyme Q10 administration on effort capacity of athletes (Note I). Palestri-ca Milen III—Civilizaţie Şi Sport 2009, 10, 77–79. [Google Scholar]
  45. Cooke, M.; Iosia, M.; Buford, T.; Shelmadine, B.; Hudson, G.; Kerksick, C.; Rasmussen, C.; Greenwood, M.; Leutholtz, B.; Willoughby, D.; et al. Effects of acute and 14-day coenzyme Q10 supplementation on exercise performance in both trained and untrained individuals. J. Int. Soc. Sports Nutr. 2008, 5, 8. [Google Scholar] [CrossRef] [Green Version]
  46. Díaz-Castro, J.; Guisado, R.; Kajarabille, N.; García, C.; Guisado, I.M.; de Teresa, C.; Ochoa, J.J. Coenzyme Q10 supplementation ameliorates inflammatory signaling and oxidative stress associated with strenuous exercise. Eur. J. Nutr. 2011, 51, 791–799. [Google Scholar] [CrossRef]
  47. Drobnic, F.; Riera, J.; Artuch, R.; Jou, C.; Codina, A.; Montero, R.; Paredes-Fuentes, A.J.; Domingo, J.C.; Banquells, M.; Riva, A.; et al. Efficient Muscle Distribution Reflects the Positive Influence of Coenzyme Q10 Phytosome in Healthy Aging Athletes after Stressing Exercise. J. Food Sci. Nutr. Res. 2020, 3, 262–275. [Google Scholar] [CrossRef]
  48. Emami, A.; Tofighi, A.; Asri-Rezaei, S.; Bazargani-Gilani, B. The effect of short-term coenzyme Q10supplementation and pre-cooling strategy on cardiac damage markers in elite swimmers. Br. J. Nutr. 2018, 119, 381–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Fiorella, P.L.; Bargossi, A.M.; Grossi, G.; Motta, R.; Senaldi, R.; Battino, M.; Sassi, S.; Sprovieri, G.; Lubich, T.; Folkers, K.; et al. Metabolic effects of coenzyme Q10 treatment in high level athletes. In Biomedical and Clinical Aspects of Coenzyme Q; Folkers, K., Littarru, G.P., Yamagami, T., Eds.; Elsevier Science Publishers: Amsterdam, The Netherlands, 1991; pp. 513–520. [Google Scholar]
  50. Cordero, M.J.A. Efecto del Phlebodium decumanum y de la coenzyma Q10 sobre el rendimiento deportivo en jugadores profesionales de voleibol. Nutr. Hosp. 2015, 31, 401–414. [Google Scholar] [CrossRef]
  51. Geiss, K.; Hamm, M.; Littarru, G.P.; Folkers, K.; Enzmann, F. Steigerung der körperlichen Leistungsfähigkeit von Ausdauerathleten mit Hilfen von Q10 Monopräparat. In Energie und Schutz Coenzym Q10 Fakten und Perspektivem in der Biologie und Medizin; Littarru, G.P., Ed.; Litografica Iride: Rome, Italy, 2004; pp. 84–86. [Google Scholar]
  52. Gökbel, H.; Gül, I.; Belviranl, M.; Okudan, N. The Effects of Coenzyme Q10 Supplementation on Performance During Repeated Bouts of Supramaximal Exercise in Sedentary Men. J. Strength Cond. Res. 2010, 24, 97–102. [Google Scholar] [CrossRef] [PubMed]
  53. Gokbel, H.; Turk, S.; Okudan, N.; Atalay, H.; Belviranli, M.; Gaipov, A.; Solak, Y. Effects of Coenzyme Q10 Supplementation on Exercise Performance and Markers of Oxidative Stress in Hemodialysis Patients: A Double-Blind Placebo-Controlled Crossover Trial. Am. J. Ther. 2016, 23, e1736–e1743. [Google Scholar] [CrossRef]
  54. Guerra, G.; Ballardini, E.; Lippa, S.; Oradei, A.; Litarru, G. Effetto della somministrazione di Ubidecarenone nel consumo massimo di ossigeno e sulla performance fisica in un gruppo di giovani ciclisti. Med. Sport 1987, 40, 359–364. [Google Scholar]
  55. Gül, I.; Gökbel, H.; Belviranli, M.; Okudan, N.; Büyükbaş, S.; Başarali, K. Oxidative stress and antioxidant defense in plasma after repeated bouts of supramaximal exercise: The effect of coenzyme Q10. J. Sports Med. Phys. Fit. 2011, 51, 305–312. [Google Scholar]
  56. Ho, C.-C.; Chang, P.-S.; Chen, H.-W.; Lee, P.-F.; Chang, Y.-C.; Tseng, C.-Y.; Lin, P.-T. Ubiquinone Supplementation with 300 mg on Glycemic Control and Antioxidant Status in Athletes: A Randomized, Double-Blinded, Placebo-Controlled Trial. Antioxidants 2020, 9, 823. [Google Scholar] [CrossRef]
  57. Kaikkonen, J.; Kosonen, L.; Nyyssönen, K.; Porkkala-Sarataho, E.; Salonen, R.; Korpela, H.; Salonen, J.T. Effect of combined coenzyme Q10 and d-α-tocopheryl acetate supplementation on exercise-induced lipid peroxidation and muscular damage: A placebo-controlled double-blind study in marathon runners. Free Radic. Res. 1998, 29, 85–92. [Google Scholar] [CrossRef]
  58. Laaksonen, R.; Fogelholm, M.; Himberg, J.-J.; Laakso, J.; Salorinne, Y. Ubiquinone supplementation and exercise capacity in trained young and older men. Eur. J. Appl. Physiol. 1995, 72, 95–100. [Google Scholar] [CrossRef]
  59. Leelarungrayub, D.; Sawattikanon, N.; Klaphajone, J.; Pothongsunan, P.; Bloomer, R.J. Coenzyme Q10 Supplementation Decreases Oxidative Stress and Improves Physical Performance in Young Swimmers: A Pilot Study. Open Sports Med. J. 2010, 4, 1–8. [Google Scholar] [CrossRef] [Green Version]
  60. Malm, C.; Svensson, M.; Sjoberg, B.; Ekblom, B.; Sjodin, B. Supplementation with ubiquinone-10 causes cellular damage during intense exercise. Acta Physiol. Scand. 1996, 157, 511–512. [Google Scholar] [CrossRef]
  61. Malm, C.; Svensson, M.; Ekblom, B.; Sjödin, B. Effects of ubiquinone-10 supplementation and high intensity training on physical performance in humans. Acta Physiol. Scand. 1997, 161, 379–384. [Google Scholar] [CrossRef] [PubMed]
  62. Mizuno, K.; Tanaka, M.; Nozaki, S.; Mizuma, H.; Ataka, S.; Tahara, T.; Sugino, T.; Shirai, T.; Kajimoto, Y.; Kuratsune, H.; et al. Antifatigue effects of coenzyme Q10 during physical fatigue. Nutrition 2008, 24, 293–299. [Google Scholar] [CrossRef] [PubMed]
  63. Nielsen, A.N.; Mizuno, M.; Ratkevicius, A.; Mohr, T.; Rohde, M.; Mortensen, S.A.; Quistorff, B. No Effect of Antioxidant Supplementation in Triathletes on Maximal Oxygen Uptake, 31P-NMRS Detected Muscle Energy Metabolism and Muscle Fatigue. Int. J. Sports Med. 1999, 20, 154–158. [Google Scholar] [CrossRef] [PubMed]
  64. Okudan, N.; Belviranli, M.; Torlak, S. Coenzyme Q10 does not prevent exercise-induced muscle damage and oxidative stress in sedentary men. J. Sports Med. Phys. Fit. 2018, 58, 889–894. [Google Scholar] [CrossRef] [PubMed]
  65. Östman, B.; Sjödin, A.; Michaëlsson, K.; Byberg, L. Coenzyme Q10 supplementation and exercise-induced oxidative stress in humans. Nutrition 2012, 28, 403–417. [Google Scholar] [CrossRef]
  66. Porter, D.A.; Costill, D.L.; Zachwieja, J.J.; Krzeminski, K.; Fink, W.J.; Wagner, E.; Folkers, K. The Effect of Oral Coenzyme Q10 on the Exercise Tolerance of Middle-Aged, Untrained Men. Int. J. Sports Med. 1995, 16, 421–427. [Google Scholar] [CrossRef]
  67. Snider, I.P.; Bazzarre, T.L.; Murdoch, S.D.; Goldfarb, A. Effects of Coenzyme Athletic Performance System as an Ergogenic Aid on Endurance Performance to Exhaustion. Int. J. Sport Nutr. 1992, 2, 272–286. [Google Scholar] [CrossRef] [Green Version]
  68. Svensson, M.; Malm, C.; Tonkonogi, M.; Ekblom, B.; Sjödin, B.; Sahlin, K. Effect of Q10 Supplementation on Tissue Q10 Levels and Adenine Nucleotide Catabolism During High-Intensity Exercise. Int. J. Sport Nutr. 1999, 9, 166–180. [Google Scholar] [CrossRef]
  69. Tauler, P.; Ferrer, M.D.; Sureda, A.; Pujol, P.; Drobnic, F.; Tur, J.A.; Pons, A. Supplementation with an antioxidant cocktail containing coenzyme Q prevents plasma oxidative damage induced by soccer. Eur. J. Appl. Physiol. 2008, 104, 777–785. [Google Scholar] [CrossRef]
  70. Ferrer, M.D.; Tauler, P.; Sureda, A.; Pujol, P.; Drobnic, F.; Tur, J.A.; Pons, A. A Soccer Match’s Ability to Enhance Lymphocyte Capability to Produce ROS and Induce Oxidative Damage. Int. J. Sport Nutr. Exerc. Metab. 2009, 19, 243–258. [Google Scholar] [CrossRef]
  71. Vanfraechem, J.; Folkers, K. Coenzyme Q10 and physical performance. In Biomedical and Clinical Aspects of Coenzyme Q; Folkers, K., Yamamura, Y., Eds.; Elsevier/North-Holland Biomedical Publishers: Amsterdam, The Netherlands, 1981; Volume 3, pp. 235–241. [Google Scholar]
  72. Weston, S.B.; Zhou, S.; Weatherby, R.P.; Robson, S.J. Does Exogenous Coenzyme Q10 Affect Aerobic Capacity in Endurance Athletes? Int. J. Sport Nutr. 1997, 7, 197–206. [Google Scholar] [CrossRef] [Green Version]
  73. Wyss, V.; Lubich, T.; Ganzit, G.; Cesaretti, D.; Fiorella, P.; Dei Rocini, C.; Battino, M. Remarks of prolonged ubiquinone administration in physical exercise. In Highlights in Ubiquinone Research; Lenaz, G., Ed.; Taylor & Francis: London, UK, 1990; pp. 303–306. [Google Scholar]
  74. Yamabe, H.; Fukuzaki, H. The beneficial effect of coenzyme Q10 on the impaired aerobic function in middle aged women without organic disease. In Biomedical and Clinical Aspects of Coenzyme Q; Folkers, K., Littarru, G.P., Yamagami, T., Eds.; Elsevier Science Publishers: Amsterdam, The Netherlands, 1991; pp. 535–540. [Google Scholar]
  75. Ylikoski, T.; Piirainen, J.; Hanninen, O.; Penttinen, J. The effect of coenzyme Q10 on the exercise performance of cross-country skiers. Mol. Asp. Med. 1997, 18, 283–290. [Google Scholar] [CrossRef]
  76. Zeppilli, P.; Merlino, B.; De Luca, A.; Palmieri, V.; Santini, C.; Vannicelli, R.; Littarru, G.P. Influence of Coenzyme-Q10 on physical work capacity in athletes, sedentary people and patients with mitochondrial disease. In Biomedical and Clinical Aspects of Coenzyme Q; Folkers, K., Littarru, G.P., Yamagami, T., Eds.; Elsevier Science Publishers: Amsterdam, The Netherlands, 1991; pp. 541–545. [Google Scholar]
  77. Zheng, A.; Moritani, T. Influence of CoQ10 on autonomic nervous activity and energy metabolism during exercise in healthy subjects. J. Nutr. Sci. Vitaminol. 2008, 54, 286–290. [Google Scholar] [CrossRef] [Green Version]
  78. Zhou, S.; Zhang, Y.; Davie, A.; Marshall-Gradisnik, S.; Hu, H.; Wang, J.; Brushett, D. Muscle and plasma coenzyme Q10 concentration, aerobic power and exercise economy of healthy men in response to four weeks of supplementation. J. Sports Med. Phys. Fit. 2005, 45, 337–346. [Google Scholar]
  79. Zuliani, U.; Bonetti, A.; Campana, M.; Cerioli, G.; Solito, F.; Novarini, A. The influence of ubiquinone (Co Q10) on the metabolic response to work. J. Sports Med. Phys. Fit. 1989, 29, 57–62. [Google Scholar]
  80. Pham, T.; MacRae, C.L.; Broome, S.C.; D’Souza, R.F.; Narang, R.; Wang, H.W.; Mori, T.A.; Hickey, A.J.R.; Mitchell, C.J.; Merry, T.L. MitoQ and CoQ10 supplementation mildly suppresses skeletal muscle mitochondrial hydrogen peroxide levels without impacting mitochondrial function in middle-aged men. Eur. J. Appl. Physiol. 2020, 120, 1657–1669. [Google Scholar] [CrossRef]
  81. Alf, D.; Schmidt, M.E.; Siebrecht, S.C. Ubiquinol supplementation enhances peak power production in trained athletes: A double-blind, placebo controlled study. J. Int. Soc. Sports Nutr. 2013, 10, 24. [Google Scholar] [CrossRef] [Green Version]
  82. Bloomer, R.J.; Canale, R.E.; McCarthy, C.G.; Farney, T.M. Impact of Oral Ubiquinol on Blood Oxidative Stress and Exercise Performance. Oxidative Med. Cell. Longev. 2012, 2012, 465020. [Google Scholar] [CrossRef] [Green Version]
  83. Diaz-Castro, J.; Mira-Rufino, P.J.; Moreno-Fernandez, J.; Chirosa, I.; Chirosa, J.L.; Guisado, R.; Ochoa, J.J. Ubiquinol supplementation modulates energy metabolism and bone turnover during high intensity exercise. Food Funct. 2020, 11, 7523–7531. [Google Scholar] [CrossRef]
  84. Diaz-Castro, J.; Moreno-Fernandez, J.; Chirosa, I.; Chirosa, L.J.; Guisado, R.; Ochoa, J.J. Beneficial Effect of Ubiquinol on Hematological and Inflammatory Signaling during Exercise. Nutrients 2020, 12, 424. [Google Scholar] [CrossRef] [Green Version]
  85. Kizaki, K.; Terada, T.; Arikawa, H.; Tajima, T.; Imai, H.; Takahashi, T.; Era, S. Effect of reduced coenzyme Q10 (ubiquinol) supple-mentation on blood pressure and muscle damage during kendo training camp: A double-blind, randomized controlled study. J. Sports Med. Phys. Fit. 2015, 55, 797–804. [Google Scholar]
  86. Kon, M.; Tanabe, K.; Akimoto, T.; Kimura, F.; Tanimura, Y.; Shimizu, K.; Okamoto, T.; Kono, I. Reducing exercise-induced muscular injury in kendo athletes with supplementation of coenzyme Q10. Br. J. Nutr. 2008, 100, 903–909. [Google Scholar] [CrossRef] [Green Version]
  87. Kunching, S.; Nararatwanchai, T.; Chalermchai, T.; Wongsupasawat, K.; Sitiprapaporn, P.; Thipsiriset, A. The effects of ubiquinol supplementation on clinical parameters and physical performance in trained men. Songklanakarin J. Sci. Technol. 2022, 44, 231–235. [Google Scholar]
  88. Orlando, P.; Silvestri, S.; Galeazzi, R.; Antonicelli, R.; Marcheggiani, F.; Cirilli, I.; Bacchetti, T.; Tiano, L. Effect of ubiquinol supplementation on biochemical and oxidative stress indexes after intense exercise in young athletes. Redox Rep. 2018, 23, 136–145. [Google Scholar] [CrossRef]
  89. Sarmiento, A.; Diaz-Castro, J.; Pulido-Moran, M.; Moreno-Fernandez, J.; Kajarabille, N.; Chirosa, I.; Guisado, I.M.; Chirosa, L.J.; Guisado, R.; Ochoa, J.J. Short-term ubiquinol supplementation reduces oxidative stress associated with strenuous exercise in healthy adults: A randomized trial. BioFactors 2016, 42, 612–622. [Google Scholar] [CrossRef]
  90. Suzuki, Y.; Nagato, S.; Sakuraba, K.; Morio, K.; Sawaki, K. Short-term ubiquinol-10 supplementation alleviates tissue damage in muscle and fatigue caused by strenuous exercise in male distance runners. Int. J. Vitam. Nutr. Res. 2021, 91, 261–270. [Google Scholar] [CrossRef] [PubMed]
  91. Shill, D.D.; Southern, W.M.; Willingham, T.B.; Lansford, K.A.; McCully, K.K.; Jenkins, N.T. Mitochondria-specific antioxidant supplementation does not influence endurance exercise training-induced adaptations in circulating angiogenic cells, skeletal muscle oxidative capacity or maximal oxygen uptake. J. Physiol. 2016, 594, 7005–7014. [Google Scholar] [CrossRef] [PubMed]
  92. Williamson, J.; Hughes, C.M.; Cobley, J.N.; Davison, G.W. The mitochondria-targeted antioxidant MitoQ, attenuates exercise-induced mitochondrial DNA damage. Redox Biol. 2020, 36, 101673. [Google Scholar] [CrossRef] [PubMed]
  93. Broome, S.C.; Braakhuis, A.J.; Mitchell, C.J.; Merry, T.L. Mitochondria-targeted antioxidant supplementation improves 8 km time trial performance in middle-aged trained male cyclists. J. Int. Soc. Sports Nutr. 2021, 18, 58. [Google Scholar] [CrossRef] [PubMed]
  94. Guyatt, G.H.; Oxman, A.D.; Vist, G.E.; Kunz, R.; Falck-Ytter, Y.; Alonso-Coello, P.; Schünemann, H.J.; GRADE Working Group. GRADE: An emerging consensus on rating quality of evidence and strength of recommendations. BMJ 2008, 336, 924–926. [Google Scholar] [CrossRef] [Green Version]
  95. Harbour, R.; Miller, J. A new system for grading recommendations in evidence based guidelines. BMJ 2001, 323, 334–336. [Google Scholar] [CrossRef] [Green Version]
  96. De Morton, N.A. The PEDro scale is a valid measure of the methodological quality of clinical trials: A demographic study. Aust. J. Physiother. 2009, 55, 129–133. [Google Scholar] [CrossRef] [Green Version]
  97. Maher, C.G.; Sherrington, C.; Herbert, R.D.; Moseley, A.M.; Elkins, M. Reliability of the PEDro Scale for Rating Quality of Randomized Controlled Trials. Phys. Ther. 2003, 83, 713–721. [Google Scholar] [CrossRef] [Green Version]
  98. Paredes-Fuentes, A.J.; Montero, R.; Codina, A.; Jou, C.; Fernández, G.; Maynou, J.; Santos-Ocaña, C.; Riera, J.; Navas, P.; Drobnic, F.; et al. Coenzyme Q10 Treatment Monitoring in Different Human Biological Samples. Antioxidants 2020, 9, 979. [Google Scholar] [CrossRef]
  99. Sánchez-Cuesta, A.; Cortés-Rodríguez, A.B.; Navas-Enamorado, I.; Lekue, J.A.; Viar, T.; Axpe, M.; Navas, P.; López-Lluch, G. High coenzyme Q10 plasma levels improve stress and damage markers in professional soccer players during competition. Int. J. Vitam. Nutr. Res. 2020, 8, 1–12. [Google Scholar] [CrossRef]
  100. Hosoe, K.; Kitano, M.; Kishida, H.; Kubo, H.; Fujii, K.; Kitahara, M. Study on safety and bioavailability of ubiquinol (Kaneka QH™) after single and 4-week multiple oral administration to healthy volunteers. Regul. Toxicol. Pharmacol. 2007, 47, 19–28. [Google Scholar] [CrossRef]
  101. Bhagavan, H.N.; Chopra, R.K. Coenzyme Q10: Absorption, tissue uptake, metabolism and pharmacokinetics. Free Radic. Res. 2006, 40, 445–453. [Google Scholar] [CrossRef]
  102. Weis, M.; Mortensen, S.; Rassing, M.; Møller-Sonnergaard, J.; Poulsen, G.; Rasmussen, S. Bioavailability of four oral Coenzyme Q10 formulations in healthy volunteers. Mol. Asp. Med. 1994, 15, s273–s280. [Google Scholar] [CrossRef]
  103. Schulz, C.; Obermüller-Jevic, U.C.; Hasselwander, O.; Bernhardt, J.; Biesalski, H.K. Comparison of the relative bioavailability of different coenzyme Q10formulations with a novel solubilizate (Solu™ Q10). Int. J. Food Sci. Nutr. 2006, 57, 546–555. [Google Scholar] [CrossRef]
  104. Petrangolini, G.; Ronchi, M.; Frattini, E.; De Combarieu, E.; Allegrini, P.; Riva, A. A New Food-grade Coenzyme Q10 Formulation Improves Bioavailability: Single and Repeated Pharmacokinetic Studies in Healthy Volunteers. Curr. Drug Deliv. 2019, 16, 759–767. [Google Scholar] [CrossRef]
  105. Weber, C.; Bysted, A.; Hølmer, G. Coenzyme Q10 in the diet-daily intake and relative bioavailability. Mol. Asp. Med. 1997, 18, 251–254. [Google Scholar] [CrossRef]
  106. Rakuša, T.; Kristl, A.; Roškar, R. Stability of Reduced and Oxidized Coenzyme Q10 in Finished Products. Antioxidants 2021, 10, 360. [Google Scholar] [CrossRef]
  107. Singh, R.B.; Niaz, M.A.; Kumar, A.; Sindberg, C.D.; Moesgaard, S.; Littarru, G.P. Effect on absorption and oxidative stress of different oral Coenzyme Q10dosages and intake strategy in healthy men. BioFactors 2005, 25, 219–224. [Google Scholar] [CrossRef]
  108. Lenaz, G. Coenzyme Q saturation kinetics of mitochondrial enzymes: Theory, experimental aspects and biomedical implica-tions. In Biomedical and Clinical Aspects of Coenzyme Q; Folkers, K., Yamagami, T., Littarru, G.P., Eds.; Elsevier Science Publishers: Amsterdam, The Netherlands, 1991; pp. 11–18. [Google Scholar]
  109. Molyneux, S.L.; Florkowski, C.M.; Lever, M.; George, P.M. Biological Variation of Coenzyme Q10. Clin. Chem. 2005, 51, 455–457. [Google Scholar] [CrossRef] [Green Version]
  110. Duncan, A.J.; Heales, S.J.; Mills, K.; Eaton, S.; Land, J.M.; Hargreaves, I.P. Determination of Coenzyme Q10 Status in Blood Mononuclear Cells, Skeletal Muscle, and Plasma by HPLC with Di-Propoxy-Coenzyme Q10 as an Internal Standard. Clin. Chem. 2005, 51, 2380–2382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Okamoto, T.; Mizuta, K.; Mizobuchi, S.; Usui, A.; Takahashi, T.; Fujimoto, S.; Kishi, T. Decreased serum ubiquinol-10 levels in healthy subjects during exercise at maximal oxygen uptake. BioFactors 2000, 11, 31–33. [Google Scholar] [CrossRef]
  112. Battino, M.; Amadio, E.; Oradei, A.; Littarru, G. Metabolic and antioxidant markers in the plasma of sportsmen from a Mediterranean town performing non-agonistic activity. Mol. Asp. Med. 1997, 18, 241–245. [Google Scholar] [CrossRef]
  113. Kwong, L.K.; Kamzalov, S.; Rebrin, I.; Bayne, A.-C.V.; Jana, C.K.; Morris, P.; Forster, M.J.; Sohal, R.S. Effects of coenzyme Q10 administration on its tissue concentrations, mitochondrial oxidant generation, and oxidative stress in the rat. Free Radic. Biol. Med. 2002, 33, 627–638. [Google Scholar] [CrossRef]
  114. Tsai, K.L.; Huang, Y.H.; Kao, C.L.; Yang, D.M.; Lee, H.C.; Chou, H.Y.; Chen, Y.C.; Chiou, G.Y.; Chen, L.H.; Yang, Y.P.; et al. A novel mechanism of coenzyme Q10 protects against human endothelial cells from oxidative stress-induced injury by modulating NO-related pathways. J. Nutr. Biochem. 2012, 23, 458–468. [Google Scholar] [CrossRef]
  115. Belviranlı, M.; Okudan, N. Effect of Coenzyme Q10 Alone and in Combination with Exercise Training on Oxidative Stress Biomarkers in Rats. Int. J. Vitam. Nutr. Res. 2018, 88, 126–136. [Google Scholar] [CrossRef]
  116. Sohal, R.S.; Forster, M.J. Coenzyme Q, oxidative stress and aging. Mitochondrion 2007, 7, S103–S111. [Google Scholar] [CrossRef] [Green Version]
  117. Gutierrez-Mariscal, F.M.; Larriva, A.P.A.-D.; Limia-Perez, L.; Romero-Cabrera, J.L.; Yubero-Serrano, E.M.; López-Miranda, J. Coenzyme Q10 Supplementation for the Reduction of Oxidative Stress: Clinical Implications in the Treatment of Chronic Diseases. Int. J. Mol. Sci. 2020, 21, 7870. [Google Scholar] [CrossRef]
  118. Yamamoto, Y. Coenzyme Q10 redox balance and a free radical scavenger drug. Arch. Biochem. Biophys. 2016, 595, 132–135. [Google Scholar] [CrossRef]
  119. Davies, K.J.; Quintanilha, A.T.; Brooks, G.A.; Packer, L. Free radicals and tissue damage produced by exercise. Biochem. Biophys. Res. Commun. 1982, 107, 1198–1205. [Google Scholar] [CrossRef]
  120. Halliwell, B. Biochemistry of oxidative stress. Biochem. Soc. Trans. 2007, 35, 1147–1150. [Google Scholar] [CrossRef]
  121. Leeuwenburgh, C. Oxidative Stress and Antioxidants in Exercise. Curr. Med. Chem. 2001, 8, 829–838. [Google Scholar] [CrossRef] [Green Version]
  122. Reid, M.B.; Haack, K.E.; Franchek, K.M.; Valberg, P.A.; Kobzik, L.; West, M.S. Reactive oxygen in skeletal muscle. I. Intracellular oxidant kinetics and fatigue in vitro. J. Appl. Physiol. 1992, 73, 1797–1804. [Google Scholar] [CrossRef]
  123. Aguiló, A.; Tauler, P.; Fuentespina, E.; Tur, J.A.; Córdova, A.; Pons, A. Antioxidant response to oxidative stress induced by exhaustive exercise. Physiol. Behav. 2005, 84, 1–7. [Google Scholar] [CrossRef]
  124. Pravst, I.; Rodriguez Aguilera, J.C.; Cortes Rodriguez, A.B.; Jazbar, J.; Locatelli, I.; Hristov, H.; Žmitek, K. Comparative Bioavailability of Different Coenzyme Q10 Formulations in Healthy Elderly Individuals. Nutrients 2020, 12, 784. [Google Scholar] [CrossRef] [Green Version]
  125. Clarkson, P.M.; Nosaka, K.; Braun, B. Muscle function after exercise-induced muscle damage and rapid adaptation. Med. Sci. Sports Exerc. 1992, 24, 512–520. [Google Scholar] [CrossRef]
  126. Tiidus, P.M. Radical species in inflammation and overtraining. Can. J. Physiol. Pharmacol. 1998, 76, 533–538. [Google Scholar] [CrossRef]
  127. Córdova-Martínez, A.; Caballero-García, A.; Bello, H.J.; Perez-Valdecantos, D.; Roche, E. Effects of Eccentric vs. Concentric Sports on Blood Muscular Damage Markers in Male Professional Players. Biology 2022, 11, 343. [Google Scholar] [CrossRef] [PubMed]
  128. Proske, U.; Morgan, D.L. Muscle damage from eccentric exercise: Mechanism, mechanical signs, adaptation and clinical applications. J. Physiol. 2001, 537, 333–345. [Google Scholar] [CrossRef] [PubMed]
  129. Zhai, J.; Bo, Y.; Lu, Y.; Liu, C.; Zhang, L. Effects of Coenzyme Q10 on Markers of Inflammation: A Systematic Review and Meta-Analysis. PLoS ONE 2017, 12, e0170172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Córdova, A.; Sureda, A.; Pons, A.; Alvarez-Mon, M. Modulation of TNF-α, TNF-α receptors and IL-6 after treatment with AM3 in professional cyclists. J. Sports Med. Phys. Fit. 2015, 55, 345–351. [Google Scholar]
  131. Córdova-Martínez, A.; Caballero-García, A.; Bello, H.; Pérez-Valdecantos, D.; Roche, E. Effect of Glutamine Supplementation on Muscular Damage Biomarkers in Professional Basketball Players. Nutrients 2021, 13, 2073. [Google Scholar] [CrossRef]
  132. Martínez, A.C.; Pons, M.M.; Gomila, A.S.; Marí, J.A.T.; Biescas, A.P. Changes in circulating cytokines and markers of muscle damage in elite cyclists during a multi-stage competition. Clin. Physiol. Funct. Imaging 2014, 35, 351–358. [Google Scholar] [CrossRef]
  133. Peake, J.M.; Suzuki, K.; Coombes, J. The influence of antioxidant supplementation on markers of inflammation and the relationship to oxidative stress after exercise. J. Nutr. Biochem. 2007, 18, 357–371. [Google Scholar] [CrossRef]
  134. Ji, L.L.; Gomez-Cabrera, M.C.; Vina, J. Role of nuclear factor κB and mitogen-activated protein kinase signaling in exercise-induced antioxidant enzyme adaptation. Appl. Physiol. Nutr. Metab. 2007, 32, 930–935. [Google Scholar] [CrossRef]
  135. Drobnic, F. Las agujetas, ¿una entidad clínica con nombre inapropiado? (Mecanismos de aparición, evolución y tratamiento). Apunts. Medicina de l’Esport 1989, 26, 125–134. [Google Scholar]
  136. Cordova, A.; Monserrat, J.; Villa, G.; Reyes, E.; Soto, M.A.-M. Effects of AM3 (Inmunoferon®) on increased serum concentrations of interleukin-6 and tumour necrosis factor receptors I and II in cyclists. J. Sports Sci. 2006, 24, 565–573. [Google Scholar] [CrossRef]
  137. Schwane, J.A.; Johnson, S.R.; Vandenakker, C.B.; Armstrong, R.B. Delayed-onset muscular soreness and plasma CPK and LDH activities after downhill running. Med. Sci. Sports Exerc. 1983, 15, 51–56. [Google Scholar] [CrossRef]
  138. Peake, J.M.; Suzuki, K.; Wilson, G.; Hordern, M.; Nosaka, K.; Mackinnon, L.; Coombes, J.S. Exercise-Induced Muscle Damage, Plasma Cytokines, and Markers of Neutrophil Activation. Med. Sci. Sports Exerc. 2005, 37, 737–745. [Google Scholar] [CrossRef] [Green Version]
  139. Shimomura, Y.; Suzuki, M.; Sugiyama, S.; Hanaki, Y.; Ozawa, T. Protective effect of coenzyme Q10 on exercise-induced muscular injury. Biochem. Biophys. Res. Commun. 1991, 176, 349–355. [Google Scholar] [CrossRef]
  140. Brancaccio, P.; Maffulli, N.; Limongelli, F.M. Creatine kinase monitoring in sport medicine. Br. Med. Bull. 2007, 81–82, 209–230. [Google Scholar] [CrossRef]
  141. Bessa, A.L.; Oliveira, V.N.; Agostini, G.G.; Oliveira, R.J.; Oliveira, A.C.; White, G.E.; Wells, G.D.; Teixeira, D.N.; Espindola, F.S. Exercise Intensity and Recovery: Biomarkers of Injury, Inflammation, and Oxidative Stress. J. Strength Cond. Res. 2016, 30, 311–319. [Google Scholar] [CrossRef]
  142. Brancaccio, P.; Maffulli, N.; Buonauro, R.; Limongelli, F.M. Serum Enzyme Monitoring in Sports Medicine. Clin. Sports Med. 2008, 27, 1–18. [Google Scholar] [CrossRef] [PubMed]
  143. Lee, J.; Clarkson, P.M. Plasma Creatine Kinase Activity and Glutathione after Eccentric Exercise. Med. Sci. Sports Exerc. 2003, 35, 930–936. [Google Scholar] [CrossRef]
  144. Baird, M.F.; Graham, S.M.; Baker, J.S.; Bickerstaff, G.F. Creatine-Kinase- and Exercise-Related Muscle Damage Implications for Muscle Performance and Recovery. J. Nutr. Metab. 2012, 2012, 960363. [Google Scholar] [CrossRef] [Green Version]
  145. Vincent, H. The Effect of Training Status on the Serum Creatine Kinase Response, Soreness and Muscle Function Following Resistance Exercise. Int. J. Sports Med. 1997, 28, 431–437. [Google Scholar] [CrossRef]
  146. Horská, A.; Fishbein, K.W.; Fleg, J.L.; Spencer, R.G.S. The relationship between creatine kinase kinetics and exercise intensity in human forearm is unchanged by age. Am. J. Physiol. Endocrinol. Metab. 2000, 279, E333–E339. [Google Scholar] [CrossRef]
  147. Totsuka, M.; Nakaji, S.; Suzuki, K.; Sugawara, K.; Sato, K. Break point of serum creatine kinase release after endurance exercise. J. Appl. Physiol. 2002, 93, 1280–1286. [Google Scholar] [CrossRef] [PubMed]
  148. Baltusnikas, J.; Venckunas, T.; Kilikevicius, A.; Fokin, A.; Ratkevicius, A. Efflux of creatine kinase from isolated soleus muscle de-pends on age, sex and type of exercise in mice. J. Sports Sci. Med. 2015, 14, 379–385. [Google Scholar] [PubMed]
  149. Baumert, P.; Lake, M.J.; Stewart, C.; Drust, B.; Erskine, R.M. Genetic variation and exercise-induced muscle damage: Implications for athletic performance, injury and ageing. Eur. J. Appl. Physiol. 2016, 116, 1595–1625. [Google Scholar] [CrossRef] [Green Version]
  150. Miyoshi, K.; Kawai, H.; Iwasa, M.; Kusaka, K.; Nishino, H. Autosomal recessive distal muscular dystrophy as a new type of progressive muscular dystrophy. Seventeen cases in eight families including an autopsied case. Brain 1986, 109, 31–54. [Google Scholar] [CrossRef] [PubMed]
  151. Hortobágyi, T.; Denahan, T. Variability in Creatine Kinase: Methodological, Exercise, and Clinically Related Factors. Endoscopy 1989, 10, 69–80. [Google Scholar] [CrossRef]
  152. Nosaka, K.; Newton, M.; Sacco, P. Delayed-onset muscle soreness does not reflect the magnitude of eccentric exercise-induced muscle damage. Scand. J. Med. Sci. Sports 2002, 12, 337–346. [Google Scholar] [CrossRef]
  153. Garry, J.; McShane, J.M. Postcompetition elevation of muscle enzyme levels in professional football players. MedGenMed 2000, 2, E4. [Google Scholar] [PubMed]
  154. Linnane, A.W.; Kopsidas, G.; Zhang, C.; Yarovaya, N.; Kovalenko, S.; Papakostopoulos, P.; Eastwood, H.; Graves, S.; Richardson, M. Cellular Redox Activity of Coenzyme Q 10: Effect of CoQ 10 Supplementation on Human Skeletal Muscle. Free Radic. Res. 2002, 36, 445–453. [Google Scholar] [CrossRef]
  155. Bello, R.I.; Gómez-Díaz, C.; Burón, M.I.; Alcaín, F.J.; Navas, P.; Villalba, J.M. Enhanced anti-oxidant protection of liver membranes in long-lived rats fed on a coenzyme Q10-supplemented diet. Exp. Gerontol. 2005, 40, 694–706. [Google Scholar] [CrossRef]
  156. Długosz, A.; Kuźniar, J.; Sawicka, E.; Marchewka, Z.; Lembas-Bogaczyk, J.; Sajewicz, W.; Boratyńska, M. Oxidative stress and coenzyme Q10 supplementation in renal transplant recipients. Int. Urol. Nephrol. 2004, 36, 253–258. [Google Scholar] [CrossRef]
  157. Billat, V. Fisiología y Metodología del Entrenamiento. de la Teoría a la Práctica; Paidotribo: Barcelona, Spain, 2002. [Google Scholar]
  158. Bassett, D.R., Jr.; Howley, E.T. Limiting factors for maximum oxygen uptake and determinants of endurance performance. Med. Sci. Sports Exerc. 2000, 32, 70–84. [Google Scholar] [CrossRef]
  159. Billat, V.; Renoux, J.C.; Pinoteau, J.; Petit, B.; Koralsztein, J.P. Reproducibility of running time to exhaustion at VO2max in subelite runners. Med. Sci. Sports Exerc. 1994, 26, 254–257. [Google Scholar] [CrossRef]
  160. McLellan, T.M.; Cheung, S.S.; Jacobs, I. Variability of Time to Exhaustion During Submaximal Exercise. Can. J. Appl. Physiol. 1995, 20, 39–51. [Google Scholar] [CrossRef]
  161. Pereira, M.; Freedson, P. Intraindividual Variation of Running Economy in Highly Trained and Moderately Trained Males. Endoscopy 1997, 18, 118–124. [Google Scholar] [CrossRef]
  162. Poole, D.C.; Burnley, M.; Vanhatalo, A.; Rossiter, H.B.; Jones, A.M. Critical Power: An Important Fatigue Threshold in Exercise Physiology. Med. Sci. Sports Exerc. 2016, 48, 2320–2334. [Google Scholar] [CrossRef] [Green Version]
  163. McClave, S.A.; LeBlanc, M.; Hawkins, S.A. Sustainability of Critical Power Determined by a 3-Minute All-Out Test in Elite Cyclists. J. Strength Cond. Res. 2011, 25, 3093–3098. [Google Scholar] [CrossRef]
  164. Whipp, B.J.; Ward, S.A.; Rossiter, H. Pulmonary O2 Uptake during Exercise: Conflating Muscular and Cardiovascular Responses. Med. Sci. Sports Exerc. 2005, 37, 1574–1585. [Google Scholar] [CrossRef]
  165. Boushel, R.; Gnaiger, E.; Calbet, J.A.; Gonzalez-Alonso, J.; Wright-Paradis, C.; Sondergaard, H.; Ara, I.; Helge, J.W.; Saltin, B. Muscle mitochondrial capacity exceeds maximal oxygen delivery in humans. Mitochondrion 2011, 11, 303–307. [Google Scholar] [CrossRef]
  166. Bruusgaard, J.C.; Johansen, I.B.; Egner, I.M.; Rana, Z.A.; Gundersen, K. Myonuclei acquired by overload exercise precede hypertrophy and are not lost on detraining. Proc. Natl. Acad. Sci. USA 2010, 107, 15111–15116. [Google Scholar] [CrossRef] [Green Version]
  167. Egner, I.M.; Bruusgaard, J.C.; Eftestøl, E.; Gundersen, K. A cellular memory mechanism aids overload hypertrophy in muscle long after an episodic exposure to anabolic steroids. J. Physiol. 2013, 591, 6221–6230. [Google Scholar] [CrossRef]
  168. Steele, P.E.; Tang, P.H.; Degrauw, A.J.; Miles, M.V. Clinical Laboratory Monitoring of Coenzyme Q10 Use in Neurologic and Muscular Diseases. Pathol. Patterns Rev. 2004, 121 (Suppl. S1), S113–S120. [Google Scholar] [CrossRef] [PubMed]
  169. Littarru, G.; Battino, M.; Tomasetti, M.; Mordente, A.; Santini, S.; Oradei, A.; Manto, A.; Ghirlanda, G. Metabolic implications of Coenzyme Q10 in red blood cells and plasma lipoproteins. Mol. Asp. Med. 1994, 15, s67–s72. [Google Scholar] [CrossRef]
  170. Shinozawa, S.; Araki, Y.; Oda, T. Stabilizing effects of coenzyme Q10 on potassium ion release, membrane potential and fluidity of rabbit red blood cells. Acta Med. Okayama 1980, 34, 255–261. [Google Scholar] [PubMed]
  171. Wani, M.R.; Shadab, G.G.H.A. Coenzyme Q10 protects isolated human blood cells from TiO2 nanoparticles induced oxidative/antioxidative imbalance, hemolysis, cytotoxicity, DNA damage and mitochondrial impairment. Mol. Biol. Rep. 2021, 48, 3367–3377. [Google Scholar] [CrossRef]
  172. Gao, J.; Xu, Y.; Jia, H.; Zhang, L.; Cao, X.; Zuo, X.; Cai, G.; Chen, X. Associations of coenzyme Q10 with endothelial function in hemodialysis patients. Nephrology 2020, 26, 54–61. [Google Scholar] [CrossRef]
  173. Huo, J.; Xu, Z.; Hosoe, K.; Kubo, H.; Miyahara, H.; Dai, J.; Mori, M.; Sawashita, J.; Higuchi, K. Coenzyme Q10 Prevents Senescence and Dysfunction Caused by Oxidative Stress in Vascular Endothelial Cells. Oxidative Med. Cell. Longev. 2018, 2018, 3181759. [Google Scholar] [CrossRef]
  174. Strong, M.J.; Pattee, G.L.; Strong, G.L.P.M.J. Creatine and coenzyme Q10 in the treatment of ALS. Amyotroph. Lateral Scler. Other Motor Neuron Disord. 2000, 1 (Suppl. S4), S17–S20. [Google Scholar] [CrossRef]
  175. Ahmed, M.; Anderson, S.D.; Schofield, R.S. Coenzyme q10 and creatine in heart failure: Micronutrients, macrobenefit? Clin. Cardiol. 2011, 34, 196–197. [Google Scholar] [CrossRef]
  176. Guescini, M.; Tiano, L.; Genova, M.L.; Polidori, E.; Silvestri, S.; Orlando, P.; Fimognari, C.; Calcabrini, C.; Stocchi, V.; Sestili, P. The Combination of Physical Exercise with Muscle-Directed Antioxidants to Counteract Sarcopenia: A Biomedical Rationale for Pleiotropic Treatment with Creatine and Coenzyme Q10. Oxidative Med. Cell. Longev. 2017, 2017, 7083049. [Google Scholar] [CrossRef] [Green Version]
  177. Mori, T.A.; Burke, V.; Puddey, I.B.; Irish, A.; Cowpland, C.A.; Beilin, L.J.; Dogra, G.K.; Watts, G. The effects of ω3 fatty acids and coenzyme Q10 on blood pressure and heart rate in chronic kidney disease: A randomized controlled trial. J. Hypertens. 2009, 27, 1863–1872. [Google Scholar] [CrossRef]
  178. Kucharská, J.; Poništ, S.; Vančová, O.; Gvozdjáková, A.; Uličná, O.; Slovák, L.; Taghdisiesfejir, M.; Bauerová, K. Treatment With Coenzyme Q10, ω-3-Polyunsaturated Fatty Acids and Their Combination Improved Bioenergetics and Levels of Coenzyme Q9 and Q10 in Skeletal Muscle Mitochondria in Experimental Model of Arthritis. Physiol. Res. 2021, 70, 723–733. [Google Scholar] [CrossRef]
  179. Fouad, G.I. Combination of Omega 3 and Coenzyme Q10 Exerts Neuroprotective Potential Against Hypercholesterolemia-Induced Alzheimer’s-Like Disease in Rats. Neurochem. Res. 2020, 45, 1142–1155. [Google Scholar] [CrossRef]
  180. Moon, H.-J.; Ko, W.-K.; Han, S.W.; Kim, D.-S.; Hwang, Y.-S.; Park, H.-K.; Kwon, I.K. Antioxidants, like coenzyme Q10, selenite, and curcumin, inhibited osteoclast differentiation by suppressing reactive oxygen species generation. Biochem. Biophys. Res. Commun. 2012, 418, 247–253. [Google Scholar] [CrossRef]
  181. Parohan, M.; Sarraf, P.; Javanbakht, M.H.; Foroushani, A.R.; Ranji-Burachaloo, S.; Djalali, M. The synergistic effects of nano-curcumin and coenzyme Q10 supplementation in migraine prophylaxis: A randomized, placebo-controlled, double-blind trial. Nutr. Neurosci. 2019, 24, 317–326. [Google Scholar] [CrossRef]
  182. Devadasu, V.R.; Wadsworth, R.M.; Kumar, M.N.V.R. Protective effects of nanoparticulate coenzyme Q10 and curcumin on inflammatory markers and lipid metabolism in streptozotocin-induced diabetic rats: A possible remedy to diabetic complications. Drug Deliv. Transl. Res. 2011, 1, 448–455. [Google Scholar] [CrossRef]
  183. Burke, L.M.; Peeling, P. Methodologies for Investigating Performance Changes With Supplement Use. Int. J. Sport Nutr. Exerc. Metab. 2018, 28, 159–169. [Google Scholar] [CrossRef] [Green Version]
  184. Grossman, J.; Mackenzie, F.J. The Randomized Controlled Trial: Gold standard, or merely standard? Perspect. Biol. Med. 2005, 48, 516–534. [Google Scholar] [CrossRef]
  185. WHO. WHO Guidelines on Physical Activity and Sedentary Behaviour; World Health Organization: Geneva, Switzerland, 2020. [Google Scholar]
  186. Riebe, D.; Ehrman, J.K.; Liguori, G.; Magal, M. ACSM’s Guidelines for Exercise Testing and Prescription; Kluwer, W., Ed.; American College of Sports Medicine: Filadelfia, PA, USA, 2018. [Google Scholar]
  187. Geifman, N.; Cohen, R.; Rubin, E. Redefining meaningful age groups in the context of disease. AGE 2013, 35, 2357–2366. [Google Scholar] [CrossRef] [Green Version]
  188. Beyer, R.E.; Morales-Corral, P.G.; Ramp, B.J.; Kreitman, K.R.; Falzon, M.J.; Rhee, S.Y.S.; Kuhn, T.W.; Stein, M.; Rosenwasser, M.J.; Cartwright, K.J. Elevation of tissue coenzyme Q (ubiquinone) and cytochrome c concentrations by endurance exercise in the rat. Arch. Biochem. Biophys. 1984, 234, 323–329. [Google Scholar] [CrossRef] [Green Version]
  189. Gohil, K.; Rothfuss, L.; Lang, J.; Packer, L. Effect of exercise training on tissue vitamin E and ubiquinone content. J. Appl. Physiol. 1987, 63, 1638–1641. [Google Scholar] [CrossRef]
Figure 1. Full search strategy. Eligibility criteria of the review: randomised, double-blind, placebo-controlled, parallel design, determining at least plasma CoQ10 levels, and that the human sample were athletes or subjects trained and used to practising high-intensity exercise.
Figure 1. Full search strategy. Eligibility criteria of the review: randomised, double-blind, placebo-controlled, parallel design, determining at least plasma CoQ10 levels, and that the human sample were athletes or subjects trained and used to practising high-intensity exercise.
Nutrients 14 01811 g001
Table 1. Methodological quality of the studies included in the systematic review. Assessment of the methodological quality of studies using the PEDro Scale.
Table 1. Methodological quality of the studies included in the systematic review. Assessment of the methodological quality of studies using the PEDro Scale.
ReferencesSvensson
et al. [68]
Díaz-Castro
et al. [83,84]
Kizaki
et al. [85]
Kon
et al. [86]
Sarmiento
et al. [89]
Suzuki
et al. [90]
Braun
et al. [41]
Ho
et al. [56]
Weston
et al. [72]
Criteria 1 *111111111
Criteria 2011111111
Criteria 3111111111
Criteria 4110011110
Criteria 5111111111
Criteria 6111111111
Criteria 7111111111
Criteria 8111100111
Criteria 9.111111111
Criteria 10111111111
Criteria 11111111111
PEDro Score910999910109
Criteria 1 * = The eligibility criterion is not scored for being related to external validity and therefore does not reflect the quality dimensions assessed by the PEDro Scale.
Table 2. Studies with the inclusion criteria.
Table 2. Studies with the inclusion criteria.
ReferenceMoleculeCoQ10
mg/d
Duration (d: Days)Placebon
Total/CoQ10
Type of SubjectsSexSport/ActivityExercise TestingAge (Years)Impact on Phys./Sport Perf.
Svensson et al. (1999)[68]Ubiquinone12020 dYes17/9Well trained subjectsMale-Graded max and Anaerobic tests28 ± 5No
Díaz-Castro et al. (2020)[83,84]Ubiquinol20014 dYes100/50Well trained subjectsMaleFiremenCircuit edurance exercises39 ± 1-
Kizaki et al. (2015)[85]Ubiquinol60011 dYes32/17Well trained subjectsMaleKendo4 training days20 ± 1No
Kon et al. (2008)[86]Ubiquinol30020 dYes18/10High levelMaleKendoMuscle injury induced exercise20 ± 1Yes
Sarmiento et al. (2016)[89]Ubiquinol20014 dYes100/50Well trained subjectsMaleFiremenCircuit edurance exercises39 ± 9-
Suzuki et al. (2021)[90]Ubiquinol30012 dYes16/8Well trained subjectsMaleDistance runners25 & 40 K races20 ± 2Yes
Braun et al. (1991)[41]Ubiquinone10056 dYes12/6High levelMaleCyclistsGraded max test22 ± 2No
Ho et al. (2020)[56]Ubiquinone30084 dYes31/15Moderately trained subjectsMale & FemaleSoccer & TaekwondoNone20 ± 1-
Weston et al. (1997)[72]Ubiquinone≃7028 dYes18/6High levelMaleCyclists and TriathletesGraded max.25 ± 3No
Table 3. Studies with the inclusion criteria.
Table 3. Studies with the inclusion criteria.
ReferenceChange CoQ10 TotalMeasured Parameters and Effects of CoQ10
TissuePlasma
Pre ➝ Post
(μg/mL)
Plasma
Pre ➝ Post
(μmol/mol Chol)
Muscle
Pre ➝ Post
(nmol/g Protein)
Inflammatory PatternAntioxidant
Pattern
Physical PerformanceSport PerformanceMuscle InjuryOther
Svensson et al. (1999)Plasma ↑ Muscle ↔ 0.74 ➝ 1.2340.8 ➝ 44.2 mgl/kgHX, MDA, UA ↔
Díaz-Castro et al. (2020)Plasma ↑1.00 ➝ 5.22VEGF, NO, EGF, IL-1ra, IL-10 ↑, and IL-1, IL-8, MCP-1 ↓PTH, OC, OPG, phosphatase al., leptin, insulin, noradrenaline and PGC-1α ↑,
Kizaki et al. (2015)Plasma↑0.7 ➝ 10.8CK, Mb ↔,
Kon et al. (2008)Plasma↑≃0.8 ➝ ≃3.8LPO ↓CK, Mb ↓
Sarmiento et al. (2016)Plasma ↑0.9 ➝ 4.5NO ↓ hydroperoxydes Isoprostanes, oxidized LDL, TAC ↑
Suzuki et al. (2021)Plasma ↑0.7 ➝ 5.6Perception of fatigue ↓CK, ALT, LDH ↓
Braun et al. (1991)Plasma ↑≃0.8 ➝ 1.5MDA ↓ (NS)Total work load, VO2peak, HR ↔ CoQ10 postexercise ↑
Ho et al. (2020)Plasma ↑0.57 ➝ 1.14130 ➝ 270MDA ↓, TAC ↔ Improve glycemic control
Weston et al. (1997)Plasma ↑0.9 ➝ 2.0Oxygen uptake at 6 min ↑, VO2max, max power, Anaerobic threshold, and HRTime ↔
CoQ10/Chol: CoQ10 in relation to cholesterol level; pre/post: previous and post supplementation; Ns: No specified; Phys. Physical; Perf: Performance; ↔ similar response to placebo or previous supplementation, ↑ Higher response to the presupplementation phase, ↓ Lower response to the presupplementation phase. HX hypoxanthine, MDA malondyaldehide, UA uric acid, PTH parathormone, OC osteocalcin, OPG osteoprotegerin, PGC-1α peroxisome proliferator activated receptor-γ coactivator-1α VEGF vascular endothelial growth factor, NO nitric oxide, EGF epidermal growth factor, CK creatinkinase, Mb myoglobin, ALT alanine transaminase, LDH lactate dehydrogenase, TAC Total antioxidant capacity, MCP-1 monocyte chemotactic protein, LPO lipid peroxidation.
Table 4. Studies without the inclusion criteria.
Table 4. Studies without the inclusion criteria.
ReferenceMoleculeCoQ10
mg/d
Duration (d: Days)Placebon
Total/CoQ10
Type of SubjectsSexSport/ActivityExercise TestingAge (Years)Impact on Phys./Sport Perf.
Alf et al. (2013)[81]Ubiquinol30042 dYes100/50High levelMale & FemaleOlympic AthletesCycling submaximal test19 ± 3Yes/Yes
Bloomer et al. (2012)[82]Ubiquinol30028 dYes15/15Well trained subjectsMale & FemaleNsGraded and Anaerobic max. Tests43 ± 10No
Kunching et al. (in press)[87]Ubiquinol20042 dYes29/15Moderately trained subjectsMaleDiverseIndirect max. Test, 1RM and flexibility24 ± 2Yes
Orlando et al. (2018)[88]Ubiquinol20028 dYes21/21Moderately trained subjectsMaleRugby40 min 85%max. Treadmill26 ± 5No
Pham et al. (2020)[80]Ubiquinol20042 dNo22/22Physically active subjectsMale-None51 ± 1-
Broome et al. (2021)[93]Mitoquinone2028 dYes19/19Well trained subjectsMaleCyclists8 km race44 ± 4Yes
Pham et al. (2020)[80]Mitoquinone2042 dNo22/22Physically active subjectsMale-None51 ± 1-
Shill et al. (2021)[91]Mitoquinone1021 dYes20/10Physically active subjectsMale-Graded max test22 ± 1No
Williamsom et al. (2020)[92]Mitoquinone2021 dYes24/12Physically active subjectsMale-Anaerobic repeated tests25 ± 4Yes
Amadio et al. (1991)[38]Ubiquinone10040 dControl10/5Well trained subjectsMaleBasketballSubmaximal test19 ± 5Yes
Armanfar et al. (2015)[39]Ubiquinone300–400 14 dYes18/9Well trained subjectsMaleMiddle distance runners3000 m race20 ± 3No
Bonetti et al. (2000)[40]Ubiquinone10056 dYes28/14Moderately trained subjects and NAMaleCyclistsGraded max test41 ± 6Yes
Cerioli (1991)[42]Ubiquinone10030 dNs12Non activeMale-Graded max test26Yes
Cinquegrana et al. (1987)[43]Ubiquinone6035 dYes14/14Non activeMale-Graded max test48 ± 3
Ciocoi-Pop et al. (Note I & II) (2009)[44]Ubiquinone3021 dYes10/5Well trained subjectsMaleSoccerGraded max and Anaerobic tests19 ± 0Yes
Cooke et al. (2008)[45]Ubiquinone20014 dYes31/21Moderately trained subjects and untrainedMale & Female-Grade max, Isokinetic, and Anaerobic tests26 ± 8No
Díaz-Castro et al. (2012)[46]Ubiquinone30 × 2 days, 120 day test 3 dYes20/10Well trained subjectsMale-50 km running41 ± 3Yes
Table 5. Studies without the inclusion criteria.
Table 5. Studies without the inclusion criteria.
ReferenceChange CoQ10 TotalMeasured Parameters and Effects of CoQ10
TissuePlasma
Pre ➝ Post
(μg/mL)
Plasma
Pre ➝ Post
(μmol/mol Chol)
Muscle
Pre ➝ Post
(nmol/g Protein)
Inflammatory PatternAntioxidant
Pattern
Physical PerformanceSport PerformanceMuscle InjuryOther
Alf et al. (2013)- Power output ↑
Bloomer et al. (2012)Plasma ↑0.98 ➝ 2.330.48 ➝ 1.13 MDA, hydrogen peroxide,Lactate ↔ perceived vigour ↔
Kunching et al. (in press)- VO2max ml/kg ↑,Max strength ↔ Sistolic pressure ↓
Orlando et al. (2018)Plasma ↔
CoQ10/Chol ↑
≃180 ➝ >500 ROS ↓Time to exhaustion, speed ↔ CK, DNA damage ↔
Pham et al. (2020)- H2O2 mit ↓, Isoprost ↔
Broome et al. (2021)- ROS, Isoprost ↓Power output ↑Faster time trial.
Pham et al. (2020)- H2O2 mit ↓, TAC ↑, Isoprostanes ↔
Shill et al. (2021)- CDseries, VEGFR2+ and peripheral blood mononuclear cells ↔ VO2max ↔ Muscle mitochondrial capacity ↔
Williamsom et al. (2020)- DNA damage ↓
Amadio et al. (1991)Plasma ↑0.9 ➝ 1.6 VO2max ↑ Cardiac parameters
Armanfar et al. (2015)- TNFa, CRP, IL6 ↓ CK ↔
Bonetti et al. (2000)Plasma ↑0.8 ➝ 2.2 hypoxanthine, xanthine and inosine ↔ Max work load ↑ VO2peak, anaerobic threshold and lactate ↔,
Cerioli (1991)- Aerobic Capacity↑ CK ↔ FFA ↓,Fat metabolism ↑
Cinquegrana et al. (1987)-
Ciocoi-Pop et al. (Note I&II) (2009)- MDA ↑, HD ↑ in salivaVO2max ↑, Anaerobic power ↔
Cooke et al. (2008)Plasma ↑
Muscle ↔
≃0.6 ➝ ≃2.5 ≃1.2 ➝ ≃1.4
μg/mg
MDA ↑, SOD ↓VO2max, anaerobic capacity, Anaerobic power ↔
Díaz-Castro et al. (2012)- IL-6 ↔, 8-OH-dG, TNF-α ↓CAT ↑, TAS ↑, GPx ↔, hydroperoxide ↓, isoprostane ↓,
CoQ10/Chol: CoQ10 in relation to cholesterol level; pre/post: previous and post supplementation; Ns: No specified; Phys. Physical; Perf: Performance; ↔ similar response to placebo or previous supplementation, ↑ Higher response to the presupplementation phase, ↓ Lower response to the presupplementation phase. GPx Glutathione peroxidase, MDA malondyaldehide, ROS reactive oxygen species, CK creatinquinase, VO2 oxygen consumption, HD hydrogen donors, SOD superoxide dismutase, CAT catalase, TAS Plasma total antioxidant status, 8-OH-dG 8-Hydroxy-20-deoxyguanosine, TNF-α Tumor necrosis factor a, FFA free fatty acids, CRP C-reactive protein, IL6 interleukin 6.
Table 6. Studies without the inclusion criteria.
Table 6. Studies without the inclusion criteria.
ReferenceMoleculeCoQ10
mg/d
Duration (d: Days)Placebon
Total/CoQ10
Type of SubjectsSexSport/ActivityExercise TestingAge (Years)Impact on Phys./Sport Perf.
Drobnic et al. (2020) [47]Ubiquinone10030 dControl20/12Well trained subjectsMaleMarathon runnersTreadmill Hot & Humid environment55 ± 4Yes
Emami et al. (2018)[48]Ubiquinone30014 dYes36/9Well trained subjectsMaleSwimmwersSwimming training18 ± 1
Fiorella et al. (1991)[49]Ubiquinone10040 dControl22/11Well trained subjectsMaleAthletesGraded max test + Incremental running test until exhaustion29 ± 6Yes
García-Verazaluce et al. (2015)[50]Ubiquinone +120 + Phlebodium d.28 dYes30/10Well trained subjectsMaleVolleyballNone25 ± 2Ns
Geiss et al. (2004)[51]Ubiquinone18028 dYes10/10Well trained subjectsMale?EnduranceSubmaximal fatigue testNsYes
Gökbel et al. (2010)[52]Ubiquinone10056 dYes15/15Non activeMaleNsAnaerobic test(Repeated Wingate test)20 ± 1No
Gökbel et al. (2016)[53]Ubiquinone20098 dYes23/23Patients (hemodyalisis) MaleNs6 Mins walking test47 ± 12No
Guerra et al. (1987)[54]Ubiquinone6035 dNs Moderately trained subjectsMaleCyclingGraded max test. Race 9 km.NsYes/Yes
Gül et al. (2011)[55]Ubiquinone10056 dYes15Non activeMale-Anaerobic repeated tests20 ± 1Yes
Kaikkonen et al. (1998)[57]Ubiquinone
+
90 + Vit E21 dYes37/18Moderately trained subjectsMaleMarathonMarathon40 ± 7No
Laaksonen et al. (1995)[58]Ubiquinone
+
120 +
Omega 3
42 dYes11/11Well trained subjectsMale
Male
Marathon & triathletesGraded max. test28
(22–38)
No
8/864
(60–74)
No
Leelarungrayub (2010)[59]Ubiquinone30012 dNo16/16Moderately trained subjectsMale & FemaleSwimmimgTreadmill time to exhaustion & Swimmimg 100–800 m15 ± 1Yes 100 m,
No 800 m
Malm et al. (1996)[60]Ubiquinone12020 dYes15/9Moderately trained subjectsMale-Anaerobic tests20–34No
Malm et al. (1997)[61]Ubiquinone12022 dYes18/9Moderately trained subjectsMale-Graded max and Anaerobic tests25 ± 3No
Mizuno et al. (2008)[62]Ubiquinone100 & 30056 dYes17/17Physically active subjectsMale & Female-Anaerobic repeated tests under fatigue38 ± 10Yes
Nielsen et al. (1999)[63]Ubiquinone
+
100 + Vit E & Vit C42 dYes7/7Well trained subjectsMaleTriathletesGraded max. & Local fatigue (31P-NMRS)22–32No
+: indicates that the administration of Ubiquinone is supplemented by one or more other substances (indicated in the next column).
Table 7. Studies without the inclusion criteria.
Table 7. Studies without the inclusion criteria.
ReferenceChange CoQ10 TotalMeasured Parameters and Effects of CoQ10
TissuePlasma
Pre ➝ Post
(μg/mL)
Plasma
Pre ➝ Post
(μmol/mol Chol)
Muscle
Pre ➝ Post
(nmol/g Protein)
Inflammatory PatternAntioxidant
Pattern
Physical PerformanceSport PerformanceMuscle InjuryOther
Drobnic et al. (2020)Plasma ↑ Muscle ↑1.11 ➝ 2.34212 ➝ 476245 ➝ 299nmol/g proteinIL-6, IL-8, IL-10, MCP-1, TNFa ↓MDA ↓, TAC ↑ after exerciseLactate and fatigue perception ↑,
Emami et al. (2018)Plasma ↑≃0.8 ➝ ≃2.5 TAC ↑, LPO ↓, LDH, CK-MB, Mb, Troponin I ↓
Fiorella et al. (1991)Plasma ↑ Thrombocites↑0.6 ➝ 1.4 37.5➝61.8 LA, UA, Ammonia ↔ Running distance and time to exhaustion ↑,CK, LDH ↓,
García-Verazaluce et al. (2015)- IL6 ↓ Costisol ↓
Geiss et al. (2004)Plasma ↑0.6 ➝ 1.7 Power output ↑
Gökbel et al. (2010)- Peak Power ↔, Mean Power ↑, Fatigue Index ↔
Gökbel et al. (2016)Plasma ↑1.3 ➝ 3.0 MDA ↓, GPX↓, SOD ↔, after exercise (NS)
Guerra et al. (1987)Plasma ↑Ns VO2max ↑Possible better race time
Gül et al. (2011)- MDA ↓, NO, XO, SOD, GPx ↔, UA ↑
Kaikkonen et al. (1998)Plasma ↑1.96 ➝ 2.03 GSH, UA, LDLox, TRAP ↔
Laaksonen et al. (1995)Plasma ↑
Muscle ↑
0.9 ➝ 2.0 118 ➝ 128
nmol/g protein
MDA ↔ VO2max ↔, Time to exhaustion
Plasma ↑
Muscle ↔
1.31 ➝ 3.592 ➝ 78
nmol/g protein
Leelarungrayub et al. (2010)Plasma ↑1.1 ➝ 2.3 MDA, NO ↓,
TAC, SOD ↔, GSH ↑
Increase time to fatigueNo better 800 m swimming time
Malm et al. (1996)- CK ↑
Malm et al. (1997)- ROS ↑VO2max ↔, Max power ↔
Mizuno et al. (2008)Plasma ↑100 mg. 0.5 ➝ 2.0
300 mg. 0.5 ➝ 3.3
Perception of fatigue ↓, Perception of recovery, Max velocity ↑
Nielsen et al. (1999)Plasma ↑0.9 ➝ 1.8 VO2max ↔ Muscle metabolism ↔
CoQ10/Chol: CoQ10 in relation to cholesterol level; pre/post: previous and post supplementation; ➝: Concentration change; Ns: No specified, Phys. Physical; Perf: Performance; ↔ similar response to placebo or previous supplementation, ↑ Higher response to the presupplementation phase, ↓ Lower response to the presupplementation phase, NS: Non statistical significance. MDA malondyaldehide, TAC total antioxidant capacity, LPO lipid peroxidation, LDH lactate dehydrogenase, CK-MB miocardic creatinquinase, Mb myoglobin, LA lactic acid, UA uric acid, CK creatinquinase, SOD superoxide dismutase, NO nitric oxide.
Table 8. Studies without the inclusion criteria.
Table 8. Studies without the inclusion criteria.
ReferenceMoleculeCoQ10
mg/d
Duration (d: Days)Placebon
Total/CoQ10
Type of SubjectsSexSport/ActivityExercise TestingAge (Years)Impact on Phys./Sport Perf
Okudan et al. (2017)[64]Ubiquinone20028 dYes21/11Non activeMale-Exccentric ex.23 ± 0No
Östman et al. (2012)[65]Ubiquinone9056 dYes23/11Moderately trained subjectsMale-Various exercise capacity tests28 ± 9No
Porter et al. (1995)[66]Ubiquinone15056 dYes13/6Physically active subjectsMaleSome with hypertensionGraded Max and Forearm Handgrip tests45 ± 2Yes
Snider et al. (1992)[67]Ubiquinone +100 + Vit E, C inosine, citochrome C28 dYes11/11High levelMaleTriathletesGraded max. test25 ± 1No
Tauler et al. (2008) Ferrer et al. (2009)[69,70]Ubiquinone +100+ Multivitamin90 dYes19/8High levelMaleSoccerCompetition Match20 ± 0Yes
Vanfraechem et al. (1981)[71]Ubiquinone6056 dYes6Non activeMaleNoneGraded max. 4w–8w22 ± 2Yes
Wyss et al. (1990)[73]Ubiquinone10030 dYes18/18Physically active subjectsMaleRunningGraded max.25 ± 4Yes
Yamabe et al. (1991)[74]Ubiquinone906 monthsNo9/9Non active with inabilities to do exerciseMale-Graded max.51 ± 5Yes
Ylikoski et al. (1997)[75]Ubiquinone9042 dYes18/18High levelMalecross-country skiersGraded max.NsYes
Zeppilli et al. (1991)[76]Ubiquinone10030 dYes9/9High level MaleVolleyball Graded max test17–2Yes
10Non active -23–29Yes
8PatientsMit. Disease23–29Yes
Zheng et al. (2008)[77]Ubiquinone301dYes11/11Non activeMale-Rest & Low intensity exercise (30% HRmax)26 ± 1Yes
Zhou et al. (2005)[78]Ubiquinone +150+ Vit E14 dYes6/6Physically active subjectsMale-Submaximal exercise and Graded max tests30 ± 7No
Zuliani et al. (1989)[79]Ubiquinone10028 dNo12Non activeMale-60’ ciloergometry26No
Sanchez-Cuesta et al. (2020)[99].-2 Sport seasons-24 & 25High levelMaleSoccerWeekly competition26 ± 4Yes
+: indicates that the administration of Ubiquinone is supplemented by one or more other substances (indicated in the next column).
Table 9. Studies without the inclusion criteria.
Table 9. Studies without the inclusion criteria.
ReferenceChange CoQ10 TotalMeasured Parameters and Effects of CoQ10
TissuePlasma
Pre ➝ Post
(μg/mL)
Plasma
Pre ➝ Post
(μmol/mol Chol)
Muscle
Pre ➝ Post
(nmol/g Protein)
Inflammatory PatternAntioxidant
Pattern
Physical PerformanceSport PerformanceMuscle InjuryOther
Okudan et al. (2017)Plasma ↑1.0 ➝ 1.7 MDA, SOD ↔ CK, Myogb. ↔
Östman et al. (2012)- MDA, UA, hypoxanthine ↔ Exercise capacity, VO2max, Max power, Lactate ↔ CK, UA ↔
Porter et al. (1995)Plasma ↑0.7 ➝ 1.0 VO2max ↔ LA ↓(NS) Vigor perception ↑
Snider et al. (1992)- Time to exhaustion,
RPE ↔
Tauler et al. (2008) Ferrer et al. (2009)Plasma ↑3.0 ➝ 3.6 SOD ↑, MDA ↓ ↑ Time spend in active working zone (Z3-Z5)
Vanfraechem et al. (1981)- Maximal load and O2 consumption 170 w and max. Cardiaovascular and cardiorespiratory parameters ↑
Wyss et al. (1990)Plasma ↑Ns VO2max ↑, Max work ↑,
Yamabe et al. (1991)- VO2max, Max power and Anaerobic threshold ↑
Ylikoski et al. (1997)Plasma ↑0.8 ➝ 2.8 VO2max, max power, Anaerobic threshold, and HRTime↑
Zeppilli et al. (1991)Plasma ↑0.6 ➝ 1.3 Vb 0.7 ➝ 1.0 NA VO2max, max power ↑
Zheng et al. (2008)- VCO2/VO2 ↓, HR ↔ ↑ Power HR variability
Zhou et al. (2005)Plasma↑, Muscle ↔ 0.8 ➝ 2.6 207 ➝ 220
(nmol/g protein)
VO2max ↔, HR ↔, RPE ↔, Anaerobic threshold ↔
Zuliani et al. (1989)Plasma ↑0.5 ➝ 1.3 Glucose, Insulin, LA ↔ CK ↔,Glycerol ↔, FFA ↓
Sanchez-Cuesta et al. (2020)Plasma ↑Preseason 0.6 Middle season 0.9 The highest values have better competition parameters CK ↓Better Testosterone/cortisol pattern
CoQ10/Chol: CoQ10 in relation to cholesterol level; pre/post: previous and post supplementation; Ns: No specified; Phys. Physical; Perf: Performance; ↔ similar response to placebo or previous supplementation, ↑ Higher response to the presupplementation phase, ↓ Lower response to the presupplementation phase, NS: Non statistical significance. The study of Sanchez-Cuesta as it is explained in the text, is the follow up of the CQ10 plasma level throughout 2 competition seasons. Vb volleyball, NA No physical activity, HR heart rate, RPE rating perceived perception.
Table 10. Results of the Ubiquinone and Ubiquinol supplementation on different sport and exercise variables depending on the physical condition of the subjects evaluated.
Table 10. Results of the Ubiquinone and Ubiquinol supplementation on different sport and exercise variables depending on the physical condition of the subjects evaluated.
Physical Condition CategoriesNumber of StudiesCoQ10
Effect
TotalSport
Performance
Exercise
Performance
Oxidative
Pattern
Muscle
INJURY
Inflammatory
Pattern
Other
High level/Well trained athletes28Positive3931210464
No effect12 61311
Moderate trained/Physically active20Positive11 451 2
No effect181953
Negative3 21
Non active
subjects
9Positive11 61 4
No effect4 2 2
Patients1Positive2 11
TOTAL58number tests100 *3402514711
Positive6322317 5610
No effect341176811
Negative3 21
* In the Zeppilli research [77] there are three populations (<30 a) that are studied with the same methodology. That is the reason why there are 2 more tests than in Table 11.
Table 11. Results of the Ubiquinone and Ubiquinol supplementation on different sport and exercise variables depending on the age categories of the subjects evaluated.
Table 11. Results of the Ubiquinone and Ubiquinol supplementation on different sport and exercise variables depending on the age categories of the subjects evaluated.
Age
Categories
Number of StudiesCoQ10
Effect
TotalSport
Performance
Exercise
Performance
Oxidative
Pattern
Muscle
Injury
Inflammatory PatternOther
<3037Positive382129528
No effect261133711
Negative3 21
31–5011Positive15136 32
No effect6 321
>507Positive5 22 1
No effect2 1
No specified3Positive3 3
Table 12. Recommended methodological guidelines for a future study on human physical performance of CoQ10.
Table 12. Recommended methodological guidelines for a future study on human physical performance of CoQ10.
Physical activity:Four types: No physically active or light activity (<3 d/week), Moderately active (3–5 d/week), Very active (5–7 d/week), High level athletes.
Age (years):Four types: ≤33, 34–48, 49–64, ≥65
Diet:Homogenise the sample according to the type of diet
Dosage4.0–4.5 mg/kg/d of Ubiquinol or Ubiquinone with Phytosome or Ubiquinone with vehicle to increase bioavailability, or explaining perfectly the diet related to the administration.
PlaceboYes, double blinded.
Type of studyParallel, never crossover to avoid training and supplementation effect
Tissue concentration (plasma)Mandatory. Always after 24 h after last doses of placebo or study substance.
Recommended before and after exercise stress test.
Determined always as “μmol/mol Chol” and added as “μg/mL”.
Tissue concentration (muscle)Very recommended.
Indispensable to know mitochondrial density.
Determined as “nmol/g protein”
Treatment period>1 week.
Effort test before and after supplementationCharacterization of the maximal oxygen consumption of every subject by a graded maximal exercise test.
Evaluation of the metabolic efficiency under, at and upper the anaerobic threshold.
Long duration exercise at a submaximal level to evaluate oxidative and inflammatory pattern.
Evaluation of subjective fatigue perception
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Drobnic, F.; Lizarraga, M.A.; Caballero-García, A.; Cordova, A. Coenzyme Q10 Supplementation and Its Impact on Exercise and Sport Performance in Humans: A Recovery or a Performance-Enhancing Molecule? Nutrients 2022, 14, 1811. https://doi.org/10.3390/nu14091811

AMA Style

Drobnic F, Lizarraga MA, Caballero-García A, Cordova A. Coenzyme Q10 Supplementation and Its Impact on Exercise and Sport Performance in Humans: A Recovery or a Performance-Enhancing Molecule? Nutrients. 2022; 14(9):1811. https://doi.org/10.3390/nu14091811

Chicago/Turabian Style

Drobnic, Franchek, Mª Antonia Lizarraga, Alberto Caballero-García, and Alfredo Cordova. 2022. "Coenzyme Q10 Supplementation and Its Impact on Exercise and Sport Performance in Humans: A Recovery or a Performance-Enhancing Molecule?" Nutrients 14, no. 9: 1811. https://doi.org/10.3390/nu14091811

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop