Summary
Coenzyme Q10 (CoQ10), a lipophilic substituted benzoquinone, is present in animal and plant cells. It is endogenously synthetized in every cell and involved in a variety of cellular processes. CoQ10 is an obligatory component of the respiratory chain in inner mitochondrial membrane. In addition, the presence of CoQ10 in all cellular membranes and in blood. It is the only endogenous lipid antioxidant. Moreover, it is an essential factor for uncoupling protein and controls the permeability transition pore in mitochondria. It also participates in extramitochondrial electron transport and controls membrane physicochemical properties. CoQ10 effects on gene expression might affect the overall metabolism. Primary changes in the energetic and antioxidant functions can explain its remedial effects. CoQ10 supplementation is safe and well-tolerated, even at high doses. CoQ10 does not cause any serious adverse effects in humans or experimental animals. New preparations of CoQ10 that are less hydrophobic and structural derivatives, like idebenone and MitoQ, are being developed to increase absorption and tissue distribution. The review aims to summarize clinical and experimental effects of CoQ10 supplementations in some neurological diseases such as migraine, Parkinson’s disease, Huntington’s disease, Alzheimer’s disease, amyotrophic lateral sclerosis, Friedreich’s ataxia or multiple sclerosis. Cardiovascular hypertension was included because of its central mechanisms controlling blood pressure in the brainstem rostral ventrolateral medulla and hypothalamic paraventricular nucleus. In conclusion, it seems reasonable to recommend CoQ10 as adjunct to conventional therapy in some cases. However, sometimes CoQ10 supplementations are more efficient in animal models of diseases than in human patients (e.g. Parkinson’s disease) or rather vague (e.g. Friedreich’s ataxia or amyotrophic lateral sclerosis).
Keywords: Idebenone, MitoQ, Migraine, Parkinson’s disease, Alzheimer’s disease, Multiple sclerosis, Hypertension
Introduction
Coenzyme Q (CoQ), a highly hydrophobic molecule, is known from 1957 when it was isolated from beef heart mitochondria by Professor Frederick Crane on Madison University (Crane et al. 1957, Crane 1989). CoQ is composed of a benzoquinone ring and a polyisoprenoid lipid tail containing varying chain length depending on the species (Fig. 1). Human isoform contains ten isoprene units (CoQ10) but rodents have mainly nine units (CoQ9) besides some amount of CoQ10. Briefly, the benzoquinone ring is derived from the essential amino acid phenylalanine, which is converted into tyrosine and then 4-hydroxybenzoate. The polyisoprenoid lipid tail subunits are generated from acetyl-CoA (and through joint intermediate of cholesterol – farnesyl-pyrophosphate) by the mevalonate pathway. The last phase of a CoQ biosynthesis is the condensation of the benzoquinone ring and the polyisoprenoid tail. A lot of knowledge on CoQ biosynthesis originated from experiments on simple organisms such as the budding yeast Saccharomyces cerevisia, fission yeast Schizosaccharomyces plombe or coliform bacterium Escherichia coli etc. (Awad et al. 2018, Wang and Hekimi 2013). Stefely and Pagliarini (2017) summarized the recent progress and defined still existing knowledge gaps in CoQ biosynthesis.
Fig. 1.
Chemical structure composed from benzoquinone head and hydrophobic polyisoprenoid tail of fully reduced (ubiquinol, bottom) and fully oxidized (ubiquinone, top) forms of human derivative of coenzyme Q10 (CoQ10)
CoQ is present in most aerobic organisms, all animal and plant organs (Battino et al. 1990, Elmberger et al. 1987, Ramasarma 1985). It is endogenously produced in every cell and its intracellular synthesis is its major source, although a small proportion is acquired through the diet. Meat, fish, nuts, and some oils are the richest nutritional sources of CoQ, while much lower levels can be found in most dairy products, vegetables, fruits, and cereals (Cabrini et al. 2001, Kamei et al. 1986, Kettawan et al. 2007, Pravst et al. 2010, Pyo 2010). Interestingly, the daily intake in different countries is very similar and represents around 3–5 mg (Kubo et al. 2008, Mattila and Kumpulainen 2001, Weber et al. 1997a,b).
CoQ concentrations are modified in humans and rats during lifespan (Beyer et al. 1985, Kalén et al. 1989, Zhang et al. 1996). Although the results of the various studies are not in total agreement, generally, in the early phase of life, CoQ concentrations increase, whereas during aging they decrease. Wada et al. (2007) demonstrated that not only the amount of CoQ but also its redox status is essential to balance the oxidative stress associated with the aging process. CoQ concentrations also decrease in mice and pigs with the age. Similarly, an oxidized form of CoQ10 increases indicating higher oxidative stress or a decreased anti-oxidative capacity of aged animals (Onur et al. 2014). Battino et al. (1995) described in rats that CoQ9 and CoQ10 contents in different mitochondrial fractions (synaptic and non-synaptic) are slowly decreasing in three brain areas (cortex, striatum and hippocampus), reaching their minimum in age of 18 months, then increased in the older rats. Nevertheless, the CoQ9/CoQ10 ratio remained constant during aging.
Apart from the earlier study by Kalén et al. (1989) there are few recent human trials. A large cohort of 860 European adults aged 18–82 years shows a decrease of CoQ10 blood concentrations with a shift in redox status in favour of the oxidized fraction in old people of both sexes (Nicklowitz et al. 2016). The recent study on Japanese centenarians (25 males and 74 females) compared to 76-year-old controls confirms decreased serum total levels of CoQ10 significantly shifted to the oxidized form (Nagase et al. 2018). Thus, exogenous CoQ10 supplementation may show benefits as an antiaging agent during aging process.
Subjects are not generally dependent on exogenous supplies of CoQ10. However, apart ageing CoQ10 concentrations are also affected during different pathological disorders. Under these circumstances, dietary supplementation may be needed and would fulfill important functions by counteracting CoQ10 depletion. Most of the clinical work with CoQ10 is focused on the wide spectrum of heart diseases such as ischemic heart disease, congestive heart failure or different cardiomyopathies (Rauchová and Vokurková 2009).
In this short review I have attempted to show the changes of CoQ10 content in different diseases and possible brain effects of the treatment with CoQ10 (and its derivatives) supplementation. This review is focused on CoQ10 applications in CoQ10 deficiency, very frequent type of headache – migraine – and other nervous diseases (Parkinson’s disease, Alzheimer’s diseases, Friedreich’s ataxia or Huntington disease, amyloid lateral sclerosis, sclerosis multiplex) and improvement in noice-induced hearing loss or in cardiovascular hypertension control.
Multiple functions of coenzyme Q
CoQ, also known as ubiquinone, acts as a mobile electron and proton transporter from complex I (NADH: ubiquinone reductase) and complex II (succinate: ubiquinone reductase) to complex III (ubiquinone cytochrome c oxidase) in the inner mitochondrial membrane (Trumpower 1981). The subcellular distribution shows that the inner mitochondrial membrane has the largest portion of CoQ. In comparison with other respiratory carriers the content of CoQ exceeds other redox components by about tenfold (Capaldi 1982). Moreover, CoQ accepts electrons from other dehydrogenases, which are present in lower amounts, and seems to be rate-limiting in the integrated electron transfer (Genova and Lenaz 2011, 2014). Among them localized on the outer surface of the inner membrane there is mitochondrial FAD-linked glycerol-3-phosphate dehydrogenase – the simplest branch of the respiratory chain and a part of glycerol-3-phosphate shuttle (Rauchová et al. 1992, Rauchová et al. 1997). CoQ is also an obligatory cofactor for FMN-linked dihydroorotate dehydrogenase – a key enzyme of de novo pyrimidine biosynthesis, which is loosely associated with the outer surface of the inner membrane (Evans and Guy 2004, Reis et al. 2017). On the matrix side of inner membrane there is electron transport flavoprotein dehydrogenase forming a short pathway that transfer electron from 11 different mitochondrial flavoprotein dehydrogenases to the quinone pool – an essential enzyme involved in the fatty acid β-oxidation and branched-chain amino acid oxidation pathways (Frerman 1988, Watmough and Frerman 2010). Further there are FAD-dependent proline dehydrogenase (an enzyme required for proline and arginine metabolism) and sulphide-quinone oxidoreductase (Blake et al. 1976, Quinzii et al. 2017). Fig. 2 shows a schematic illustration of the CoQ integration in the mammalian mitochondrial respiratory chain.
Fig. 2.
A schematic illustration of CoQ10 intergration in the mammalian mitochondrial respiratory chain. Electrons (e−) accept oxidized form of coenzyme Q10 (CoQ10) from complex I (C I) and complex II (C II), mitochondrial glycerol-3-phosphate dehydro-genase (GPDH), dihydroorotate dehydro-genase (DHODH), electron transport flavoprotein dehydro-genase (ETFDH), proline dehydrogenase (ProDH) and sulfide dehydro-genase (SulDH) for delivery to complex III (C III) and finally to complex IV (C IV). Glycerol-3-phosphat (GP), dihydroxyaceton phosphate (DHAP), dihydroorotate (DHO), orotate (ORO), cytochrome c (Cyt c)
Beyond fundamental role of an electron carrier associated with cellular energy production in the respiratory chain, CoQ functions as a principal cofactor in the activation of protein uncoupling (Echtay et al. 2000) and it controls permeability of transition pores (Fontaine et al. 1998, Walter et al. 2002). However, CoQ occurs in other cellular membranes such as Golgi apparatus, lysosomes or plasma membrane having a role in functioning of oxido-reductase systems. Gille and Nohl (2000) described the lysosomal membrane redox chain, which provides the low pH inside the organel for the best functioning hydrolytic enzymes necessary for the removal of useless proteins. Moreover, CoQ can also change membrane stability or fluidity what could be important for physiological functions of receptors, carriers and membrane-bound enzymes (Agmo Hernández et al. 2015, Fato et al. 1984). The bioinformatic analyses show that CoQ affects the expression of many genes involved in different cellular pathways (Schmelzer et al. 2007).
In addition, CoQ turns between fully oxidized form, quinone, – through a formation of its semiquinone – and fully reduced form (Fig. 1). The reduced form, ubiquinol, functions as a potent antioxidant and free radical scavenger by either directly scavenging free radicals or recycling and regenerating other antioxidants such as tocopherol (vitamin E) and ascorbic acid (vitamin C). So, by this way it protects membrane lipids, proteins and mitochondrial DNA against oxidative damage. CoQ represents the only lipid soluble antioxidant synthesized by own cells. On the other hand, oxido-reduction character of CoQ has also the important prooxidant role for the formation of superoxide anion (O2−) and hydrogen peroxide (H2O2) in cell signaling systems (Linnane et al. 2007). Furthermore, CoQ acts as a regulator of cell growth and differentiation and a blocker of apoptosis (Kagan et al. 1999, Li et al. 2019) and has anti-inflammatory effects (Schmelzer et al. 2007, Schmelzer et al. 2008). Systematic reviews evaluated the effects of CoQ10 supplementation on various inflammatory markers including C-reactive protein, interleukin-6 or tumor necrosis factor-α (Fan et al. 2017, Zhai et al. 2017).
The functional diversity reflects a suitability of CoQ for applications in clinical studies as an alternative medication or dietary supplement for several diseases, which also includes neurological disorders.
Safety and bioavailability of coenzyme Q10
As a natural component, CoQ10 is well tolerated. However, its therapeutic applications are greatly limited because CoQ10 is almost insoluble in water (due to its high hydrophobicity) and has relatively large molecular mass (863.34 g/mol). In rats, only about 2–4 % of orally administered CoQ10 could be absorbed (Zhang et al. 1995). Divided dosages (2 ×100 mg) produce a larger increase in plasma levels than a single peroral 200 mg dose (Singh et al. 2005). Hence, large daily doses are recommended to be divided into several doses to maximize the CoQ10 absorption. To improve its solubility and bioavailability, several advancements have been made and a variety of preparations has been developed such as compressed tablets, chewable tablets, powder-filled capsules or soft gels containing a suspension in oil (Bhagavan and Chopra 2007). Very recent papers summarize data about various preparations of CoQ10 with improved solubility and biological availability (Ehrenhaus Masotta et al. 2020, Pastor-Maldonado et al. 2020, Pravst et al. 2020, Takahashi and Takahashi 2019). Water-soluble CoQ10 that improves the bioavailability of natural CoQ10 is non-toxic and stable over long periods of time Ubisol-Q10 prepared by method using polyethylene glycol-derivatised natural compounds (Borowy-Borowski et al. 2004).
Orally administered CoQ10 is absorbed from the gastrointestinal tract (GIT) similarly as vitamin E or different lipid soluble substances (Bhagavan and Chopra 2007). The data disclosed regional differences in permeability throughout the GIT (Palamakula et al. 2005). The maximum permeability occurred in the duodenum pointing at the peculiar expression of transporters along the GIT. The second most favorable region for absorption was the colon followed by the jejunum and stomach. After the absorption in GIT, CoQ10 is reduced and transported to the liver, where it is incorporated into chylomicrons. In the circulation CoQ10 is transported as very low-density lipoprotein (VLDL) or low-density lipoprotein (LDL) particles. After oral administration, the maximum plasma CoQ10 concentration occurs at 6 to 8 hours (Bhagavan and Chopra 2006). Ochiai et al. (2007) found that intestinal absorption of CoQ10 after administration of the emulsion formulation of CoQ10 was also enhanced by food intake in Wistar rats: intestinal absorption was three times faster with food intake. However, the large interindividual variations following the CoQ10 administration were demonstrated. Mantle and Dybring (2020) summarized that the relative bioavaibility and efficacy of administered oxidized and reduced forms of CoQ10 seems to be similar. Moreover, various cell types lining the GIT may have a capacity to reduce CoQ10 and facilitate its conversion. Evidence from pharmacokinetic studies suggest that exogenous supplementation of CoQ10 does not influence the biosynthesis of endogenous CoQ9/CoQ10 nor does it accumulate in plasma or tissues after the cessation of supplementation (Hidaka et al. 2008).
As concerns CoQ10 crossing brain capillary endothelial cells i.e. the blood-brain barrier (BBB) Matthews et al. (1998) showed that oral administration of CoQ10 (200 mg/kg/day for two months) increased both brain and brain mitochondrial concentrations in 12- and 24-month-old male Sprague-Dawley or 24-month-old Fisher 344 rats. Kwong et al. (2002) reported small but significant increase in brain mitochondria in 14-month-old male Sprague-Dawley for 13 weeks feeding (150 mg/kg/day). Smith et al. (2006) used high doses (1,000–5,000 mg/kg/day) of CoQ10 in the R6/2 transgenic mouse model of Huntington’s disease and significantly increased plasma and brain levels of CoQ10 and CoQ9. Kamzalov et al. (2003) found significantly elevated CoQ10 in brain mitochondria but not in the brain homogenate in male mouse C57B1 for 11 weeks feeding. On the other hand, other reports show no increase of CoQ10 with oral administration in 3-month-old or 24-month-old male C57B1 mouse or C65/B16 (Lass et al. 1999, Sohal et al. 2006, Wadsworth et al. 2008).
CoQ10 (30 mg/kg) crossed BBB of adult male Wistar rats and accumulated in the brain when administered intravenously (Belousova et al. 2016). A single intravenous injection of CoQ10 in the rat model of transient focal ischemia led to a 67 % reduction in brain lession size 24 hours after CoQ10 administration and although the infarct had increased by 7 days, it remained smaller than that of saline-treated rats (Belousova et al. 2016). Recently, Wainwright et al. (2020) investigated mechanisms of CoQ10 transport across the BBB, using normal and pathophysiological (CoQ10-deficient) cell culture models and identified lipoprotein-associated uptake and efflux mechanisms regulating CoQ10 transfer. They showed that there is a dynamic interplay of multiple transport receptors with varying degrees of influence. They brought a substantial evidence for the involvement of receptor for advanced glycation endproducts, low density lipoprotein receptor and scavenger receptor in the transport of exogenous CoQ10 into brain.
CoQ10 is well tolerated and safe. Data from experimental and clinical studies indicated repeatedly that CoQ10 is highly safe for the use as a dietary supplement compared with placebo group (Ferrante et al. 2005, Huntington Study Group Pre2CARE Investigators et al. 2010, Kitano et al. 2008, Liu and Artmann 2009, Zhu et al. 2017). Kitano et al. (2008) evaluated the potential subchronic toxicity of reduced and oxidized CoQ10 orally administered to Sprague-Dawley rats (rodents with the primary form of CoQ9) and beagle dogs (mammals with the primary form of CoQ10). The authors observed no adverse effects in male or female rats administered 600 and 200 mg/kg/day, respectively. In the dog study, the highest dose tested (600 mg/kg/day) indicated no adverse effects in males and females. The observed safe level risk assessment method indicated that the evidence of safety is strong at intakes up to 1,200 mg/day (Hathcock and Shao 2006, Ikematsu et al. 2006). Nevertheless, even a daily dosage up to 3,600 mg was found to be tolerated by healthy as well as unhealthy persons (Huntington Study Group Pre2CARE Investigators et al. 2010). In general, CoQ10 shows only a low incidence of adverse events: dizziness, nausea/vomiting, heartburn, stomach upset or related mild and transient gastrointestinal effects (Hathcock and Shao 2006, Raizner 2019).
It should be noted that CoQ10 is not approved by the US Food and Drug Administration for treatment of any medical condition (Raizner 2019). It is sold as a food additive, not as drugs, and its manufacturing is not regulated in the same way as drugs. The European Medicine Agency approved ubiquinol – the reduced form of CoQ10 – as an orphan drug for the treatment of primary CoQ10 deficiency in 2016. According to my knowledge, Myoquinon (Pharma Nord ApS) is approved for medical treatment of proven CoQ10 deficiency or adjunctive therapy to relieve symptoms of chronic heart failure in Hungary.
Coenzyme Q10 analogues
A synthetic short-chain analogue of CoQ10 idebenone (hydroxydecylubiquinone, IDB) was launched by Japanese Takeda Chemical Industries in 1986 (Fig. 3). IDB with a hydroxydecyl side chain (10 carbon atoms) and m.w. 339.44 is less hydrophobic than natural CoQ10 with m.w. 863.49 (Zs-Nagy et al. 1990). IDB acts as an electron carrier in the mitochondrial electron transport chain but it inhibits complex I – NADH dehydrogenase (Esposti et al. 1996, Rauchová et al. 2008). On the other hand, IDB transfers electrons from complex II – succinate dehydrogenase or from glycerol-3-phosphate dehydrogenase to complex III (Brière et al. 2004, Rauchová et al. 2012). Similarly, as CoQ10, IDB acts as a potent antioxidant agent (Lin et al. 2015, Muscoli et al. 2002, Rauchová et al. 2006). IDB is well-tolerated in experiments and clinical trials, e.g. for patients with Friedreich’s ataxia, Huntington’s disease, Alzheimer’s disease, multiple sclerosis etc. (Jaber and Polster 2015). To improve IDB effectiveness in the therapeutic treatment, IDB similarly to CoQ10 can be incorporated in liposomes, cyclodextrins or nanoparticles (Montenegro et al. 2018). Gueven et al. (2015) compared the structural similarity, pharmacokinetics and modulation of cellular energy production of IDB and natural CoQ10. Consequently, IDB and CoQ10 are unable to substitute for each other. A very recent review of Gueven et al. (2021) summarized that IDB (and its metabolites) protect against the multitude of toxic stimuli. The authors also mentioned some new discoveries e.g., competitive inhibition of p52Shc, retinal expression of the RNA-binding protein Lin28A or impact on inflammation and endoplasmatic reticulum stress. Thus, the effects of IDB cannot be explained only by an antioxidant activity or a normalization of mitochondrial energy supply. It seems more likely that IDB activates one or several essential pathways that explain its broad activity (Gueven et al. 2021). A broad range of IDB activity is very similar to multiple functions of coenzyme Q but the activities might also be different because of different chemical structure.
Fig. 3.
Chemical structure of synthetic analogue of CoQ10, idebenone with only 10 carbons in its aliphatic side chain
Mitoquinone (MitoQ) was developed for oxidative stress treatment (James et al. 2005, Smith and Murphy 2010). Structure of MitoQ consists of lipophilic cation – triphenylphosphonium (TPP) attached to ubiquinone head group via a 10-carbon aliphatic chain (Fig. 4). MitoQ crosses easily through all biological membranes, including the blood-brain barrier and neuronal membranes. MitoQ concentrates hundredfolds in mitochondria driven by high membrane potential across the inner mitochondrial membrane. Oral delivery of MitoQ protects mitochondria from damage and may therefore form the basis for mitochondria-protective therapies (Murphy and Smith 2007). It was shown that MitoQ had a protective role in animal and cell models of several human disease, including Parkinson’s disease, Alzheimer’s disease or sclerosis multiplex.
Fig. 4.
Chemical structure of MitoQ
CoQ10 deficiency
Primary CoQ10 deficiency, which can cause reduced levels of CoQ10 in tissues, includes mutations in the genes participating in the complicated biosynthesis of CoQ10, as described by Mollet et al. (2007), Ogasahara et al. (1989), Quinzii et al. (2006), Rötig et al. (2000) and others. In many cases the family history suggests an autosomal recessive mode of inheritance. Alzár-Fabra et al. (2018) presented the updated comprehensive review of the very heterogeneous clinical spectrum associated with primary CoQ10 deficiency. At present 10 genes encoding biosynthesis of CoQ10 have their pathogenic variants bringing about human CoQ10 deficiency (Alzár-Fabra et al. 2018). Besides of cardiac, muscular and renal manifestations the peripheral and central nervous system is often affected including encephalopathy, seizures, cerebellar ataxia, epilepsy, sensorineural hearing loss, optic atrophy or intellectual disability in these patients.
Primary CoQ10 deficiency is very rare (Yu et al. 2019). Much more frequent secondary CoQ10 deficiency includes the defects not directly connected with biosynthesis of CoQ10 (Alzár-Fabra et al. 2018, Desbats et al. 2015, Yubero et al. 2016). Many reports focus on skeletal muscle and central nervous system. Muscular manifestations consist of weakness, hypotonia, exercise intolerance and myoglobinuria, while the main CNS manifestations include ataxia and general CNS impairment (Desbats et al. 2015).
CoQ10 suplementation can be effective for treatment of both primary and secondary CoQ10 deficiencies (Potgieter et al. 2013). Duberley et al. (2014) evaluated the effect of CoQ10 supplementation on the well-established neuronal model of CoQ10 deficiency. Human SH-SY5Y neuronal cells with 1 mM competitive inhibitor of mammalian CoQ biosynthesis in cell cultures – para-aminobenzoic acid – induced approximately a half decrease of cellular CoQ10 concentration accompaining by a fourfold increase in mitochondrial oxidative stress and global loss of mitochondrial respiratory chain enzyme activities. Following CoQ10 supplementation for 5 days there was a markedly increased cellular CoQ10 status. CoQ10 treatment (2.5 μM) significantly decreased the level of mitochondrial superoxide in the CoQ10-deficient neurons. CoQ10 treatment (5 μM) restored mitochondrial membrane potential to 90 % of the control level. However, CoQ10 treatment (10 μM) was only partially effective in restoring enzyme activities of mitochondrial respiratory chain. This study indicated that although mitochondrial oxidative stress can be attenuated in CoQ10-deficient neurons following CoQ10 supplementation, mitochondrial respiratory chain enzyme activities appear to be partially refractory to this treatment. The therapy with >10 μM CoQ10 may be required to restore mitochondrial respiratory chain enzyme activities to the control level.
Very important for the treatment of primary or secondary CoQ10 deficiency is early identification and early supplementation to omit irreversible damages in the central nervous system (and other organs).
Migraine
Migraine is a debilitaiting condition characterized by headaches and nausea with a usual onset around puberty. Associated symptoms may be sensitivity to light, sound or smell and vomiting. Migraine affects 10 % people worldwide and it is approximately three times more common in women than in men (Vos et al. 2012, Woldeamanuel and Cowan 2017). Although migraine ranks among the most frequent neurological disorders, its pathophysiology remains not fully understood. Mitochondria and energetic metabolism have long been postulated to be involved in the etiology of migraines, although a direct link has been difficult to identify (Sparaco et al. 2006, Yorns and Hardison 2013).
A small open-label study with 32 adults showed some effectiveness of 150 mg of CoQ10 supplementation as a preventive treatment for migraine headaches (Rozen et al. 2002). The other (double-blind, randomized, placebo-controlled) study with 42 adult migraine patients used CoQ10 supplementation (3×100 mg/day). CoQ10 was superior to placebo for attack frequency, headache-days and days-with-nausea in the third treatment month (Sándor et al. 2005).
Hershey et al. (2007) found that deficiency of CoQ10 may be quite common in pediatric and adolescent migraine. They measured plasma CoQ10 in 1550 patients (aged 13 to 22 years). Consequent CoQ10 supplementation in patients with low plasma CoQ10 (1 to 3 mg/kg/day of CoQ10 in liquid gel capsule formulation) resulted in clinical improvement. In a double-blinded, placebo-controled study with 120 pediatric and adolescent migraine receiving 100 mg CoQ10 supplementation for 32 weeks, the improvement was seen in weeks 1–4 but there was no difference in headache outcomes between the CoQ10 supplementation and placebo in week 32 (Slater et al. 2011). CoQ10 supplementation (400 mg/day per 3 months together with prophylactic medication) showed significant improvement in frequency, severity and duration of migraine attacks in CoQ10 group compared to placebo in 45 women aged 18–50 years. Interestingly, the levels of inflammatory marker tumor necrosis factor-α (TNF-α) were reduced significantly but interleukin IL-6 and IL-10 levels were not affected (Dahri et al. 2019).
Recently, Zeng et al. (2019) included five studies from 2002–2018 with 346 patients (120 pediatric and 226 adult subjects) in the meta-analysis. CoQ10 was comparable with placebo in respect to migraine attacks/month and migraine severity/day. However, CoQ10 was more effective than placebo in reducing migraine days/month and migraine duration. Another recent meta-analysis included four studies from 2005–2018 with 221 patients (Parohan et al. 2020). They concluded that the greatest impact of CoQ10 supplementation was on the frequency of attacts per month without affecting the severity or duration of migraine attacks.
In addition to CoQ10 several other nutraceuticals have been demonstrated to have potential effectiveness for migraine prevention such as magnesium, riboflavin, carnitine or herbal remedies and their different combinations together with aerobic exercise or other forms of relaxation training, cognitive therapies and acupuncture (Mauskop 2012). A double-blind, placebo-controled study with 130 adult patients (18–65 years) evaluated the efficacy of a proprietary supplement containing magnesium, riboflavin and Q10 (commercially available food supplement Migravent® in Germany or Dolovent® in USA) (Gaul et al. 2015). After three months of treatment with this supplement, a pain and burden of migraine were statistically significantly reduced compared to placebo patients, but it did not show statistically significant efficacy on migraine frequency. Results with another proprietary supplement containing feverfew (Tanacetum parthenium), CoQ10 and magnesium (Antemig® in France since 2014) could be beneficial and safe for the prevention of migraine in adult patients (Guilbot et al. 2017). A double-blind 8-week lasting study with concurrent CoQ10 (30 mg/day) and carnitine (500 mg/day) supplementation provided the evidence supporting some beneficial effects on mitochondrial metabolism (decrease of lactate serum level) and migraine symptoms (Hajihashemi et al. 2019). Parohan et al. (2021) reported a synergistic effect of nano-curcumin and CoQ10 (300 mg/day) given for 8 weeks in a double-blind, placebo-controled study with 91 adult patients.
Parkinson’s disease
Parkinson’s disease (PD) is one of the most common neurodegenerative diseases with still unknown primary cause. There is a long-term degenerative disorder of the central nervous system that mainly affects the motor system and affects about 1–2 % of the elderly population with usual onset over 60. It is prevalent in males. The most obvious symptoms are shaking (due to decrease in dopaminergic modulation of the substantia nigra pars compacta neurons altering motor systems), rigidity, slowness of movement and difficulty with walking. Inclusions of Lewy bodies in midbrain and presence of immunoreactive α-synuclein and ubiquitin on autopsy are hallmarks of the disease (Oertel and Schulz 2016, Stanga et al. 2020). Mitochondrial dysfunction contributes significantly to neuronal loss in PD (McCoy and Cookson 2012, Stanga et al. 2020). Mitochondria of patients have decreased complex I activity not only in substantia nigra (Schapira et al. 1990a, b), striatum (Mizuno et al. 1989) and frontal cortex (Parker et al. 2008) but also in skeletal muscle (Bindoff et al. 1991) or platelets (Haas et al. 1995, Krige et al. 1992). The other study found significant deficiency of CoQ10 in cortex but not in substantia nigra, striatum or cerebellum in brains samples of PD patients (Hargreaves et al. 2008). Studies also reported deficiency of CoQ10 platelets (Krige et al. 1992, Shults et al. 1997). Functional intracellular assay (FIA) methodology analyzing blood samples also assessed CoQ10 deficiency in patients with PD compared to age- and gender-matched controls (Mischley et al. 2012). In addition, their mitochondria show oxidative stress, disturbance in calcium homeostasis, impaired clearance of dysfunctional mitochondria, changes in mitochondrial biogenesis and the loss of membrane potential (McCoy and Cookson 2012).
Inhibitors of mitochondrial complex I of the oxidative phosphorylation pathway induce degeneration of dopaminergic neurotransmission in rodents and several other animal models of PD (Hamadjida et al. 2019, Tieu 2011). The most often used chemicals are MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (Beal et al. 1998, Jackson-Lewis and Przedborski 2007, Przedborski et al. 2004), nonselective herbicide paraquat (PQ), N,N′-dimethyl-4-4′-bipiridinium (Cicchetti et al. 2005, McCarthy et al. 2004), or botanical insecticide and nonselective piscicide rotenone (Caboni et al. 2004, Cannon et al. 2009, Sherer et al. 2003). Many scientists found some benefit of CoQ10 supplementation in the above-mentioned experimental animal models of PD. Cleren et al. (2008) showed significant neuroprotective effects of CoQ10 against acute treatment with MPTP, which produced severe dopamine depletion in 5-month-old male mice. Transgenic mice with DJ-1 (Parkinson disease protein 7) deficiency (DJ-1/PARK7) with the hypersensitivity to MPTP showed a clear neuroprotection by a prophylactic use of Ubisol-Q10, water-soluble nanomicellar formulation of CoQ10 (Muthukumaran et al. 2014b). Attia and Maklad (2018) indicated that CoQ10 supplement (200 mg/kg) caused a remarkable improvement in most of the behavioral tests and decreased protein carbonyl content in the brain in mouse PQ model, particularly when treatment of CoQ10 started prior rather than after PQ induction of PD. Ubisol-Q10 given in drinking solution (12 mg/kg/day) was effective in blocking the progression of neurodegeneration in PQ rat model when administered therapeutically (after PQ injection) (Muthukumaran et al. 2014a). However, Ubisol-Q10 must be given continuously and cannot be withdrawn in order to continue neuroprotection. The withdrawal led to further neurodegeneration. The authors suppose that Ubisol-Q10 halts neurodegeneration by supporting of remaining neurons (Muthukumaran et al. 2014a).
Furthermore, there are also cellular models, which develop pathology more quickly, do not require ethical approval and are less costly (Falkenburger et al. 2016). Treatment with water-soluble CoQ10 was succesful in human neuroblastoma (SH-SY5Y) cells paraquat model of PD (i.e. 10 μM paraquat in complete media for 48 hours at 37 °C), where it inhibited reactive oxygen species generation, reduced the number of apoptotic cells and DNA fragmentation (McCarthy et al. 2004). Another in vitro PD model of SH-SY5Y cells involved the addition of 6-hydroxydopamine (6-OHDA). 6-OHDA did not induce extensive oxidative damage of mitochondria but only a mild redox signal, which activated the machinery of mitochondrial fission and disrupted the mitochondrial morphology (Solesio et al. 2013). Treatment with mitochondria-targeted antioxidant MitoQ hand, IDB transfers electrons from complex II–succinate through its capacity to alter mitochondrial redox processes hand, IDB transfers electrons from complex II–succinate inhibited the migration of cytosolic dynamin-related protein-1 (Drp1) and pro-apoptotic protein Bax to the mitochondria and reduced the mitochondrial morphology alterations induced by 6-OHDA (Solesio et al. 2013). Xi et al. (2018) confirmed the protective effects of MitoQ on mitochondrial dynamics in 6-OHDA-induced in vitro (SN4741 cells derived from substantia nigra of mouse embryos) and in vivo models of PD (adult C57bl/6 male mice).
However, there are differences in the pathogenesis of the PD models and of human PD. PD models acts on complex I through a competitive inhibitory process for which CoQ10 supplementation is beneficial. The idea that supplementation of CoQ10 could retard the rate of PD progression of patients is rather old (Müller et al. 2003, Shults et al. 2002, Strijks et al. 1997, Yoritaka et al. 2015). Strijks et al. (1997) did not notice any beneficial effect in 10 patients treated with 200 mg of CoQ10 for 3 months. Shults et al. (2002) used three dosage of CoQ10 (300, 600 and 1200 mg/day) in 80 subjects with early PD for 16 months. The patients developed less disability, and the benefit was greatest in subjects receiving the highest dosage. Müller et al. (2003) indicated a moderate beneficial effect in PD patients (seven males and seven females) after oral application of 360 mg of CoQ10 lasting 4 weeks. Yoritaka et al. (2015) also indicated benefit for Japanese PD patients, who took 300 mg ubiquinol-10 capsules for 96 weeks. The reason can be that not all PD patients have low serum CoQ10 levels. Jiménez-Jiménez et al. (2000) found similar serum CoQ10 levels in 33 PD patients and 31 matched controls. However, earlier report showed decreased serum CoQ10 levels in PD patients (Matsubara et al. 1991).
Unfortunately, recent reviews and meta-analysis disclose that CoQ10 supplement plays only a limited role in the treatment of PD. It does not slow functional decline nor provides any symptomatic benefit for patients with PD (Negida et al. 2016, Oertel and Schulz 2016, Parkinson Study Group QE3 et al. 2014, Zhu et al. 2017).
Huntington’s disease
Huntington’s disease (HD), also known as Huntington’s chorea or Saint Vitus’ dance, is a rare autosomal dominant inherited neurodegenerative disease with the origin of symptoms at the age of 30–50 years. HD is characterized by progressive motor, behavioral and cognitive decline, resulting in death within 15 to 20 years after the diagnosis for which there is no effective therapy. The genetic defect of HD is caused by cytosine-adenine-guanine (CAG) trinucleotide expansion in the huntingtin gene resulting in the production of an expanded polyglutamine in the mutant huntingtin protein. The cure is based largely on symptomatic treatment and lifestyle interventions with supportive management (Kumar et al. 2020, Yu and Bega 2019).
Mitochondria and energetic metabolism dysfunction as well as oxidative stress contribute to the neurodegenerative process (Koroshetz et al. 1997, Tobore 2019). However, antioxidant treatment with CoQ10 and vitamin E was not very successful in HD models. Combined administration (250 mg CoQ10 + 530 mg vitamin E/kg/day) could not prevent the decline of brain respiratory chain function in the HD model of Wistar rats injected with 3-nitropropionic acid (Kasparová et al. 2006). Combined supplementation did not also increase survival rates and pupal mortality of flies in the HD model of Drosophila melanogaster (Bahadorani and Hilliker 2008). It seems that antioxidants alone are not enough to delay or to stop the progress of HD. Two recent extensive reviews tried to answer the questions connected with the role of antioxidants and their therapeutic possibilities in HD (Bono-Yagüe et al. 2020, Essa et al. 2019).
Ranen et al. (1996) undertook a one-year, placebo-controlled, double-blind study with IDB. They did not find any significant differences between 91 patients receiving synthetic analogue of CoQ10 IDB (90 mg/day) or placebo group. The next placebo-controlled 30-month-trial tried to assess the impact of CoQ10 (300 mg twice daily) and a noncompetitive glutamatergic NMDA (N-methyl D-aspartate) receptor antagonist, remacemide hydrochloride (200 mg three times daily) but neither CoQ10 nor remacemide produced significant slowing in functional decline in early HD patients (Huntington study group 2001). On the other hand, several studies report beneficial effects of CoQ10 on behavior and pathology in mouse models of HD. The transgenic HD mice (R6/2) are widely used as a fast-progressing model in trials testing potential therapies. R6/2 mice with oral administration of CoQ10 or remacemide significantly increased survival and delayed the development of many HD symptoms. Combined treatment (CoQ10+remacemide) was even more efficient (Ferrante et al. 2002). The authors suggest several potential explanations for the observed discrepancy: the dose of remacemide was 2.5-fold higher in mice, the pathophysiology of neurodegeneration in the transgenic mice may not be entirely reminiscent of that occurring in human patients and the disease stage in which the therapeutic trials were initiated was markedly different (Ferrante et al. 2002). Schilling et al. (2001) also found the amelioration of motor deficit but no prolonged survival of transgenic HD-N171-82Q mice after a combined treatment (CoQ10+remacemide). Combined tetracycline antibiotic with antimicrobial and antiinflammatory properties minocycline and CoQ10 treatment provided an amelioration of behavior and neuropathological alterations (Stack et al. 2006). Moreover, this therapy improved motor performance to a greater degree than either minocycline or CoQ10 alone and significantly extended the survival of the R6/2 mice. In addition, combined minocycline and CoQ10 treatment attenuated gross brain atrophy, striatal neuron atrophy and specific protein huntingtin aggregation relative to the treatment with other drugs. However, Menalled et al. (2010) did not confirm the previously reported benefits. They found that neither CoQ10 nor minocycline caused significant improvements on measures of motor function or general health, e.g. open field, rotarod, grip strength, rearing-climbing, body weight and survival in the R6/2 mouse model. The authors discuss possible reasons of discrepancies, such as different founder lines of R2/6 mice, effect of nutrition and husbandry or number of experimental animals and behavior tests used. On the other hand, Hickey et al. (2012) used a slowly progressing HD model, the homozygote mutant CAG140 knock-in mouse, which expresses the full-length protein in the proper genomic context and may better reproduce human pathology. The mice display progressive motor, cognitive and emotional anomalies, transcriptional disturbances and late striatal degeneration. The authors report beneficial effects of 0.2 % CoQ10 in diet on motor behavior. Suprissingly, the lower (0.2 %) dose of CoQ10 was more effective than the higher (0.6 %) dose. The data emphasize that maximum benefit may be observed when treatment is begun at early stages of the disease, when neuropathological changes are minimal. It can explain the different results between Menalled et al. (2010) using the more rapid R6/2 mouse model and Hickey et al. (2012) who studied a slowly progressing CAG140 knock-in mouse model.
Another combined therapy (with creatine) produced additive neuroprotective effects, such as improving of motor performance and survival extension, in R6/2 transgenic mouse model of HD (Yang et al. 2009). Smith et al. (2006) performed a study administering high levels of CoQ10 to R6/2 transgenic mice (from two different commercial sources). High doses of CoQ10 (1,000, 5,000, 10,000 or 20,000 mg/kg/day significantly extended survival and improved motor performance and grip strength, reduced weight loss and brain atrophy in R6/2 at 90 days.
Unfortunately, a recent multicenter randomized, double-blind study with 609 patients with early stage of HD (from the United States, Canada and Australia) obtaining 2,400 mg per day (or placebo) for 60 months showed no beneficial effect (McGarry et al. 2017). The trial was concluded early on the basis of an interim analysis and futility.
Alzheimer’s disease
The most common, progressive, irreversible and fatal brain disease is Alzheimer’s disease (AD), which disturbs cognition and memory functions. AD is strongly associated with increasing age with usual onset over 65 years old. Globaly, the greatest contributors to AD risk are smoking following by diabetes, mid-life hypertension, mid-life obesity, depression and physical inactivity (Barnes and Yaffe 2011). AD affects about 40 million individuals worldwide. AD’s Association reports extensive analysis of information of AD, including incidence and prevalence, mortality rates, heath expenditures, cost of care and effect on caregivers and society in general in the Unites States (Alzheimer’s Association 2013). Unfortunately, the cause of AD is not quite understood, the cure is not known, the prognosis remains poor, and the number of suffering people is increasing what impose an extreme burden to public healthcare systems worldwide. AD is characterized by the presence of pathological features in brain: amyloid-β peptide plaques and neurofibrillary tangles in pyramidal neurons. In addition, AD covers many neuropathologies and pathophysiological processes, such as neuroinflammation (Sochocka et al. 2017), oxidative stress (Atwood et al. 1999), vascular dysfunction (Di Marco et al. 2015), dendritic (Baloyannis 2009) and synaptic pathologies (Manyevitch et al. 2018). Damaged mitochondria also have an important role (Baloyannis 2006, Stanga et al. 2020, Wang et al. 2020). Moreover, Joshi et al. (2015) described Golgi apparatus fragmentation, which results in enhanced amyloid precursor protein and amyloid-β production.
Södeberg et al. (1992) reported increased levels of CoQ10 in postmortem brain samples. In observed brain regions of AD patients such as frontal cortex, precentral cortex, temporal cortex, frontal white matter, nucleus caudatus, hippocampus, pons, cerebellum and medulla oblongata, CoQ10 levels increased significantly compared to the controls, with the elevations varying between 30 and 100 %. The elevated levels of CoQ10 may reflect a change in oxidative stress (Södeberg et al. 1992). As concerns on plasma CoQ10, De Bustos et al. (2000) compared serum CoQ10 level and CoQ10/cholesterol ratio in 44 patients with AD, 17 patients with vascular dementia and 21 matched controls and the data were not significantly different. The values did not correlate with age, age at onset, duration of disease or score in cognitive tests. Other study confirmed no significant difference between 42 cognitively intact and 23 AD patients on plasma CoQ10 level (Giavarotti et al. 2013). However, Fišar et al. (2019) revealed an association between plasma CoQ10 concentration and the MMSE score in AD patients. The authors supposed that an even insignificant decrease in plasma CoQ10 concentration might play a role in the cellular dysfunction found in AD patients.
In the nineties, two smaller studies described improvements of memory, attention and behavior after IDB administration (Bergamasco et al. 1994, Senin et al. 1992). Gutzmann and Hadler (1998) conducted the two-year prospective, randomized, double-blind multicentre study with 450 patients with dementia of the AD type of mild to moderate degree. Their results suggested that IDB exerted its beneficial therapeutic effects on the course of the disease by slowing down its progression. However, Thal et al. (2003) in a one-year, multicenter, double-blind, placebo-controlled, randomized trial found that IDB failed to slow cognitive significant decline in 536 patients aged over 50 years with a diagnosis of probable AD and mild to moderate cognitive test (MMSE) scores. Patients were treated with three different doses of IDB 120, 240, or 360 mg three times daily. Galasko et al. (2012) discovered that antioxidants (400 mg of CoQ10 3 times daily or 800 IU of vitamin E plus 500 mg of vitamin C plus 900 mg of α-lipoic acid daily) for 4 months did not influence cerebrospinal fluid biomarkers related to amyloid or tau pathology of patients with mild to moderate AD.
There are different mice models of AD disease. Yang et al. (2008) tested the effect of CoQ10 on β-amyloid in the 16-month-old transgenic mice overexpressing the Alzheimer presenilin 1-L235P mutation (leucine-to-proline mutation at codon 235). Mice, which were fed with CoQ10 (1,200 mg kg/day) for 60 days, effectively decreased amyloid-β overproduction and depressed oxidative stress. Additionally, CoQ10 treatment improved markers of oxidative stress such as downregulation of superoxide dismutase and increased levels of malondialdehyde in transgenic mice relative to the wild-type mice. Another study found that CoQ10 treatment of TG19959 transgenic mouse model of AD decreased brain levels of protein carbonyls, a marker of oxidative stress, and provided protection against plaques and memory loss, as measured by the Morris water maze testing (Dumont et al. 2011). Transgenic mice with the P301S tau mutation, which causes frontotemporal dementia in man, were fed a control or 0.5 % CoQ10 diet (Elipenahl et al. 2012). The results show that CoQ10 significantly improved behavioral deficits and survival in transgenic mice with the P301S tau mutation. The authors also described a significant increase in mitochondrial complex I activity and protein levels and reduced oxidative stress with a reduction in lipid peroxidation. Muthukumaran et al. (2018) evaluated the neuroprotective effects of water-soluble formulation of CoQ10, Ubisol-Q10 in drinking water (at a dose of 6 mg/kg/day) in one-month-old double transgenic male AD mice containing human/mouse chimeric amyloid β precursor and a mutant presenilin-1 gene. Ubisol-Q10 treatment reduced circulating amyloid peptide, improved long term memory, preserved working spatial memory and drastically inhibited amyloid plaque formation in 18-month-old transgenic mice compared to an untreated transgenic group. Ubisol-Q10 treatment also activated autophagy in AD fibroblasts (presenilin-1 mutated) as well as in the brains of transgenic AD mice (Vegh et al. 2019). The authors found increased expression of autophagy-related genes beclin-1 and JNK1 following Ubisol-Q10 treatment. However, withdrawal of Ubisol-Q10 treatment led to the return of the former phenotype in AD fibroblasts indicating that constant supplementation of Ubisol-Q10 is required.
The mitochondria-targeting antioxidant MitoQ significantly increased neurite outgrowth and synaptic connectivity in neuron cell culture from a mouse model of AD, amyloid-β precursor protein transgenic mice (Manczak et al. 2010). MitoQ treatment also prevented amyloid-β-induced oxidative stress and all death of mouse cortical neuron cell culture from another model of AD, triple transgenic mice expressing three mutant human genes: amyloid-β precursor protein, presenilin-1 and four-repeat tau (McManus et al. 2011). The study focused on prevention of AD development described the improvement of cognitive decline, oxidative stress, β-amyloid accumulation, astrogliosis, synaptic loss and caspase activation in young triple transgenic mice given MitoQ in the drinking watter at two months of age and continued for 5 months, i.e. the period during which the first AD-like pathologies become manifest (McManus et al. 2011). Young and Franklin (2019) focused on the therapy for AD. They evaluated the effects of MitoQ treatment on cognitive decline and neuropathologies in triple transgenic mice starting at 12 months after birth and continuing until 18 months of age, i.e. in the period during which all of the known AD-like pathologies are present and progressing. The authors found that MitoQ treatment of older AD mice was effective in improving memory retention compared to untreated mice. MitoQ-treated mice showed improved memory retention compared to untreated triple transgenic AD mice as well as reduced brain oxidative stress, synapse loss, astrogliosis, microglial cell proliferation, amyloid-β accumulation, caspase activation, and tau hyperphosphorylation. Additionally, MitoQ treatment inhibited synapse loss and significantly increased the abbreviated lifespan of the triple transgenic AD mice. These findings support the involvement of mitochondria-derived oxidative stress in the etiology of AD and suggest that MitoQ may lessen symptoms in AD patients.
Amyotrophic lateral sclerosis
Amyotrophic lateral sclerosis (ALS) is a devastating disease characterized by the death of motor neurons. The loss of motor neurons controlling voluntary muscles causes generalized progressive muscle weakness. Over time, patients lose the ability to walk, use their hands, speak, swallow, and breathe. Most ALS patients die within 3 to 5 years after diagnosis, usually as a result of respiratory failure. The ALS cause is mostly not known (about 10 % is familiar) and the effective therapy is still lacking. Mitochondrial alteration and oxidative stress are associated with ALS (Barber and Shaw 2010, Smith et al. 2019). Recently, Bond et al. (2018) and Wang et al. (2019) reported two systematic reviews on oxidative stress markers in ALS.
Molina et al. (2000) compared serum levels of CoQ10 and CoQ10/cholesterol ratio in 30 patients with ALS and 42 matched controls using a high-performance liquid chromatography. The mean serum CoQ10 levels and the CoQ10/cholesterol ratio did not differ significantly between the two studied groups. These values were not influenced by the clinical form (spinal vs. bulbar) of ALS, and they did not correlate with age, age at onset, and duration of the disease. These results suggest that serum CoQ10 concentrations are unrelated to the risk for ALS. However, Sohmiya et al. (2005) observed a significant increase in oxidative form of CoQ10 and in the ratio of oxidized form of CoQ10 to total CoQ10. Moreover, percentage of CoQ10 correlated significantly with the duration of illness suggesting systematic oxidative stress in the pathogenesis of the disease. Similarly, a significantly increased percentage of oxidized CoQ10 in total plasma CoQ10 in ALS patients was reported by Nagase et al. (2016). Significantly higher plasma levels of thiobarbituric acid-reactive species (TBARS), marker of lipid peroxidation damage, but without changes in concentrations of plasma antioxidants (α-tocopherol, β-carotene, ubiquinol-10 and glutathione) in ALS patients compared to healthy controls were described by Oteiza et al. (1997).
Matthews et al. (1998) found that daily oral administration of 200 mg CoQ10/kg attenuated striatal lesions produced by systemic administration of 3-nitropropionic acid and slightly but significantly increased life span in a transgenic mouse model of familial ALS. On the other hand, the treatment of mouse model of familiar ALS (SOD1G93A) with oxidized (or reduced) CoQ10 had no effect on disease progression (Lucchetti et al. 2013). Experimental studies with targeted metabolic therapies supporting energy metabolism, which also contain CoQ10, improved motor function, quality of life and survival time of ALS patients (Ari et al. 2014). A promising candidate for ALS treatment seems mitochondria-targeted antioxidant MitoQ, which has already been authorized for human medicine (Smith and Murphy 2010). Oral administration of MitoQ to SODG93A mice starting at the onset of the symptoms extended their survival and improved grip strength (Miquel et al. 2014). MitoQ in SODG93A mice also improved mitochondrial respiratory function in spinal cord and muscle, decreased nitrosative markers in central nervous system, decreased motor neuron loss and astrocyte reactivity in the spinal cord and maintained motor unit integrity.
In a clinical pilot trial, Ferrante et al. (2005) assessed the safety and tolerability of high doses of CoQ10 in ALS. They disclosed that doses as high as 3,000 mg CoQ10/day are safe and well tolerated in 31 ALS patients for as long as 8 months. However, the serum CoQ10 level reached a plateau at a dose 2,400 mg/kg indicating that further studies need not exceed this high dose. Phase II trial of CoQ10 for ALS (dose selection with 35 participants per group) compared two doses: high (2,700 mg/day) and moderately high (1,800 mg/day) in 9-month-period (Levy et al. 2006, Kaufmann et al. 2009). Unfortunately, the authors did not find any significant differences between selected dose 2,700 mg CoQ10/day and placebo subjects with 75 participants per group. Thus, the results did not provide sufficient evidence to justify a phase III trial for ALS treatment.
Friedreich’s ataxia
Friedreich’s ataxia (FA) is an autosomal recessive genetic and slowly progressive disease that causes difficulty in walking, a loss of sensation in the arms and legs and impaired speech. Moreover, life-threatening hypertrophic cardiomyopathy is the most severe manifestation. Reduced levels of the mitochondrial protein frataxin lead to cell-damaging oxidative stress (Bűrk 2017). CoQ10, IDB and vitamin E were used as antioxidant treatment.
In an open-label study of 10 FA patients, antioxidant treatment (400 mg CoQ10 plus 2,100 IU vitamin E per day) did not show any consistent benefits in the neurological evaluation after 6-month treatment (Lodi et al. 2001). In all patient’s serum CoQ10 level was increased and cardiac and skeletal muscle bioenergetics showed an improvement after 3-month-treatment (Lodi et al. 2001). The 4-year-follow-up study with 77 patients confirmed an improvement in cardiac and skeletal muscle mitochondrial energy synthesis but only 7 patients showed some neurological improvement (Hart et al. 2005). A high dose antioxidant therapy (600 mg CoQ10 plus 2,100 IU vitamin E per day) provided no additional benefit when compared to a very low-dose antioxidant therapy (30 mg CoQ10 plus 24 IU vitamin E per day) according to a two-year double-blind study with 50 FA patients (Cooper et al. 2008). Serum CoQ10 levels increased significantly in all patients. When compared cross-sectional data 49 % of all patients with CoQ10 brought some evidence for stabilizing effects on the progression of some neurological parameters monitored by International Cooperative Ataxia Ratings Scale (ICARS).
The early studies with antioxidant and synthetic analogue of CoQ10, idebenone (IDB) used very low dose (5 mg/kg/day) and the numbers of participants were usually low (Artuch et al. 2002, Buyse et al. 2003, Mariotti et al. 2003, Rustin et al. 1999). The studies showed various results in neurological aspects: from some improvement (Artuch et al. 2002, Di Prospero et al. 2007, Rustin et al. 1999) or no significant changes (Mariotti et al. 2003) till progressive worsening (Buyse et al. 2003). A retrospective analysis of 35 patients used IDB (5 mg/kg/day) for up to five years showed significantly deteriorated neurological parameters (Rinaldi et al. 2009). Brandsema et al. (2010) used intermediate dose of IDB (20 mg/kg/day) in 7 children’s patients. More than one-year therapy did not statistically change ICARS and Pediatric Quality of Life Inventory scores. Markedly higher doses of IDB were used in randomized, double-blind, placebo-controlled study (IONIA) with 70 ambulatory pediatric patients who were treated with low doses: 450 or 900 mg of IDB per day depending on body weight (≤ or > 45 kg) or high doses: 1,350–2,250 mg of IDB per day depending on body weight (≤ or > 45 kg) (Lynch et al. 2010). The study did not show any significant neurological improvement during the 6-month period. Sixty-eight patients followed by a 12-month-study (IONIA-E) were treated only with a high dose of IDB: 1,350–2,250 mg/day depending on body weight (≤ or > 45 kg). The authors concluded that this dose may offer a therapeutic benefit to pediatric FA patients by stabilizing the overall neurological function and improving fine motor skills and speech (Meier et al. 2012). Comprehensive reviews on the use of CoQ10 and IDB in FA were prepared by Spindler et al. (2009) and Parkinson et al. (2013).
Multiple sclerosis
Multiple sclerosis (MS) is a chronic, autoimmune, inflammatory and demyelinating neurodegenerative disease of central nervous system with a broad spectrum of motor, sensory and neuropsychiatric symptoms but the etiology is still unknown. Although there are currently drugs that can alleviate the symptoms of MS, the cure is not known. Mitochondrial dysfunction and abnormalities play their role in MS pathology (Campbell et al. 2012, Witte et al. 2013). Oxidative stress also accompanies this disease (Acar et al. 2012, Choi et al. 2018, Haider et al. 2011, Tasset et al. 2012).
Gironi et al. (2014) estimated significantly lower blood CoQ10 levels in MS patients in comparison with healthy subjects. On the other hand, de Bustos et al. (2000) did not find any significant differences between serum CoQ10 (and the CoQ10/cholesterol ratio) in a series of 31 patients with MS and 19 controls. Moreover, they did not find any correlation with age, age at onset or the duration of MS suggesting that the serum CoQ10 levels are an unrelated marker of risk or activity of MS. The possible explanation of the above difference can be that Gironi et al. (2014) had MS patients free of relapse or disease progression in the past 30 days, while de Bustos et al. (2000) studied patients during MS exacerbation. Therefore, the normal CoQ10 levels could be explained by an attempt of the organism to increase antioxidant mechanisms during an inflammatory phase of disease, which might thus compensate constitutive low CoQ10 levels Gironi et al. 2014).
CoQ10 supplementation (500 mg/day for 12 weeks) decreased levels of the marker of oxidative damage, malondialdehyde and increased the activity of antioxidant enzyme superoxide dismutase. Nevertheless, total antioxidant capacity was decreased, and glutathione peroxidase activity was not affected in 48 patients with relapsing-remitting MS in a double-blind, placebo-controlled randomized clinical trial (Sanoobar et al. 2013). The same CoQ10 supplementation (500 mg/day for 12 weeks) lowered levels of two pro-inflammatory markers: serum tumor necrosis factor-α and interleukin-6 but did not change levels of anti-inflammatory markers: tumor necrosis factor-β and interleukin-4 in 48 patients with relapsing-remitting MS (Sanoobar et al. 2015). Participants reported reduced fatigue and depression compared to the placebo group (Sanoobar et al. 2016). However, further clinical trials with long-term observation and more participants are needed to elucidate the benefit of CoQ10 particularly in the immune-related inflammation processes (Zahednasab et al. 2015). Moccia et al. (2019) evaluated the wide set of laboratory markers of oxidative stress and inflammation in 60 relapsing-remitting patients with MS treated with 44 μg interferon-ß1a and with 200 mg CoQ10. After 3-month-period, CoQ10 supplementation improved the scavenging activity, reduced the oxidative damage and induced shift towards a more anti-inflammatory milieu in peripheral blood of patients.
CoQ10 is also mentioned in some papers dealing with the nutritional parameters and suitable diets for MS patients (Armon-Omer et al. 2019, Bagur et al. 2017, Evans et al. 2018, Marx et al. 2020, Zuliani and Baroni 2015).
The most widely used animal model of MS is mouse experimental autoimmune encephalomyelitis (EAE). Mao et al. (2013) found that EAE (C57BL/6) mice with MitoQ pretreatment and treatment (i.p. 100 nmol/mouse (~30 g) twice per week for several weeks) reduced axonal loss and neurological disabilities associated with EAE. Moreover, a mitochondria-targeted antioxidant MitoQ significantly suppressed demyelination and inflammation of EAE, including the inhibition of inflammatory cytokines and chemokines. The authors confirmed neuroprotective and antioxidant roles of MitoQ by a co-culture of cortical neurons and microglia designed to mimic the mechanism of MS and EAE in vitro. Similarly, CoQ10 administration (i.p. 10 mg/kg/three weeks) decreased significantly clinical symptoms and the level of the tumor necrosis factor α (TNF-α) versus interleukin 10 (IL-10) in EAE (C57BL/6) mice. Thus, CoQ10 was capable to suppress the inflammatory pathway of MS (Soleimani et al. 2014).
Another experimental model for studying demyelination-remyelination uses a copper chelator, cuprizol (CPZ), which inhibits copper ions and causes oxidative stress, oligodendrocyte apoptosis and demyelination. CoQ10 treatment of C57Bl/6 mice alleviated oxidative stress induced by CPZ and dramatically suppress infammatory biomarkers. (Khalilian et al. 2021).
Noise-induced hearing loss
Constant increased noice exposure is one of the most common causes of hearing loss. It is estimated that approximately 5 % of the world population suffers from noise-induced hearing loss (NIHL) but its management and treatment are only poorly understood. Recent systematic review includes eleven articles with 701 patients and determines the effectiveness of current pharmacologic agents for the prevention of NIHL (Gupta et al. 2021). Various regimens included administration several well-tolerated agents and known supplements such as α-lipoic acid, ambient oxygen, beta-carotene, carbogen, ebselen, Mg-aspartate, N-acetylcysteine, and vitamins C, E, and B12. Unfortunately, there is only limited number of heterogenous studies and future prospective, double-blinded, randomized, placebo-controlled clinical trials with standardized reporting of audiometric data are necessary to evaluate the clinical efficacy of pharmacological prevention for NIHL (Gupta et al. 2021).
Oxidative stress is one of the current theories of NIHL. Fetoni et al. (2013) followed the relationship between cochlear oxidative damage and repeated noise exposure in rat model of adult male Wistar rats. NIHL caused hearing loss, damage in hair cells and spiral ganglion. In addition, NIHL changed dendritic morphology and decreased spine number of pyramidal neurons of auditory cortices. Systemic administration of a hydrophilic CoQ10 formulation (CoQ10 terclaclate – Qter, i.p. 10 mg CoQ10/kg one hour before the acustic trauma) reduced oxidative-induced cochlear damage, hearing loss and cortical dendritic injury.
Hypertension
Hypertension belongs to cardiovascular diseases but the brain plays an essential role in arterial blood pressure regulation. Rostral ventrolateral medulla (RVLM) is a brainstem site that generates sympathetic vasomotor tone and is an important center of blood pressure control (Guynet 2006, Hirooka et al. 2010, Sved et al. 2003). Animal models of neurogenic hypertension such as spontaneously hypertensive rats (SHR) have elevated the levels of superoxide anion and hydrogen peroxide in RVLM (Kimura et al. 2005, Kishi et al. 2004, Konno et al. 2012, Tai et al. 2005). Chan et al. (2009) recognized reduced mitochondrial electron capacity in RVLM of SHR. Local microinjection of CoQ10 into RVLM of SHR restored a reduced mitochondrial electron transport chain capacity. Added CoQ10 lessened the rotenone inhibition of respiratory chain complex I or antimycin A inhibition of respiratory chain complex III. Local application of CoQ10 into RVLM of SHR promoted a dose-dependent decrease in mean arterial pressure and sympathetic vasomotor tone. On the other hand, there was only negligible effect of CoQ10 treatment on superoxide level in RVLM of prehypertensive SHR or normotensive Wistar Kyoto rats (Chan et al. 2009).
The paraventricular nucleus (PVN) in the hypothalamus also plays an important role in the development of hypertension. PVN is an important central site for the coordination and regulation of autonomic response involving several excitatory and inhibitory neurotransmitters and pro- and anti-inflammatory cytokines. Adult male Sprague-Dawley rats fed a high-salt (8 % NaCl) diet for 15 weeks developed hypertension with higher mean arterial pressure as compared to rats fed a normal salt (0.3 % NaCl) diet (Gao et al. 2016). High-salt diet also increased levels of noradrenaline, tyrosine hydroxylase, interleukin-1β, NADPH oxidase 2 (NOX2) and NADPH oxidase 4 (NOX4) and lowered levels of gamma-aminobutyric acid (GABA), interleukin-10 and Cu/Zn superoxide dismutase (SOD) in PVN. The concomitant CoQ10 treatment (10 mg/kg/day via oral gavage in rats fed a high-salt diet) for 15 weeks attenuated salt-induced hypertension. Moreover, CoQ10 supplementation restored the balance between excitatory and inhibitory neurotransmitters and the balance between pro- and anti-inflammatory cytokines in PVN. Thus, Gao et al. (2016) concluded that CoQ10 exerts its protective effects on hypertension just via restoring the appropriated balance in PVN.
Rats fed a high-fructose diet are the well-established rodent model for the study of human metabolic syndrome (Wu et al. 2014). The feeding of a high-fructose diet for 8 weeks caused an increase in sympathetic vasomotor tone and neurogenic hypertension in adult male Sprague-Dawley rats. RVLM of rats fed a high-fructose diet had increased level of reactive oxygen species due to depression of mitochondrial electron transport chain capacity and neuronal NO synthase (nNOS) uncoupling via upregulation of its protein inhibitor. Intracisternal infusion of CoQ10 attenuated sympathoexcitation and hypertension and significantly ameliorated all molecular events in rats fed high-fructose diet.
These three examples of hypertension show the variety of CoQ10 actions – the restoration of mitochondrial respiratory chain (Chan et al. 2009) or the correction of the balance in neurotransmitters and cytokines (Gao et al. 2016).
Hypertension affects approximately one billion subjects worldwide and is a major risk factor associated with cardiovascular events including coronary heart disease and cerebrovascular accidents. Some reviews summarized that CoQ10 administration can lower blood pressure without significant side effects in patients (Ho et al. 2016, Rosenfeldt et al. 2007).
Conclusions
From the first pioneering clinical administration of CoQ10 to patients with heart failure in Japan in the 60s of the last century the number of CoQ10 applications keeps increasing. In the context of mitochondrial dysfunction and oxidative stress in the above-mentioned serious neurological diseases the most prominent and relevant functions are the energetic role and antioxidant capacity of CoQ10. New promising formulations improve bioavailability and could make possible the more efficient administration. Unfortunately, the listed neurological diseases need a more causative treatment. CoQ10 administration can serve only as a corooborative substance. It is important to note that numerous clinical and experimental studies repeteadly provide the evidence that CoQ10 is highly safe and good tolerated with negligible side effects or drug interactions.
Acknowledgements
I thank Dr. Martina Vokurkova for drawing figures, Dr. Anna Avenarius and Dr. Michal Bencze for the data on Myoquinon and Dr. Josef Zicha for critical reading of the manuscript. The study was supported by an international joint project of GACR and MOST, Taiwan (research grant No. 19-08260J) and by the Institute of Physiology, Czech Academy of Sciences (No. RVO 67985823).
Footnotes
This paper is dedicated to the 70th anniversary of the founding of Physiologia Bohemoslovaca (currently Physiological Research)
Conflict of Interest
There is no conflict of interest.
References
- ACAR A, UGUR CEVIK M, EVLIYAOGLU O, UZAR E, TAMAM Y, ARIKANOGLU A, YUCEL Y, VAROL S, ONDER H, TAŞDEMIR N. Evaluation of serum oxidant/antioxidant balance in multiple sclerosis. Acta Neurol Belg. 2012;112:275–280. doi: 10.1007/s13760-012-0059-4. [DOI] [PubMed] [Google Scholar]
- AGMO HERNÁNDEZ V, ERIKSSON EK, EDWARDS K. Ubiquinone-10 alters mechanical properties and increases stability of phospholipid membranes. Biochim Biophys Acta. 2015;1848:2233–2243. doi: 10.1016/j.bbamem.2015.05.002. [DOI] [PubMed] [Google Scholar]
- ALCÁZAR-FABRA M, TREVISSON E, BREA-CALVO G. Clinical syndromes associated with coenzyme Q10 deficiency. Essays Biochem. 2018;62:377–398. doi: 10.1042/EBC20170107. [DOI] [PubMed] [Google Scholar]
- ALZHEIMER’S ASSOCIATION. Alzheimer’s disease facts and figures. Alzheimers Dement. 2013;9:208–245. doi: 10.1016/j.jalz.2013.02.003. [DOI] [PubMed] [Google Scholar]
- ARI C, POFF AM, HELD HE, LANDON CS, GOLDHAGEN CR, MAVROMATES N, D’AGOSTINO DP. Metabolic therapy with Deanna Protocol supplementation delays disease progression and extends survival in amyotrophic lateral sclerosis (ALS) mouse model. PloS One. 2014;9:e103526. doi: 10.1371/journal.pone.0103526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ARMON-OMER A, WALDMAN C, SIMAAN N, NEUMAN H, TAMIR S, SHAHIEN R. New insights on the nutrition status and antioxidant capacity in multiple sclerosis patients. Nutrients. 2019;11:427. doi: 10.3390/nu11020427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ARTUCH R, ARACIL A, MAS A, COLOMÉ C, RISSECH M, MONRÓS E, PINEDA M. Friedreich’s ataxia: idebenone treatment in early stage patients. Neuropediatrics. 2002;33:190–193. doi: 10.1055/s-2002-34494. [DOI] [PubMed] [Google Scholar]
- ATTIA HN, MAKLAD YA. Neuroprotective effects of coenzyme Q10 on paraquat-induced Parkinson’s disease in experimental animals. Behav Pharmacol. 2018;29:79–86. doi: 10.1097/FBP0000000000000342. [DOI] [PubMed] [Google Scholar]
- ATWOOD CS, HUANG X, MOIR RD, TANZI RE, BUSH AI. Role of free radicals and metal ions in the pathogenesis of Alzheimer’s disease. Met Ions Biol Syst. 1999;36:309–364. [PubMed] [Google Scholar]
- AWAD AM, BRADLEY MC, FERNÁNDEZ-DEL-RÍO L, NAG A, TSUI HS, CLARKE CF. Coenzyme Q10 deficiencies: pathways in yeast and humans. Essays Biochem. 2018;62:361–376. doi: 10.1042/EBC20170106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BAGUR MJ, MURCIA MA, JIMÉNEZ-MONREAL AM, TUR JA, BIBILONI MM, ALONSO GL, MARTÍNEZ-TOMÉ M. Influence of diet in multiple sclerosis: A systematic review. Adv Nutr. 2017;8:463–472. doi: 10.3945/an.116.014191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BAHADORANI S, HILLIKER AJ. Antioxidants cannot suppress the lethal phenotype of a Drosophila melanogaster model of Huntington’s disease. Genome. 2008;51:392–395. doi: 10.1139/G08-012. [DOI] [PubMed] [Google Scholar]
- BALOYANNIS SJ. Mitochondrial alterations in Alzheimer’s disease. J Alzheimers Dis. 2006;9:119–126. doi: 10.3233/jad-2006-9204. [DOI] [PubMed] [Google Scholar]
- BALOYANNIS SJ. Dendritic pathology in Alzheimer’s disease. J Neurol Sci. 2009;283:153–157. doi: 10.1016/j.jns.2009.02.370. [DOI] [PubMed] [Google Scholar]
- BARBER SC, SHAW PJ. Oxidative stress in ALS: key role in motor neuron injury and therapeutic target. Free Radic Biol Med. 2010;48:629–641. doi: 10.1016/j.freeradbiomed.2009.11.018. [DOI] [PubMed] [Google Scholar]
- BARNES DE, YAFFE K. The projected effect of risk factor reduction on Alzheimer’s disease prevalence. Lancet Neurol. 2011;10:819–828. doi: 10.1016/S1474-4422(11)70072-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BATTINO M, FERRI E, GORINI A, VILLA RF, RODRIGUEZ HUERTAS JF, FIORELLA P, GENOVA ML, LENAZ G, MARCHETTI M. Natural distribution and occurrence of coenzyme Q homologues. Membr Biochem. 1990;9:179–190. doi: 10.3109/09687689009025839. https://org.doi/10.3109/09687689009025839 . [DOI] [PubMed] [Google Scholar]
- BATTINO M, GORINI A, VILLA RF, GENOVA ML, BOVINA C, SASSI S, LITTARRU GP, LENAZ G. Coenzyme Q content in synaptic and non-synaptic mitochondria from different brain regions in the ageing rat. Mech Ageing Dev. 1995;78:173–187. doi: 10.1016/0047-6374(94)01535-t. https://org.doi:10/1016/0047-6374(94)01535-t . [DOI] [PubMed] [Google Scholar]
- BEAL MF, MATTHEWS RT, TIELEMAN A, SHULTS CW. Coenzyme Q10 attenuates the 1-methyl-4-phenyl-1,2,3,tetrahydropyridine (MPTP) induced loss of striatal dopamine and dopaminergic axons in aged mice. Brain Res. 1998;783:109–114. doi: 10.1016/s0006-8993(97)01192-x. [DOI] [PubMed] [Google Scholar]
- BELOUSOVA M, TOKAREVA OG, GORODETSKAYA E, KALENIKOVA EI, MEDVEDEV OS. Intravenous treatment with coenzyme Q10 improves neurological outcome and reduces infarct volume after transient focal brain ischemia in rats. J Cardiovasc Pharmacol. 2016;67:103–109. doi: 10.1097/FJC.0000000000000320. [DOI] [PubMed] [Google Scholar]
- BERGAMASCO B, SCARZELLA L, LA COMMARE P. Idebenone, a new drug in the treatment of cognitive impairment in patients with dementia of the Alzheimer type. Funct Neurol. 1994;9:161–168. [PubMed] [Google Scholar]
- BEYER RE, BURNETT BA, CARTWRIGHT KJ, EDINGTON DW, FALZON MJ, KREITMAN KR, KUHN TW, RAMP BJ, RHEE SY, ROSENWASSER MJ, STEIN M, AN LC. Tissue coenzyme Q (ubiquinone) and protein concentrations over the life span of the laboratory rat. Mech Ageing Dev. 1985;32:267–281. doi: 10.1016/0047-6374(85)90085-5. [DOI] [PubMed] [Google Scholar]
- BHAGAVAN HN, CHOPRA RK. Coenzyme Q10: absorption, tissue uptake, metabolism and pharmacokinetics. Free Radic Res. 2006;40:445–453. doi: 10.1080/10715760600617843. [DOI] [PubMed] [Google Scholar]
- BHAGAVAN HN, CHOPRA RK. Plasma coenzyme Q10 response to oral ingestion of coenzyme Q10 formulations. Mitochondrion. 2007;7(Suppl):S78–S88. doi: 10.1016/j.mito.2007.03.003. [DOI] [PubMed] [Google Scholar]
- BINDOFF LA, BIRCH-MACHIN MA, CARTLIDGE NE, PARKER WD, JR, TURNBULL DM. Respiratory chain abnormalities in skeletal muscle from patients with Parkinson’s disease. J Neurol Sci. 1991;104:203–208. doi: 10.1016/0022-510x(91)90311-t. [DOI] [PubMed] [Google Scholar]
- BLAKE RL, HALL JG, RUSSELL ES. Mitochondrial proline dehydrogenase deficiency in hyperprolinemic PRO/Re mice: genetic and enzymatic analyses. Biochem Genet. 1976;14:739–757. doi: 10.1007/BF00485338. [DOI] [PubMed] [Google Scholar]
- BOND L, BERNHARDT K, MADRIA P, SORRENTINO K, SCELSI H, MITCHELL CS. A metadata analysis of oxidative stress etiology in preclinical amyotrophic lateral sclerosis: Benefits of antioxidant therapy. Front Neurosci. 2018;12:10. doi: 10.3389/fnins.2018.00010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BONO-YAGÜE J, GÓMEZ-ESCRIBANO AP, MILLÁN JM, VÁZQUEZ-MANRIQUE RP. Reactive species in Huntington disease: are they really the radicals you want to catch? Antioxidants (Basel) 2020;9:577. doi: 10.3390/antiox9070577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BOROWY-BOROWSKI H, SODJA C, DOCHERTY J, WALKER PR, SIKORSKA M. Unique technology for solubilization and delivery of highly lipophilic bioactive molecules. J Drug Target. 2004;12:415–424. doi: 10.1080/10611860412331285233. [DOI] [PubMed] [Google Scholar]
- BRANDSEMA JF, STEPHENS D, HARTLEY J, YOON G. Intermediate-dose idebenone and quality of life in Friedreich ataxia. Pediatr Neurol. 2010;42:338–342. doi: 10.1016/j.pediatrneurol.2010.01.004. [DOI] [PubMed] [Google Scholar]
- BRIÈRE JJ, SCHLEMMER D, CHRETIEN D, RUSTIN P. Quinone analogues regulate mitochondrial substrate competitive oxidation. Biochem Biophys Res Commun. 2004;316:1138–1142. doi: 10.1016/j.bbrc.2004.03.002. [DOI] [PubMed] [Google Scholar]
- BÜRK K. Friedreich Ataxia: current status and future prospects. Cerebellum Ataxias. 2017;4:4. doi: 10.1186/s40673-017-0062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- BUYSE G, MERTENS L, DI SALVO G, MATTHIJS I, WEIDEMANN F, EYSKENS B, GOOSSENS W, GOEMANS N, SUTHERLAND GR, VAN HOVE JL. Idebenone treatment in Friedreich’s ataxia: neurological, cardiac, and biochemical monitoring. Neurology. 2003;60:1679–1681. doi: 10.1212/01.wnl.0000068549.52812.0f. [DOI] [PubMed] [Google Scholar]
- CABONI P, SHERER TB, ZHANG N, TAYLOR G, NA HM, GREENAMYRE JT, CASIDA JE. Rotenone, deguelin, their metabolites, and the rat model of Parkinson’s disease. Chem Res Toxicol. 2004;17:1540–1548. doi: 10.1021/tx049867r. [DOI] [PubMed] [Google Scholar]
- CABRINI L, BARZANTI V, CIPOLLONE M, FIORENTINI D, GROSSI G, TOLOMELLI B, ZAMBONIN L, LANDI L. Antioxidants and total peroxyl radical-trapping ability of olive and seed oils. J Agric Food Chem. 2001;49:6026–6032. doi: 10.1021/jf010837t. [DOI] [PubMed] [Google Scholar]
- CAMPBELL GR, KRAYTSBERG Y, KRISHNAN KJ, OHNO N, ZIABREVA I, REEVE A, TRAPP BD, NEWCOMBE J, REYNOLDS R, LASSMANN H, KHAPKO K, TURNBULL DM, MAHAD DJ. Clonally expanded mitochondrial DNA deletions within the choroid plexus in multiple sclerosis. Acta Neuropathol. 2012;124:209–220. doi: 10.1007/s00401-012-1001-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CANNON JR, TAPIAS V, NA HM, HONICK AS, DROLET RE, GREENAMYRE JT. A highly reproducible rotenone model of Parkinson’s disease. Neurobiol Dis. 2009;34:279–290. doi: 10.1016/j.nbd.2009.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CAPALDI RA. Arrangement of proteins in the mitochondrial inner membrane. Biochim Biophys Acta. 1982;694:291–306. doi: 10.1016/0304-4157(82)90009-0. [DOI] [PubMed] [Google Scholar]
- CHAN SH, WU KL, CHANG AY, TAI MH, CHAN JY. Oxidative impairment of mitochondrial electron transport chain complexes in rostral ventrolateral medulla contributes to neurogenic hypertension. Hypertension. 2009;53:217–227. doi: 10.1161/HYPERTENSIONAHA.108.116905. [DOI] [PubMed] [Google Scholar]
- CHOI IY, LEE P, ADANY P, HUGHES AJ, BELLISTON S, DENNEY DR, LYNCH SG. In vivo evidence of oxidative stress in brains of patients with progressive multiple sclerosis. Mult Scler. 2018;24:1029–1038. doi: 10.1177/1352458517711568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- CICCHETTI F, LAPOINTE N, ROBERGE-TREMBLAY A, SAINT-PIERRE M, JIMENEZ L, FICKE BW, GROSS RE. Systemic exposure to paraquat and maneb models early Parkinson’s disease in young adult rats. Neurobiol Dis. 2005;20:360–371. doi: 10.1016/j.nbd.2005.03.018. [DOI] [PubMed] [Google Scholar]
- CLEREN C, YANG L, LORENZO B, CALINGASAN NY, SCHOMER A, SIRECI A, WILLE EJ, BEAL MF. Therapeutic effects of coenzyme Q10 (CoQ10) and reduced CoQ10 in the MPTP model of Parkinsonism. J Neurochem. 2008;104:1613–1621. doi: 10.1111/j.1471-4159.2007.05097.x. [DOI] [PubMed] [Google Scholar]
- COOPER JM, KORLIPARA LV, HART PE, BRADLEY JL, SCHAPIRA AH. Coenzyme Q10 and vitamin E deficiency in Friedreich’s ataxia: predictor of efficacy of vitamin E and coenzyme Q10 therapy. Eur J Neurol. 2008;15:1371–1379. doi: 10.1111/j.1468-1331.2008.02318.x. [DOI] [PubMed] [Google Scholar]
- CRANE FL. Comments on the discovery of coenzyme Q: a commentary on ‘Isolation of a quinone from beef heart mitochondria’. Biochim Biophys Acta. 1989;1000:358–361. doi: 10.1016/s0006-3002(89)80030-7. [DOI] [PubMed] [Google Scholar]
- CRANE FL, HATEFI Y, LESTER RL, WIDMER C. Isolation of a quinone from beef heart mitochondria. Biochim Biophys Acta. 1957;25:220–221. doi: 10.1016/0006-3002(57)90457-2. [DOI] [PubMed] [Google Scholar]
- DAHRI M, TARIGHAT-ESFANJANI A, ASGHARI-JAFARABADI M, HASHEMILAR M. Oral coenzyme Q10 supplementation in patients with migraine: Effects on clinical features and inflammatory markers. Nutr Neurosci. 2019;22:607–615. doi: 10.1080/1028415X.2017.1421039. [DOI] [PubMed] [Google Scholar]
- DE BUSTOS F, JIMÉNEZ-JIMÉNEZ FJ, MOLINA JA, GÓMEZ-ESCALONILLA C, DE ANDRÉS C, DEL HOYO P, ZURDO M, TALLÓN-BARRANCO A, BERBEL A, PORTA-ETESSAM J, PARRILLA G, ARENAS J. Serum levels of coenzyme Q10 in patients with multiple sclerosis. Acta Neurol Scand. 2000;101:209–211. doi: 10.1034/j.1600-0404.2000.101003209.x. [DOI] [PubMed] [Google Scholar]
- DE BUSTOS F, MOLINA JA, JIMÉNEZ-JIMÉNEZ FJ, GARCÍA-REDONDO A, GÓMEZ-ESCALONILLA C, PORTA-ETESSAM J, BERBEL A, ZURDO M, BARCENILLA B, PARRILLA G, ENRIQUEZ-DE-SALAMANCA R, ARENAS J. Serum levels of coenzyme Q10 in patients with Alzheimer’s disease. J Neural Transm (Vienna) 2000;107:233–239. doi: 10.1007/s007020050019. [DOI] [PubMed] [Google Scholar]
- DESBATS MA, LUNARDI G, DOIMO M, TREVISSON E, SALVIATI L. Genetic bases and clinical manifestations of coenzyme Q10 (CoQ 10) deficiency. J Inherit Metab Dis. 2015;38:145–156. doi: 10.1007/s10545-014-9749-9. [DOI] [PubMed] [Google Scholar]
- DI MARCO LY, VENNERI A, FARKAS E, EVANS PC, MARZO A, FRANGI AF. Vascular dysfunction in the pathogenesis of Alzheimer’s disease--A review of endothelium-mediated mechanisms and ensuing vicious circles. Neurobiol Dis. 2015;82:593–606. doi: 10.1016/j.nbd.2015.08.014. [DOI] [PubMed] [Google Scholar]
- DI PROSPERO NA, BAKER A, JEFFRIES N, FISCHBECK KH. Neurological effects of high-dose idebenone in patients with Friedreich’s ataxia: a randomised, placebo-controlled trial. Lancet Neurol. 2007;6:878–886. doi: 10.1016/S1474-4422(07)70220-X. [DOI] [PubMed] [Google Scholar]
- DUBERLEY KE, HEALES SJ, ABRAMOV AY, CHALASANI A, LAND JM, RAHMAN S, HARGREAVES IP. Effect of coenzyme Q10 supplementation on mitochondrial electron transport chain activity and mitochondrial oxidative stress in coenzyme Q10 deficient human neuronal cells. Int J Biochem Cell Biol. 2014;50:60–63. doi: 10.1016/j.biocel.2014.02.003. [DOI] [PubMed] [Google Scholar]
- DUMONT M, KIPIANI K, YU F, WILLE E, KATZ M, CALINGASAN NY, GOURAS GK, LIN MT, BEAL MF. Coenzyme Q10 decreases amyloid pathology and improves behavior in a transgenic mouse model of Alzheimer’s disease. J Alzheimers Dis. 2011;27:211–223. doi: 10.3233/JAD-2011-110209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ECHTAY KS, WINKLER E, KLINGENBERG M. Coenzyme Q is an obligatory cofactor for uncoupling protein function. Nature. 2000;408(6812):609–613. doi: 10.1038/35046114. [DOI] [PubMed] [Google Scholar]
- EHRENHAUS MASOTTA N, HÖCHT C, CONTIN M, LUCANGIOLI S, ROJAS AM, TRIPODI VP. Bioavailability of coenzyme Q10 loaded in an oleogel formulation for oral therapy: Comparison with a commercial-grade solid formulation. Int J Pharm. 2020;582:119315. doi: 10.1016/j.ijpharm.2020.119315. [DOI] [PubMed] [Google Scholar]
- ELIPENAHLI C, STACK C, JAINUDDIN S, GERGES M, YANG L, STARKOV A, BEAL MF, DUMONT M. Behavioral improvement after chronic administration of coenzyme Q10 in P301S transgenic mice. J Alzheimers Dis. 2012;28:173–182. doi: 10.3233/JAD-2011-111190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ELMBERGER PG, KALÈN A, APPELKVIST EL, DALLNER G. In vitro and in vivo synthesis of dolichol and other main mevalonate products in various organs of the rat. Eur J Biochem. 1987;168:1–11. doi: 10.1111/j.1432-1033.1987.tb13379.x. [DOI] [PubMed] [Google Scholar]
- ESPOSTI MD, NGO A, GHELLI A, BENELLI B, CARELLI V, MCLENNAN H, LINNANE AW. The interaction of Q analogs, particularly hydroxydecyl benzoquinone (idebenone), with the respiratory complexes of heart mitochondria. Arch Biochem Biophys. 1996;330:395–400. doi: 10.1006/abbi.1996.0267. https://doi.org10.1006/abbi.1996.0267 . [DOI] [PubMed] [Google Scholar]
- ESSA MM, MOGHADAS M, BA-OMAR T, WALID QORONFLEH M, GUILLEMIN GJ, MANIVASAGAM T, JUSTIN-THENMOZHI A, RAY B, BHAT A, CHIDAMBARAM SB, FERNANDES AJ, SONG BJ, AKBAR M. Protective effects of antioxidants in Huntington’s disease: an extensive review. Neurotox Res. 2019;35:739–774. doi: 10.1007/s12640-018-9989-9. https://doi,org/10.1007/s12640-018-9989-9 . [DOI] [PubMed] [Google Scholar]
- EVANS DR, GUY HI. Mammalian pyrimidine biosynthesis: fresh insights into an ancient pathway. J Biol Chem. 2004;279:33035–33038. doi: 10.1074/jbc.R400007200. [DOI] [PubMed] [Google Scholar]
- EVANS E, PICCIO L, CROSS AH. Use of vitamins and dietary supplements by patients with multiple sclerosis: A review [published correction appears in JAMA Neurol 75: 1028, 2018] JAMA Neurol. 2018;75:1013–1021. doi: 10.1001/jamaneurol.2018.0611. [DOI] [PubMed] [Google Scholar]
- FALKENBURGER BH, SARIDAKI T, DINTER E. Cellular models for Parkinson’s disease. J Neurochem. 2016;139(Suppl 1):121–130. doi: 10.1111/jnc.13618. [DOI] [PubMed] [Google Scholar]
- FAN L, FENG Y, CHEN GC, QIN LQ, FU CL, CHEN LH. Effects of coenzyme Q10 supplementation on inflammatory markers: A systematic review and meta-analysis of randomized controlled trials. Pharmacol Res. 2017;119:128–136. doi: 10.1016/j.phrs.2017.01.032. [DOI] [PubMed] [Google Scholar]
- FATO R, BERTOLI E, PARENTI CASTELLI G, LENAZ G. Fluidizing effect of endogenous ubiquinone in bovine heart mitochondrial membranes. FEBS Lett. 1984;172:6–10. doi: 10.1016/0014-5793(84)80861-3. [DOI] [PubMed] [Google Scholar]
- FERRANTE RJ, ANDREASSEN OA, DEDEOGLU A, FERRANTE KL, JENKINS BG, HERSCH SM, BEAL MF. Therapeutic effects of coenzyme Q10 and remacemide in transgenic mouse models of Huntington’s disease. J Neurosci. 2002;22:1592–1599. doi: 10.1523/JNEUROSCI.22-05-01592.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- FERRANTE KL, SHEFNER J, ZHANG H, BETENSKY R, O’BRIEN M, YU H, FANTASIA M, TAFT J, BEAL MF, TRAYNOR B, NEWHALL K, DONOFRIO P, CARESS J, ASHBURN C, FREIBERG B, O’NEILL C, PALADENECH C, WALKER T, PESTRONK A, ABRAMS B, FLORENCE J, RENNA R, SCHIERBECKER J, MALKUS B, CUDKOWICZ M. Tolerance of high-dose (3,000 mg/day) coenzyme Q10 in ALS. Neurology. 2005;65:1834–1836. doi: 10.1212/01.wnl.0000187070.35365.d7. [DOI] [PubMed] [Google Scholar]
- FETONI AR, DE BARTOLO P, ERAMO SL, ROLESI R, PACIELLO F, BERGAMINI C, FATO R, PALUDETTI G, PETROSINI L, TROIANI D. Noise-induced hearing loss (NIHL) as a target of oxidative stress-mediated damage: cochlear and cortical responses after an increase in antioxidant defense. J Neurosci. 2013;33:4011–4023. doi: 10.1523/JNEUROSCI.2282-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- FIŠAR Z, HANSÍKOVÁ H, KŘÍŽOVÁ J, JIRÁK R, KITZLEROVÁ E, ZVĚŘOVÁ M, HROUDOVÁ J, WENCHICH L, ZEMAN J, RABOCH J. Activities of mitochondrial respiratory chain complexes in platelets of patients with Alzheimer’s disease and depressive disorder. Mitochondrion. 2019;48:67–77. doi: 10.1016/j.mito.2019.07.013. [DOI] [PubMed] [Google Scholar]
- FONTAINE E, ICHAS F, BERNARDI P. A ubiquinone-binding site regulates the mitochondrial permeability transition pore. J Biol Chem. 1998;273:25734–25740. doi: 10.1074/jbc.273.40.25734. [DOI] [PubMed] [Google Scholar]
- FRERMAN FE. Acyl-CoA dehydrogenases, electron transfer flavoprotein and electron transfer flavoprotein dehydrogenase. Biochem Soc Trans. 1988;16:416–418. doi: 10.1042/bst0160416. [DOI] [PubMed] [Google Scholar]
- GALASKO DR, PESKIND E, CLARK CM, QUINN JF, RINGMAN JM, JICHA GA, COTMAN C, COTTRELL B, MONTINE TJ, THOMAS RG, AISEN P Alzheimer’s Disease Cooperative Study. Antioxidants for Alzheimer disease: a randomized clinical trial with cerebrospinal fluid biomarker measures. Arch Neurol. 2012;69:836–841. doi: 10.1001/archneurol.2012.85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GAO HL, YU XJ, QI J, YI QY, JING WH, SUN WY, CUI W, MU JJ, YUAN ZY, ZHAO XF, LIU KL, ZHU GQ, SHI XL, LIU JJ, KANG YM. Oral CoQ10 attenuates high salt-induced hypertension by restoring neurotransmitters and cytokines in the hypothalamic paraventricular nucleus. Sci Rep. 2016;6:30301. doi: 10.1038/srep30301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GAUL C, DIENER HC, DANESCH U MIGRAVENT® STUDY GROUP. Improvement of migraine symptoms with a proprietary supplement containing riboflavin, magnesium and Q10: a randomized, placebo-controlled, double-blind, multicenter trial. J Headache Pain. 2015;16:516. doi: 10.1186/s10194-015-0516-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GENOVA ML, LENAZ G. New developments on the functions of coenzyme Q in mitochondria. Biofactors. 2011;37:330–354. doi: 10.1002/biof.168. [DOI] [PubMed] [Google Scholar]
- GENOVA ML, LENAZ G. Functional role of mitochondrial respiratory supercomplexes. Biochim Biophys Acta. 2014;1837:427–443. doi: 10.1016/j.bbabio.2013.11.002. [DOI] [PubMed] [Google Scholar]
- GIAVAROTTI L, SIMON KA, AZZALIS LA, FONSECA FL, LIMA AF, FREITAS MC, BRUNIALTI MK, SALOMÃO R, MOSCARDI AA, MONTAÑO MB, RAMOS LR, JUNQUEIRA VB. Mild systemic oxidative stress in the subclinical stage of Alzheimer’s disease. Oxid Med Cell Longev. 2013;2013:609019. doi: 10.1155/2013/609019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GILLE L, NOHL H. The existence of a lysosomal redox chain and the role of ubiquinone. Arch Biochem Biophys. 2000;375:347–354. doi: 10.1006/abbi.1999.1649. [DOI] [PubMed] [Google Scholar]
- GIRONI M, BORGIANI B, MARIANI E, CURSANO C, MENDOZZI L, CAVARRETTA R, SARESELLA M, CLERICI M, COMI G, ROVARIS M, FURLAN R. Oxidative stress is differentially present in multiple sclerosis courses, early evident, and unrelated to treatment. J Immunol Res. 2014;2014:961863. doi: 10.1155/2014/961863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GUEVEN N, RAVISHANKAR P, ERI R, RYBALKA E. Idebenone: When an antioxidant is not an antioxidant. Redox Biol. 2021;38:101812. doi: 10.1016/j.redox.2020.101812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GUEVEN N, WOOLLEY K, SMITH J. Border between natural product and drug: comparison of the related benzoquinones idebenone and coenzyme Q10. Redox Biol. 2015;4:289–295. doi: 10.1016/j.redox.2015.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GUILBOT A, BANGRATZ M, AIT ABDELLAH S, LUCAS C. A combination of coenzyme Q10, feverfew and magnesium for migraine prophylaxis: a prospective observational study. BMC Complement Altern Med. 2017;17:433. doi: 10.1186/s12906-017-1933-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GUPTA A, KOOCHAKZADEH S, NGUYEN SA, BRENNAN EA, MEYER TA, LAMBERT PR. Pharmacological prevention of noise-induced hearing loss: A systematic review. Otol Neurotol. 2021;42:2–9. doi: 10.1097/MAO.0000000000002858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- GUTZMANN H, HADLER D. Sustained efficacy and safety of idebenone in the treatment of Alzheimer’s disease: update on a 2-year double-blind multicentre study. J Neural Transm Suppl. 1998;54:301–310. doi: 10.1007/978-3-7091-7508-8_30. [DOI] [PubMed] [Google Scholar]
- GUYENET PG. The sympathetic control of blood pressure. Nat Rev Neurosci. 2006;7:335–346. doi: 10.1038/nrn1902. [DOI] [PubMed] [Google Scholar]
- HAAS RH, NASIRIAN F, NAKANO K, WARD D, PAY M, HILL R, SHULTS CW. Low platelet mitochondrial complex I and complex II/III activity in early untreated Parkinson’s disease. Ann Neurol. 1995;37:714–722. doi: 10.1002/ana.410370604. [DOI] [PubMed] [Google Scholar]
- HAIDER L, FISCHER MT, FRISCHER JM, BAUER J, HÖFTBERGER R, BOTOND G, ESTERBAUER H, BINDER CJ, WITZTUM JL, LASSMANN H. Oxidative damage in multiple sclerosis lesions. Brain. 2011;134:1914–1924. doi: 10.1093/brain/awr128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- HAJIHASHEMI P, ASKARI G, KHORVASH F, REZA MARACY M, NOURIAN M. The effects of concurrent coenzyme Q10, L-carnitine supplementation in migraine prophylaxis: A randomized, placebo-controlled, double-blind trial. Cephalalgia. 2019;39:648–654. doi: 10.1177/0333102418821661. [DOI] [PubMed] [Google Scholar]
- HAMADJIDA A, FROUNI I, KWAN C, HUOT P. Classic animal models of Parkinson’s disease: a historical perspective. Behav Pharmacol. 2019;30:291–310. doi: 10.1097/FBP.0000000000000441. [DOI] [PubMed] [Google Scholar]
- HARGREAVES IP, LANE A, SLEIMAN PM. The coenzyme Q10 status of the brain regions of Parkinson’s disease patients. Neurosci Lett. 2008;447:17–19. doi: 10.1016/j.neulet.2008.09.069. [DOI] [PubMed] [Google Scholar]
- HART PE, LODI R, RAJAGOPALAN B, BRADLEY JL, CRILLEY JG, TURNER C, BLAMIRE AM, MANNERS D, STYLES P, SCHAPIRA AH, COOPER JM. Antioxidant treatment of patients with Friedreich ataxia: four-year follow-up. Arch Neurol. 2005;62:621–626. doi: 10.1001/archneur.62.4.621. [DOI] [PubMed] [Google Scholar]
- HATHCOCK JN, SHAO A. Risk assessment for coenzyme Q10 (Ubiquinone) Regul Toxicol Pharmacol. 2006;45:282–288. doi: 10.1016/j.yrtph.2006.05.006. [DOI] [PubMed] [Google Scholar]
- HERSHEY AD, POWERS SW, VOCKELL AL, LECATES SL, ELLINOR PL, SEGERS A, BURDINE D, MANNING P, KABBOUCHE MA. Coenzyme Q10 deficiency and response to supplementation in pediatric and adolescent migraine. Headache. 2007;47:73–80. doi: 10.1111/j.1526-4610.2007.00652.x. [DOI] [PubMed] [Google Scholar]
- HICKEY MA, ZHU C, MEDVEDEVA V, FRANICH NR, LEVINE MS, CHESSELET MF. Evidence for behavioral benefits of early dietary supplementation with CoEnzymeQ10 in a slowly progressing mouse model of Huntington’s disease. Mol Cell Neurosci. 2012;49:149–157. doi: 10.1016/j.mcn.2011.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- HIDAKA T, FUJII K, FUNAHASHI I, FUKUTOMI N, HOSOE K. Safety assessment of coenzyme Q10 (CoQ10) Biofactors. 2008;32:199–208. doi: 10.1002/biof.5520320124. [DOI] [PubMed] [Google Scholar]
- HIROOKA Y, SAGARA Y, KISHI T, SUNAGAWA K. Oxidative stress and central cardiovascular regulation. Pathogenesis of hypertension and therapeutic aspects. Circ J. 2010;74:827–835. doi: 10.1253/circj.cj-10-0153. [DOI] [PubMed] [Google Scholar]
- HO MJ, LI EC, WRIGHT JM. Blood pressure lowering efficacy of coenzyme Q10 for primary hypertension. Cochrane Database Syst Rev. 2016;3:CD007435. doi: 10.1002/14651858.CD007435.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- HUNTINGTON STUDY GROUP. A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington’s disease. Neurology. 2001;57:397–404. doi: 10.1212/wnl.57.3.397. [DOI] [PubMed] [Google Scholar]
- HUNTINGTON STUDY GROUP PRE2CARE INVESTIGATORS. HYSON HC, KIEBURTZ K, ET AL. Safety and tolerability of high-dosage coenzyme Q10 in Huntington’s disease and healthy subjects. Mov Disord. 2010;25:1924–1928. doi: 10.1002/mds.22408. [DOI] [PubMed] [Google Scholar]
- IKEMATSU H, NAKAMURA K, HARASHIMA S, FUJII K, FUKUTOMI N. Safety assessment of coenzyme Q10 (Kaneka Q10) in healthy subjects: a double-blind, randomized, placebo-controlled trial. Regul Toxicol Pharmacol. 2006;44:212–228. doi: 10.1016/j.yrtph.2005.12.002. [DOI] [PubMed] [Google Scholar]
- JABER S, POLSTER BM. Idebenone and neuroprotection: antioxidant, pro-oxidant, or electron carrier? J Bioenerg Biomembr. 2015;47:111–118. doi: 10.1007/s10863-014-9571-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- JACKSON-LEWIS V, PRZEDBORSKI S. Protocol for the MPTP mouse model of Parkinson’s disease. Nat Protoc. 2007;2:141–151. doi: 10.1038/nprot.2006.342. [DOI] [PubMed] [Google Scholar]
- JAMES AM, COCHEMÉ HM, SMITH RA, MURPHY MP. Interactions of mitochondria-targeted and untargeted ubiquinones with the mitochondrial respiratory chain and reactive oxygen species. Implications for the use of exogenous ubiquinones as therapies and experimental tools. J Biol Chem. 2005;280:21295–21312. doi: 10.1074/jbc.M501527200. [DOI] [PubMed] [Google Scholar]
- JIMÉNEZ-JIMÉNEZ FJ, MOLINA JA, DE BUSTOS F, GARCÍA-REDONDO A, GÓMEZ-ESCALONILLA C, MARTÍNEZ-SALIO A, BERBEL A, CAMACHO A, ZURDO M, BARCENILLA B, ENRÍQUEZ DE SALAMANCA R, ARENAS J. Serum levels of coenzyme Q10 in patients with Parkinson’s disease. J Neural Transm (Vienna) 2000;107:177–181. doi: 10.1007/s007020050015. [DOI] [PubMed] [Google Scholar]
- JOSHI G, BEKIER ME, 2ND, WANG Y. Golgi fragmentation in Alzheimer’s disease. Front Neurosci. 2015;9:340. doi: 10.3389/fnins.2015.00340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KAGAN T, DAVIS C, LIN L, ZAKERI Z. Coenzyme Q10 can in some circumstances block apoptosis, and this effect is mediated through mitochondria. Ann N Y Acad Sci. 1999;887:31–47. doi: 10.1111/j.1749-6632.1999.tb07920.x. [DOI] [PubMed] [Google Scholar]
- KALÉN A, APPELKVIST EL, DALLNER G. Age-related changes in the lipid compositions of rat and human tissues. Lipids. 1989;24:579–584. doi: 10.1007/BF02535072. [DOI] [PubMed] [Google Scholar]
- KAMEI M, FUJITA T, KANBE T, SASAKI K, OSHIBA K, OTANI S, MATSUI-YUASA I, MORISAWA S. The distribution and content of ubiquinone in foods. Int J Vit Nutr Res. 1986;56:57–63. [PubMed] [Google Scholar]
- KAMZALOV S, SUMIEN N, FORSTER MJ, SOHAL RS. Coenzyme Q intake elevates the mitochondrial and tissue levels of coenzyme Q and alpha-tocopherol in young mice. J Nutr. 2003;133:3175–3180. doi: 10.1093/jn/133.10.3175. [DOI] [PubMed] [Google Scholar]
- KASPAROVÁ S, SUMBALOVÁ Z, BYSTRICKÝ P, KUCHARSKÁ J, LIPTAJ T, MLYNÁRIK V, GVOZDJÁKOVÁ A. Effect of coenzyme Q10 and vitamin E on brain energy metabolism in the animal model of Huntington’s disease. Neurochem Int. 2006;48:93–99. doi: 10.1016/j.neuint.2005.09.002. [DOI] [PubMed] [Google Scholar]
- KAUFMANN P, THOMPSON JL, LEVY G, BUCHSBAUM R, SHEFNER J, KRIVICKAS LS, KATZ J, ROLLINS Y, BAROHN RJ, JACKSON CE, TIRYAKI E, LOMEN-HOERTH C, ARMON C, TANDAN R, RUDNICKI SA, REZANIA K, SUFIT R, PIORO EP, MONTES J, ARBING R, VECCHIO D, BARSDORF A, MITSUMOTO H, LEVIN B QALS STUDY GROUP. Phase II trial of CoQ10 for ALS finds insufficient evidence to justify phase III. Ann Neurol. 2009;66:235–244. doi: 10.1002/ana.21743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KETTAWAN A, KUNTHIDA C, TAKAHASHI T, KISHI T, CHIKAZAWA J, SAKATA Y, YANO E, WATABE K, YAMAMOTO Y, OKAMOTO T. The quality control assessment of commercially available coenzyme Q10-containing dietary and health supplements in Japan. J Clin Biochem Nutr. 2007;41:124–131. doi: 10.3164/jcbn.2007017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KHALILIAN B, MADADI S, FATTAHI N, ABOUHAMZEH B. Coenzyme Q10 enhances remyelination and regulate inflammation effects of cuprizone in corpus callosum of chronic model of multiple sclerosis. J Mol Histol. 2021;52:125–134. doi: 10.1007/s10735-020-09929-x. [DOI] [PubMed] [Google Scholar]
- KIMURA Y, HIROOKA Y, SAGARA Y, ITO K, KISHI T, SHIMOKAWA H, TAKESHITA A, SUNAGAWA K. Overexpression of inducible nitric oxide synthase in rostral ventrolateral medulla causes hypertension and sympathoexcitation via an increase in oxidative stress. Circ Res. 2005;96:252–260. doi: 10.1161/01.RES.0000152965.75127.9d. [DOI] [PubMed] [Google Scholar]
- KISHI T, HIROOKA Y, KIMURA Y, ITO K, SHIMOKAWA H, TAKESHITA A. Increased reactive oxygen species in rostral ventrolateral medulla contribute to neural mechanisms of hypertension in stroke-prone spontaneously hypertensive rats. Circulation. 2004;109:2357–2362. doi: 10.1161/01.CIR.0000128695.49900.12. [DOI] [PubMed] [Google Scholar]
- KITANO M, WATANABE D, ODA S, KUBO H, KISHIDA H, FUJII K, KITAHARA M, HOSOE K. Subchronic oral toxicity of ubiquinol in rats and dogs. Int J Toxicol. 2008;27:189–215. doi: 10.1080/10915810801978060. [DOI] [PubMed] [Google Scholar]
- KONNO S, HIROOKA Y, KISHI T, SUNAGAWA K. Sympathoinhibitory effects of telmisartan through the reduction of oxidative stress in the rostral ventrolateral medulla of obesity-induced hypertensive rats. J Hypertens. 2012;30:1992–1999. doi: 10.1097/HJH.0b013e328357fa98. [DOI] [PubMed] [Google Scholar]
- KOROSHETZ WJ, JENKINS BG, ROSEN BR, BEAL MF. Energy metabolism defects in Huntington’s disease and effects of coenzyme Q10. Ann Neurol. 1997;41:160–165. doi: 10.1002/ana.410410206. [DOI] [PubMed] [Google Scholar]
- KRIGE D, CARROLL MT, COOPER JM, MARSDEN CD, SCHAPIRA AH. Platelet mitochondrial function in Parkinson’s disease. The Royal Kings and Queens Parkinson Disease Research Group. Ann Neurol. 1992;32:782–788. doi: 10.1002/ana.410320612. [DOI] [PubMed] [Google Scholar]
- KUBO H, FUJII K, KAWABE T, MATSUMOTO S, KISHIDA H, HOSOE K. Food content of ubiquinol-10 and ubiquinone-10 in the Japanese diet. J Food Compost Anal. 2008;21:199–210. [Google Scholar]
- KUMAR A, KUMAR V, SINGH K, KUMAR S, KIM YS, LEE YM, KIM JJ. Therapeutic advances for Huntington’s disease. Brain Sci. 2020;10:43. doi: 10.3390/brainsci10010043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- KWONG LK, KAMZALOV S, REBRIN I, BAYNE AC, JANA CK, MORRIS P, FORSTER MJ, SOHAL RS. Effects of coenzyme Q(10) administration on its tissue concentrations, mitochondrial oxidant generation, and oxidative stress in the rat. Free Radic Biol Med. 2002;33:627–638. doi: 10.1016/s0891-5849(02)00916-4. [DOI] [PubMed] [Google Scholar]
- LASS A, FORSTER MJ, SOHAL RS. Effects of coenzyme Q10 and alpha-tocopherol administration on their tissue levels in the mouse: elevation of mitochondrial alpha-tocopherol by coenzyme Q10. Free Radic Biol Med. 1999;26:1375–1382. doi: 10.1016/s0891-5849(98)00330-x. [DOI] [PubMed] [Google Scholar]
- LEVY G, KAUFMANN P, BUCHSBAUM R, MONTES J, BARSDORF A, ARBING R, BATTISTA V, ZHOU X, MITSUMOTO H, LEVIN B, THOMPSON JLP. A two-stage design for a phase II clinical trial of coenzyme Q10 in ALS. Neurology. 2006;66:660–663. doi: 10.1212/01.wnl.0000201182.60750.66. [DOI] [PubMed] [Google Scholar]
- LI X, ZHAN J, HOU Y, CHEN S, HOU Y, XIAO Z, LUO D, LIN D. Coenzyme Q10 suppresses oxidative stress and apoptosis via activating the Nrf-2/NQO-1 and NF-κB signaling pathway after spinal cord injury in rats. Am J Transl Res. 2019;11:6544–6552. [PMC free article] [PubMed] [Google Scholar]
- LIN P, LIU J, REN M, JI K, LI L, ZHANG B, GONG Y, YAN C. Idebenone protects against oxidized low density lipoprotein induced mitochondrial dysfunction in vascular endothelial cells via GSK3β/β-catenin signalling pathways. Biochem Biophys Res Commun. 2015;465:548–555. doi: 10.1016/j.bbrc.2015.08.058. [DOI] [PubMed] [Google Scholar]
- LINNANE AW, KIOS M, VITETTA L. Coenzyme Q(10)--its role as a prooxidant in the formation of superoxide anion/hydrogen peroxide and the regulation of the metabolome. Mitochondrion. 2007;7(Suppl):S51–S61. doi: 10.1016/j.mito.2007.03.005. [DOI] [PubMed] [Google Scholar]
- LIU ZX, ARTMANN C. Relative bioavailability comparison of different coenzyme Q10 formulations with a novel delivery system. Altern Ther Health Med. 2009;15:42–46. [PubMed] [Google Scholar]
- LODI R, HART PE, RAJAGOPALAN B, TAYLOR DJ, CRILLEY JG, BRADLEY JL, BLAMIRE AM, MANNERS D, STYLES P, SCHAPIRA AH, COOPER JM. Antioxidant treatment improves in vivo cardiac and skeletal muscle bioenergetics in patients with Friedreich’s ataxia. Ann Neurol. 2001;49:590–596. [PubMed] [Google Scholar]
- LUCCHETTI J, MARINO M, PAPA S, TORTAROLO M, GUISO G, POZZI S, BONETTO V, CACCIA S, BEGHI E, BENDOTTI C, GOBBI M. A mouse model of familial ALS has increased CNS levels of endogenous ubiquinol9/10 and does not benefit from exogenous administration of ubiquinol10. PLoS One. 2013;8:e69540. doi: 10.1371/journal.pone.0069540. Published 2013 Jul 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- LYNCH DR, PERLMAN SL, MEIER T. A phase 3, double-blind, placebo-controlled trial of idebenone in friedreich ataxia. Arch Neurol. 2010;67:941–947. doi: 10.1001/archneurol.2010.168. [DOI] [PubMed] [Google Scholar]
- MANCZAK M, MAO P, CALKINS MJ, CORNEA A, REDDY AP, MURPHY MP, SZETO HH, PARK B, REDDY PH. Mitochondria-targeted antioxidants protect against amyloid-beta toxicity in Alzheimer’s disease neurons. J Alzheimers Dis. 2010;20(Suppl 2):S609–S631. doi: 10.3233/JAD-2010-100564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MANTLE D, DYBRING A. Bioavailability of coenzyme Q10: An overview of the absorption process and subsequent metabolism. Antioxidants (Basel) 2020;9:386. doi: 10.3390/antiox9050386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MANYEVITCH R, PROTAS M, SCARPIELLO S, DELISO M, BASS B, NANAJIAN A, CHANG M, THOMPSON SM, KHOURY N, GONNELLA R, TROTZ M, MOORE DB, HARMS E, PERRY G, CLUNES L, ORTIZ A, FRIEDRICH JO, MURRAY IVJ. Evaluation of metabolic and synaptic dysfunction hypotheses of Alzheimer’s disease (AD): A meta-analysis of CSF markers. Curr Alzheimer Res. 2018;15:164–181. doi: 10.2174/1567205014666170921122458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MAO P, MANCZAK M, SHIRENDEB UP, REDDY PH. MitoQ, a mitochondria-targeted antioxidant, delays disease progression and alleviates pathogenesis in an experimental autoimmune encephalomyelitis mouse model of multiple sclerosis. Biochim Biophys Acta. 2013;1832:2322–2331. doi: 10.1016/j.bbadis.2013.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MARIOTTI C, SOLARI A, TORTA D, MARANO L, FIORENTINI C, DI DONATO S. Idebenone treatment in Friedreich patients: one-year-long randomized placebo-controlled trial. Neurology. 2003;60:1676–1679. doi: 10.1212/01.wnl.0000055872.50364.fc. [DOI] [PubMed] [Google Scholar]
- MARX W, HOCKEY M, MCGUINNESS AJ, LANE M, CHRISTODOULOU J, VAN DER MEI I, BERK M, DEAN OM, TAYLOR B, PONSONBY A-L, THE RELIEF TEAM. The effect of emerging nutraceutical interventions for clinical and biological outcomes in multiple sclerosis: A systematic review. Mult Scler Relat Disord. 2020;37:101486. doi: 10.1016/j.msard.2019.101486. [DOI] [PubMed] [Google Scholar]
- MATSUBARA T, AZUMA T, YOSHIDA S, YAMAGAMI T. Serum coenzyme Q10 level in Parkinson syndrome. In: Folkers K, Littarru GP, Yamagami T, editors. Biomedical and clinical aspects of coenzyme Q10. New York: Elsevier Science; 1991. pp. 159–166. [Google Scholar]
- MATTHEWS RT, YANG L, BROWNE S, BAIK M, BEAL MF. Coenzyme Q10 administration increases brain mitochondrial concentrations and exerts neuroprotective effects. Proc Natl Acad Sci U S A. 1998;95:8892–8897. doi: 10.1073/pnas.95.15.8892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MATTILA P, KUMPULAINEN J. Coenzymes Q9 and Q10: Contents in foods and dietary intake. J Food Compost Anal. 2001;14:409–417. doi: 10.1006/jfca.2000.0983. [DOI] [Google Scholar]
- MAUSKOP A. Nonmedication, alternative, and complementary treatments for migraine. Continuum (Minneap Minn) 2012;18:796–806. doi: 10.1212/01.CON.0000418643.24408.40. [DOI] [PubMed] [Google Scholar]
- McCARTHY S, SOMAYAJULU M, SIKORSKA M, BOROWY-BOROWSKI H, PANDEY S. Paraquat induces oxidative stress and neuronal cell death; neuroprotection by water-soluble coenzyme Q10. Toxicol Appl Pharmacol. 2004;201:21–31. doi: 10.1016/j.taap.2004.04.019. [DOI] [PubMed] [Google Scholar]
- McCOY MK, COOKSON MR. Mitochondrial quality control and dynamics in Parkinson’s disease. Antioxid Redox Signal. 2012;16:869–882. doi: 10.1089/ars.2011.4019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McGARRY A, MCDERMOTT M, KIEBURTZ K, ET AL. A randomized, double-blind, placebo-controlled trial of coenzyme Q10 in Huntington disease. Neurology. 2017;88:152–159. doi: 10.1212/WNL.0000000000003478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McMANUS MJ, MURPHY MP, FRANKLIN JL. The mitochondria-targeted antioxidant MitoQ prevents loss of spatial memory retention and early neuropathology in a transgenic mouse model of Alzheimer’s disease. J Neurosci. 2011;31:15703–15715. doi: 10.1523/JNEUROSCI.0552-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MEIER T, PERLMAN SL, RUMMEY C, COPPARD NJ, LYNCH DR. Assessment of neurological efficacy of idebenone in pediatric patients with Friedreich’s ataxia: data from a 6-month controlled study followed by a 12-month open-label extension study. J Neurol. 2012;259:284–291. doi: 10.1007/s00415-011-6174-y. [DOI] [PubMed] [Google Scholar]
- MENALLED LB, PATRY M, RAGLAND N, LOWDEN PA, GOODMAN J, MINNICH J, ZAHASKY B, PARK L, LEEDS J, HOWLAND D, SIGNER E, TOBIN AJ, BRUNNER D. Comprehensive behavioral testing in the R6/2 mouse model of Huntington’s disease shows no benefit from CoQ10 or minocycline. PLoS One. 2010;5:e9793. doi: 10.1371/journal.pone.0009793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MIQUEL E, CASSINA A, MARTÍNEZ-PALMA L, SOUZA JM, BOLATTO C, RODRÍGUEZ-BOTTERO S, LOGAN A, SMITH RA, MURPHY MP, BARBEITO L, RADI R, CASSINA P. Neuroprotective effects of the mitochondria-targeted antioxidant MitoQ in a model of inherited amyotrophic lateral sclerosis. Free Radic Biol Med. 2014;70:204–213. doi: 10.1016/j.freeradbiomed.2014.02.019. [DOI] [PubMed] [Google Scholar]
- MISCHLEY LK, ALLEN J, BRADLEY R. Coenzyme Q10 deficiency in patients with Parkinson’s disease. J Neurol Sci. 2012;318:72–75. doi: 10.1016/j.jns.2012.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MIZUNO Y, OHTA S, TANAKA M, TAKAMIYA S, SUZUKI K, SATO T, OYA H, OZAWA T, KAGAWA Y. Deficiencies in complex I subunits of the respiratory chain in Parkinson’s disease. Biochem Biophys Res Commun. 1989;163:1450–1455. doi: 10.1016/0006-291x(89)91141-8. [DOI] [PubMed] [Google Scholar]
- MOCCIA M, CAPACCHIONE A, LANZILLO R, CARBONE F, MICILLO T, PERNA F, DE ROSA A, CAROTENUTO A, ALBERO R, MATARESE G, PALLADINO R, BRESCIA MORRA V. Coenzyme Q10 supplementation reduces peripheral oxidative stress and inflammation in interferon-β1a-treated multiple sclerosis. Ther Adv Neurol Disord. 2019;12:1756286418819074. doi: 10.1177/1756286418819074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MOLINA J, DE BUSTOS F, JIMÉNEZ-JIMÉNEZ FJ, GÓMEZ-ESCALONILLA C, GARCÍA-REDONDO A, ESTEBAN J, GUERRERO-SOLA A, DEL HOYO P, MARTÍNEZ-SALIO A, RAMÍREZ-RAMOS C, INDURAIN GR, ARENAS J. Serum levels of coenzyme Q10 in patients with amyotrophic lateral sclerosis. J Neural Transm. 2000;107:1021–1026. doi: 10.1007/s007020070050. [DOI] [PubMed] [Google Scholar]
- MOLLET J, GIURGEA I, SCHLEMMER D, DALLNER G, CHRETIEN D, DELAHODDE A, BACQ D, DE LONLAY P, MUNNICH A, RÖTIG A. Prenyldiphosphate synthase, subunit 1 (PDSS1) and OH-benzoate polyprenyltransferase (COQ2) mutations in ubiquinone deficiency and oxidative phosphorylation disorders. J Clin Invest. 2007;117:765–772. doi: 10.1172/JCI29089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MONTENEGRO L, TURNATURI R, PARENTI C, PASQUINUCCI L. Idebenone: Novel strategies to improve its systemic and local efficacy. Nanomaterials (Basel) 2018;8:87. doi: 10.3390/nano8020087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MÜLLER T, BÜTTNER T, GHOLIPOUR AF, KUHN W. Coenzyme Q10 supplementation provides mild symptomatic benefit in patients with Parkinson’s disease. Neurosci Lett. 2003;341:201–204. doi: 10.1016/s0304-3940(03)00185-x. [DOI] [PubMed] [Google Scholar]
- MURPHY MP, SMITH RA. Targeting antioxidants to mitochondria by conjugation to lipophilic cations. Annu Rev Pharmacol Toxicol. 2007;47:629–656. doi: 10.1146/annurev.pharmtox.47.120505.105110. [DOI] [PubMed] [Google Scholar]
- MUSCOLI C, FRESTA M, CARDILE V, PALUMBO M, RENIS M, PUGLISI G, PAOLINO D, NISTICÒ S, ROTIROTI D, MOLLACE V. Ethanol-induced injury in rat primary cortical astrocytes involves oxidative stress: effect of idebenone. Neurosci Lett. 2002;329:21–24. doi: 10.1016/s0304-3940(02)00567-0. [DOI] [PubMed] [Google Scholar]
- MUTHUKUMARAN K, KANWAR A, VEGH C, MARGINEAN A, ELLIOTT A, GUILBEAULT N, BADOUR A, SIKORSKA M, COHEN J, PANDEY S. Ubisol-Q10 (a nanomicellar water-soluble formulation of CoQ10) treatment inhibits Alzheimer-type behavioral and pathological symptoms in a double transgenic mouse (TgAPEswe, PSEN1dE9) model of Alzheimer’s dsease. J Alzheimers Dis. 2018;61:221–236. doi: 10.3233/JAD-170275. [DOI] [PubMed] [Google Scholar]
- MUTHUKUMARAN K, LEAHY S, HARRISON K, SIKORSKA M, SANDHU JK, COHEN J, KESHAN C, LOPATIN D, MILLER H, BOROWY-BOROWSKI H, LANTHIER P, WEINSTOCK S, PANDEY S. Orally delivered water soluble coenzyme Q10 (Ubisol-Q10) blocks on-going neurodegeneration in rats exposed to paraquat: potential for therapeutic application in Parkinson’s disease. BMC Neurosci. 2014a;15:21. doi: 10.1186/1471-2202-15-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MUTHUKUMARAN K, SMITH J, JASRA H, SIKORSKA M, SANDHU JK, COHEN J, LOPATIN D, PANDEY S. Genetic susceptibility model of Parkinson’s disease resulting from exposure of DJ-1 deficient mice to MPTP: evaluation of neuroprotection by Ubisol-Q10. J Parkinsons Dis. 2014b;4:523–530. doi: 10.3233/JPD-140368. [DOI] [PubMed] [Google Scholar]
- NAGASE M, YAMAMOTO Y, MATSUMOTO N, ARAI Y, HIROSE N. Increased oxidative stress and coenzyme Q10 deficiency in centenarians. J Clin Biochem Nutr. 2018;63:129–136. doi: 10.3164/jcbn.17-124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- NAGASE M, YAMAMOTO Y, MIYAZAKI Y, YOSHINO H. Increased oxidative stress in patients with amyotrophic lateral sclerosis and the effect of edaravone administration. Redox Rep. 2016;21:104–112. doi: 10.1179/1351000215Y.0000000026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- NEGIDA A, MENSHAWY A, EL ASHAL G, ELFOULY Y, HANI Y, HEGAZY Y, EL GHONIMY S, FOUDA S, RASHAD Y. Coenzyme Q10 for patients with Parkinson’s disease: A systematic review and meta-analysis. CNS Neurol Disord Drug Targets. 2016;15:45–53. doi: 10.2174/1871527314666150821103306. [DOI] [PubMed] [Google Scholar]
- NIKLOWITZ P, ONUR S, FISCHER A, LAUDES M, PALUSSEN M, MENKE T, DÖRING F. Coenzyme Q10 serum concentration and redox status in European adults: influence of age, sex, and lipoprotein concentration. J Clin Biochem Nutr. 2016;58:240–245. doi: 10.3164/jcbn.15-73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- OCHIAI A, ITAGAKI S, KUROKAWA T, KOBAYASHI M, HIRANO T, ISEKI K. Improvement in intestinal coenzyme Q10 absorption by food intake. Yakugaku Zasshi. 2007;127:1251–1254. doi: 10.1248/yakushi.127.1251. [DOI] [PubMed] [Google Scholar]
- OERTEL W, SCHULZ JB. Current and experimental treatments of Parkinson disease: A guide for neuroscientists. J Neurochem. 2016;139(Suppl 1):325–337. doi: 10.1111/jnc.13750. [DOI] [PubMed] [Google Scholar]
- OGASAHARA S, ENGEL AG, FRENS D, MACK D. Muscle coenzyme Q deficiency in familial mitochondrial encephalomyopathy. Proc Natl Acad Sci U S A. 1989;86:2379–2382. doi: 10.1073/pnas.86.7.2379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ONUR S, NIKLOWITZ P, FISCHER A, METGES CC, GRUNE T, MENKE T, RIMBACH G, DÖRING F. A comparative study into alterations of coenzyme Q redox status in ageing pigs, mice, and worms. Biofactors. 2014;40:346–354. doi: 10.1002/biof.1160. [DOI] [PubMed] [Google Scholar]
- OTEIZA PI, UCHITEL OD, CARRASQUEDO F, DUBROVSKI AL, ROMA JC, FRAGA CG. Evaluation of antioxidants, protein, and lipid oxidation products in blood from sporadic amyotrophic lateral sclerosis patients. Neurochem Res. 1997;22:535–539. doi: 10.1023/a:1027384432715. [DOI] [PubMed] [Google Scholar]
- PALAMAKULA A, SOLIMAN M, KHAN MM. Regional permeability of coenzyme Q10 in isolated rat gastrointestinal tracts. Pharmazie. 2005;60:212–214. [PubMed] [Google Scholar]
- PARKER WD, JR, PARKS JK, SWERDLOW RH. Complex I deficiency in Parkinson’s disease frontal cortex. Brain Res. 2008;1189:215–218. doi: 10.1016/j.brainres.2007.10.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- PARKINSON MH, SCHULZ JB, GIUNTI P. Co-enzyme Q10 and idebenone use in Friedreich’s ataxia. J Neurochem. 2013;126(Suppl 1):125–141. doi: 10.1111/jnc.12322. [DOI] [PubMed] [Google Scholar]
- PARKINSON STUDY GROUP QE3 INVESTIGATORS et al. A randomized clinical trial of high-dosage coenzyme Q10 in early Parkinson disease: no evidence of benefit. JAMA Neurol. 2014;71:543–552. doi: 10.1001/jamaneurol.2014.131. [DOI] [PubMed] [Google Scholar]
- PAROHAN M, SARRAF P, JAVANBAKHT MH, FOROUSHANI AR, 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. 2021;24:317–326. doi: 10.1080/1028415X.2019.1627770. [DOI] [PubMed] [Google Scholar]
- PAROHAN M, SARRAF P, JAVANBAKHT MH, RANJI-BURACHALOO S, DJALALI M. Effect of coenzyme Q10 supplementation on clinical features of migraine: a systematic review and dose-response meta-analysis of randomized controlled trials. Nutr Neurosci. 2020;23:868–875. doi: 10.1080/1028415X.2019.1572940. [DOI] [PubMed] [Google Scholar]
- PASTOR-MALDONADO CJ, SUÁREZ-RIVERO JM, POVEA-CABELLO S, ÁLVAREZ-CÓRDOBA M, VILLALÓN-GARCÍA I, MUNUERA-CABEZA M, SUÁREZ-CARRILLO A, TALAVERÓN-REY M, SÁNCHEZ-ALCÁZAR JA. Coenzyme Q10: Novel Formulations and Medical Trends. Int J Mol Sci. 2020;21:8432. doi: 10.3390/ijms21228432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- POTGIETER M, PRETORIUS E, PEPPER MS. Primary and secondary coenzyme Q10 deficiency: the role of therapeutic supplementation. Nutr Rev. 2013;71:180–188. doi: 10.1111/nure.12011. [DOI] [PubMed] [Google Scholar]
- PRAVST I, RODRÍGUEZ AGUILERA JC, CORTES RODRIGUEZ AB, JAZBAR J, LOCATELLI I, HRISTOV H, ŽMITEK K. Comparative bioavailability of different coenzyme Q10 formulations in healthy elderly individuals. Nutrients. 2020;12 doi: 10.3390/nu12030784. pii: E784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- PRAVST I, ZMITEK K, ZMITEK J. Coenzyme Q10 contents in foods and fortification strategies. Crit Rev Food Sci Nutr. 2010;50:269–280. doi: 10.1080/10408390902773037. [DOI] [PubMed] [Google Scholar]
- PRZEDBORSKI S, TIEU K, PERIER C, VILA M. MPTP as a mitochondrial neurotoxic model of Parkinson’s disease. J Bioenerg Biomembr. 2004;36:375–379. doi: 10.1023/B:JOBB.0000041771.66775.d5. [DOI] [PubMed] [Google Scholar]
- PYO Y. Coenzyme Q10 and Q9 contents in 6 commercial vegetable oils and their average daily intakes in Korea. Food Sci Biotechnol. 2010;9:837–841. doi: 10.1007/s10068-010-0118-7. [DOI] [Google Scholar]
- QUINZII C, NAINI A, SALVIATI L, TREVISSON E, NAVAS P, DIMAURO S, HIRANO M. A mutation in para-hydroxybenzoate-polyprenyl transferase (COQ2) causes primary coenzyme Q10 deficiency. Am J Hum Genet. 2006;78:345–349. doi: 10.1086/500092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- QUINZII CM, LUNA-SANCHEZ M, ZIOSI M, HIDALGO-GUTIERREZ A, KLEINER G, LOPEZ LC. The role of sulfide oxidation impairment in the pathogenesis of primary CoQ deficiency. Front Physiol. 2017;8:525. doi: 10.3389/fphys.2017.00525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- RAIZNER AE. Coenzyme Q10. Methodist Debakey Cardiovasc J. 2019;15:185–191. doi: 10.14797/mdcj-15-3-185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- RAMASARMA T. Natural occurrence and distribution of coenzyme Q. In: Lenaz G, editor. Coenzyme Q. Biochemistry, Bioenergetics and Clinical Applications of Ubiquinone. Wiley & Sons; Chichester: 1985. pp. 67–81. [Google Scholar]
- RANEN NG, PEYSER CE, COYLE JT, BYLSMA FW, SHERR M, DAY L, FOLSTEIN MF, BRANDT J, ROSS CA, FOLSTEIN SE. A controlled trial of idebenone in Huntington’s disease. Mov Disord. 1996;11:549–554. doi: 10.1002/mds.870110510. [DOI] [PubMed] [Google Scholar]
- RAUCHOVÁ H, BATTINO M, FATO R, LENAZ G, DRAHOTA Z. Coenzyme Q-pool function in glycerol-3-phosphate oxidation in hamster brown adipose tissue mitochondria. J Bioenerg Biomembr. 1992;24:235–241. doi: 10.1007/BF00762682. [DOI] [PubMed] [Google Scholar]
- RAUCHOVÁ H, DRAHOTA Z, BERGAMINI C, FATO R, LENAZ G. Modification of respiratory-chain enzyme activities in brown adipose tissue mitochondria by idebenone (hydroxydecyl-ubiquinone) J Bioenerg Biomembr. 2008;40:85–93. doi: 10.1007/s10863-008-9134-1. [DOI] [PubMed] [Google Scholar]
- RAUCHOVÁ H, FATO R, DRAHOTA Z, LENAZ G. Steady-state kinetics of reduction of coenzyme Q analogs by glycerol-3-phosphate dehydrogenase in brown adipose tissue mitochondria. Arch Biochem Biophys. 1997;344:235–241. doi: 10.1006/abbi.1997.0150. [DOI] [PubMed] [Google Scholar]
- RAUCHOVÁ H, VOKURKOVÁ M. Recent view of coenzyme Q. Chem Listy. 2009;103:32–39. (In Czech) [Google Scholar]
- RAUCHOVÁ H, VOKURKOVÁ M, DRAHOTA Z. Idebenone-induced recovery of glycerol-3-phosphate and succinate oxidation inhibited by digitonin. Physiol Res. 2012;61:259–265. doi: 10.33549/physiolres.932318. [DOI] [PubMed] [Google Scholar]
- RAUCHOVÁ H, VRBACKÝ M, BERGAMINI C, FATO R, LENAZ G, HOUSTEK J, DRAHOTA Z. Inhibition of glycerophosphate-dependent H2O2 generation in brown fat mitochondria by idebenone. Biochem Biophys Res Commun. 2006;339:362–366. doi: 10.1016/j.bbrc.2005.11.035. [DOI] [PubMed] [Google Scholar]
- REIS RAG, CALIL FA, FELICIANO PR, PINHEIRO MP, NONATO MC. The dihydroorotate dehydrogenases: Past and present. Arch Biochem Biophys. 2017;632:175–191. doi: 10.1016/j.abb.2017.06.019. [DOI] [PubMed] [Google Scholar]
- RINALDI C, TUCCI T, MAIONE S, GIUNTA A, DE MICHELE G, FILLA A. Low-dose idebenone treatment in Friedreich’s ataxia with and without cardiac hypertrophy. J Neurol. 2009;256:1434–1437. doi: 10.1007/s00415-009-5130-6. [DOI] [PubMed] [Google Scholar]
- ROSENFELDT FL, HAAS SJ, KRUM H, HADJ A, NG K, LEONG JY, WATTS GF. Coenzyme Q10 in the treatment of hypertension: a meta-analysis of the clinical trials. J Hum Hypertens. 2007;21:297–306. doi: 10.1038/sj.jhh.1002138. [DOI] [PubMed] [Google Scholar]
- RÖTIG A, APPELKVIST EL, GEROMEL V, CHRETIEN D, KADHOM N, EDERY P, LEBIDEAU M, DALLNER G, MUNNICH A, ERNSTER L, RUSTIN P. Quinone-responsive multiple respiratory-chain dysfunction due to widespread coenzyme Q10 deficiency. Lancet. 2000;356(9227):391–395. doi: 10.1016/S0140-6736(00)02531-9. [DOI] [PubMed] [Google Scholar]
- ROZEN TD, OSHINSKY ML, GEBELINE CA, BRADLEY KC, YOUNG WB, SHECHTER AL, SILBERSTEIN SD. Open label trial of coenzyme Q10 as a migraine preventive. Cephalalgia. 2002;22:137–141. doi: 10.1046/j.1468-2982.2002.00335.x. [DOI] [PubMed] [Google Scholar]
- RUSTIN P, VON KLEIST-RETZOW JC, CHANTREL-GROUSSARD K, SIDI D, MUNNICH A, RÖTIG A. Effect of idebenone on cardiomyopathy in Friedreich’s ataxia: a preliminary study. Lancet. 1999;354(9177):477–479. doi: 10.1016/S0140-6736(99)01341-0. [DOI] [PubMed] [Google Scholar]
- SÁNDOR PS, DI CLEMENTE L, COPPOLA G, SAENGER U, FUMAL A, MAGIS D, SEIDEL L, AGOSTI RM, SCHOENEN J. Efficacy of coenzyme Q10 in migraine prophylaxis: a randomized controlled trial. Neurology. 2005;64:713–715. doi: 10.1212/01.WNL.0000151975.03598.ED. [DOI] [PubMed] [Google Scholar]
- SANOOBAR M, EGHTESADI S, AZIMI A, KHALILI M, JAZAYERI S, REZA GOHARI M. Coenzyme Q10 supplementation reduces oxidative stress and increases antioxidant enzyme activity in patients with relapsing-remitting multiple sclerosis. Int J Neurosci. 2013;123:776–782. doi: 10.3109/00207454.2013.801844. [DOI] [PubMed] [Google Scholar]
- SANOOBAR M, EGHTESADI S, AZIMI A, KHALILI M, KHODADADI B, JAZAYERI S, GOHARI MR, ARYAEIAN N. Coenzyme Q10 supplementation ameliorates inflammatory markers in patients with multiple sclerosis: a double blind, placebo, controlled randomized clinical trial. Nutr Neurosci. 2015;18:169–176. doi: 10.1179/1476830513Y.0000000106. [DOI] [PubMed] [Google Scholar]
- SANOOBAR M, DEHGHAN P, KHALILI M, AZIMI A, SEIFAR F. Coenzyme Q10 as a treatment for fatigue and depression in multiple sclerosis patients: A double blind randomized clinical trial. Nutr Neurosci. 2016;19:138–143. doi: 10.1179/1476830515Y.0000000002. [DOI] [PubMed] [Google Scholar]
- SCHAPIRA AH, COOPER JM, DEXTER D, CLARK JB, JENNER P, MARSDEN CD. Mitochondrial complex I deficiency in Parkinson’s disease. J Neurochem. 1990a;54:823–827. doi: 10.1111/j.1471-4159.1990.tb02325.x. [DOI] [PubMed] [Google Scholar]
- SCHAPIRA AH, MANN VM, COOPER JM, DEXTER D, DANIEL SE, JENNER P, CLARK JB, MARSDEN CD. Anatomic and disease specificity of NADH CoQ1 reductase (complex I) deficiency in Parkinson’s disease. J Neurochem. 1990b;55:2142–2145. doi: 10.1111/j.1471-4159.1990.tb05809.x. [DOI] [PubMed] [Google Scholar]
- SCHILLING G, COONFIELD ML, ROSS CA, BORCHELT DR. Coenzyme Q10 and remacemide hydrochloride ameliorate motor deficits in a Huntington’s disease transgenic mouse model. Neurosci Lett. 2001;315:149–153. doi: 10.1016/s0304-3940(01)02326-6. [DOI] [PubMed] [Google Scholar]
- SCHMELZER C, LINDNER I, RIMBACH G, NIKLOWITZ P, MENKE T, DÖRING F. Functions of coenzyme Q10 in inflammation and gene expression. Biofactors. 2008;32:179–183. doi: 10.1002/biof.5520320121. [DOI] [PubMed] [Google Scholar]
- SCHMELZER C, LINDNER I, VOCK C, FUJII K, DÖRING F. Functional connections and pathways of coenzyme Q10-inducible genes: an in-silico study. IUBMB Life. 2007;59:628–633. doi: 10.1080/15216540701545991. [DOI] [PubMed] [Google Scholar]
- SENIN U, PARNETTI L, BARBAGALLO-SANGIORGI G, BARTORELLI L, BOCOLA V, CAPURSO A, CUZZUPOLI M, DENARO M, MARIGLIANO V, TAMMARO AE, FIORAVANTI M. Idebenone in senile dementia of Alzheimer type: a multicentre study. Arch Gerontol Geriatr. 1992;15:249–260. doi: 10.1016/0167-4943(92)90060-h. [DOI] [PubMed] [Google Scholar]
- SHERER TB, BETARBET R, TESTA CM, SEO BB, RICHARDSON JR, KIM JH, MILLER GW, YAGI T, MATSUNO-YAGI A, GREENAMYRE JT. Mechanism of toxicity in rotenone models of Parkinson’s disease. J Neurosci. 2003;23:10756–10764. doi: 10.1523/JNEUROSCI.23-34-10756.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SHULTS CW, HAAS RH, PASSOV D, BEAL MF. Coenzyme Q10 levels correlate with the activities of complexes I and II/III in mitochondria from parkinsonian and nonparkinsonian subjects. Ann Neurol. 1997;42:261–264. doi: 10.1002/ana.410420221. [DOI] [PubMed] [Google Scholar]
- SHULTS CW, OAKES D, KIEBURTZ K, BEAL MF, HAAS R, PLUMB S, JUNCOS JL, NUTT J, SHOULSON I, CARTER J, KOMPOLITI K, PERLMUTTER JS, REICH S, STERN M, WATTS RL, KURLAN R, MOLHO E, HARRISON M, LEW M PARKINSON STUDY GROUP. Effects of coenzyme Q10 in early Parkinson disease: evidence of slowing of the functional decline. Arch Neurol. 2002;59:1541–1550. doi: 10.1001/archneur.59.10.1541. [DOI] [PubMed] [Google Scholar]
- SINGH RB, NIAZ MA, KUMAR A, SINDBERG CD, MOESGAARD S, LITTARRU GP. Effect on absorption and oxidative stress of different oral Coenzyme Q10 dosages and intake strategy in healthy men. Biofactors. 2005;25:219–224. doi: 10.1002/biof.5520250127. [DOI] [PubMed] [Google Scholar]
- SLATER SK, NELSON TD, KABBOUCHE MA, LECATES SL, HORN P, SEGERS A, MANNING P, POWERS SW, HERSHEY AD. A randomized, double-blinded, placebo-controlled, crossover, add-on study of CoEnzyme Q10 in the prevention of pediatric and adolescent migraine. Cephalalgia. 2011;31:897–905. doi: 10.1177/0333102411406755. [DOI] [PubMed] [Google Scholar]
- SMITH EF, SHAW PJ, DE VOS KJ. The role of mitochondria in amyotrophic lateral sclerosis. Neurosci Lett. 2019;710:132933. doi: 10.1016/j.neulet.2017.06.052. [DOI] [PubMed] [Google Scholar]
- SMITH KM, MATSON S, MATSON WR, CORMIER K, DEL SIGNORE SJ, HAGERTY SW, STACK EC, RYU H, FERRANTE RJ. Dose ranging and efficacy study of high-dose coenzyme Q10 formulations in Huntington’s disease mice. Biochim Biophys Acta. 2006;1762:616–626. doi: 10.1016/j.bbadis.2006.03.004. [DOI] [PubMed] [Google Scholar]
- SMITH RA, MURPHY MP. Animal and human studies with the mitochondria-targeted antioxidant MitoQ. Ann N Y Acad Sci. 2010;1201:96–103. doi: 10.1111/j.1749-6632.2010.05627.x. [DOI] [PubMed] [Google Scholar]
- SOCHOCKA M, ZWOLIŃSKA K, LESZEK J. The infectious etiology of Alzheimer’s disease. Curr Neuropharmacol. 2017;15:996–1009. doi: 10.2174/1570159X15666170313122937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SÖDERBERG M, EDLUND C, ALAFUZOFF I, KRISTENSSON K, DALLNER G. Lipid composition in different regions of the brain in Alzheimer’s disease/senile dementia of Alzheimer’s type. J Neurochem. 1992;59:1646–1653. doi: 10.1111/j.1471-4159.1992.tb10994.x. [DOI] [PubMed] [Google Scholar]
- SOHAL RS, KAMZALOV S, SUMIEN N, FERGUSON M, REBRIN I, HEINRICH KR, FORSTER MJ. Effect of coenzyme Q10 intake on endogenous coenzyme Q content, mitochondrial electron transport chain, antioxidative defenses, and life span of mice. Free Radic Biol Med. 2006;40:480–487. doi: 10.1016/j.freeradbiomed.2005.08.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SOHMIYA M, TANAKA M, SUZUKI Y, TANINO Y, OKAMOTO K, YAMAMOTO Y. An increase of oxidized coenzyme Q-10 occurs in the plasma of sporadic ALS patients. J Neurol Sci. 2005;228:49–53. doi: 10.1016/j.jns.2004.09.030. [DOI] [PubMed] [Google Scholar]
- SOLEIMANI M, JAMEIE SB, BARATI M, MEHDIZADEH M, KERDARI M. Effects of coenzyme Q10 on the ratio of TH1/TH2 in experimental autoimmune encephalomyelitis model of multiple sclerosis in C57BL/6. Iran Biomed J. 2014;18:203–211. doi: 10.6091/ibj.13362.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- SOLESIO ME, PRIME TA, LOGAN A, MURPHY MP, DEL MAR ARROYO-JIMENEZ M, JORDÁN J, GALINDO MF. The mitochondria-targeted anti-oxidant MitoQ reduces aspects of mitochondrial fission in the 6-OHDA cell model of Parkinson’s disease. Biochim Biophys Acta. 2013;1832:174–182. doi: 10.1016/j.bbadis.2012.07.009. [DOI] [PubMed] [Google Scholar]
- SPARACO M, FELEPPA M, LIPTON RB, RAPOPORT AM, BIGAL ME. Mitochondrial dysfunction and migraine: evidence and hypotheses. Cephalalgia. 2006;26:361–372. doi: 10.1111/j.1468-2982.2005.01059.x. [DOI] [PubMed] [Google Scholar]
- SPINDLER M, BEAL MF, HENCHCLIFFE C. Coenzyme Q10 effects in neurodegenerative disease. Neuropsychiatr Dis Treat. 2009;5:597–610. doi: 10.2147/ndt.s5212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- STACK EC, SMITH KM, RYU H, CORMIER K, CHEN M, HAGERTY SW, DEL SIGNORE SJ, CUDKOWICZ ME, FRIEDLANDER RM, FERRANTE RJ. Combination therapy using minocycline and coenzyme Q10 in R6/2 transgenic Huntington’s disease mice. Biochim Biophys Acta. 2006;1762:373–380. doi: 10.1016/j.bbadis.2005.11.002. [DOI] [PubMed] [Google Scholar]
- STANGA S, CARETTO A, BOIDO M, VERCELLI A. Mitochondrial dysfunctions: A red thread across neurodegenerative diseases. Int J Mol Sci. 2020;21:E3719. doi: 10.3390/ijms21103719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- STEFELY JA, PAGLIARINI DJ. Biochemistry of mitochondrial coenzyme Q biosynthesis. Trends Biochem Sci. 2017;42:824–843. doi: 10.1016/j.tibs.2017.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- STRIJKS E, KREMER HP, HORSTINK MW. Q10 therapy in patients with idiopathic Parkinson’s disease. Mol Aspects Med. 1997;18(Suppl):S237–S240. doi: 10.1016/s0098-2997(97)00008-3. [DOI] [PubMed] [Google Scholar]
- SVED AF, ITO S, SVED JC. Brainstem mechanisms of hypertension: role of the rostral ventrolateral medulla. Curr Hypertens Rep. 2003;5:262–268. doi: 10.1007/s11906-003-0030-0. [DOI] [PubMed] [Google Scholar]
- TAI MH, WANG LL, WU KL, CHAN JY. Increased superoxide anion in rostral ventrolateral medulla contributes to hypertension in spontaneously hypertensive rats via interactions with nitric oxide. Free Radic Biol Med. 2005;38:450–462. doi: 10.1016/j.freeradbiomed.2004.11.015. [DOI] [PubMed] [Google Scholar]
- TAKAHASHI M, TAKAHASHI K. Water-soluble CoQ10 as a promising anti-aging agent for neurological dysfunction in brain mitochondria. Antioxidants (Basel) 2019;8:61. doi: 10.3390/antiox8030061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- TASSET I, AGÜERA E, SÁNCHEZ-LÓPEZ F, FEIJÓO M, GIRALDO AI, CRUZ AH, GASCÓN F, TÚNEZ I. Peripheral oxidative stress in relapsing-remitting multiple sclerosis. Clin Biochem. 2012;45:440–444. doi: 10.1016/j.clinbiochem.2012.01.023. [DOI] [PubMed] [Google Scholar]
- THAL LJ, GRUNDMAN M, BERG J, ERNSTROM K, MARGOLIN R, PFEIFFER E, WEINER MF, ZAMRINI E, THOMAS RG. Idebenone treatment fails to slow cognitive decline in Alzheimer’s disease. Neurology. 2003;61:1498–1502. doi: 10.1212/01.wnl.0000096376.03678.c1. [DOI] [PubMed] [Google Scholar]
- TIEU K. A guide to neurotoxic animal models of Parkinson’s disease. Cold Spring Harb Perspect Med. 2011;1(1):a009316. doi: 10.1101/cshperspect.a009316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- TOBORE TO. Towards a comprehensive understanding of the contributions of mitochondrial dysfunction and oxidative stress in the pathogenesis and pathophysiology of Huntington’s disease. J Neurosci Res. 2019;97:1455–1468. doi: 10.1002/jnr.24492. [DOI] [PubMed] [Google Scholar]
- TRUMPOWER BL. New concepts on the role of ubiquinone in the mitochondrial respiratory chain. J Bioenerg Biomembr. 1981;13:1–24. doi: 10.1007/BF00744743. [DOI] [PubMed] [Google Scholar]
- VEGH C, PUPULIN S, WEAR D, CULMONE L, HUGGARD R, MA D, PANDEY S. Resumption of autophagy by Ubisol-Q10 in presenilin-1 mutated fibroblasts and transgenic AD mice: Implications for inhibition of senescence and neuroprotection. Oxid Med Cell Longev. 2019;2019:7404815. doi: 10.1155/2019/7404815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- VOS T, FLAXMAN AD, NAGHAVI M, LOZANO R, MICHAUD C, EZZATI M, et al. Years lived with disability (YLDs) for 1160 sequelae of 289 diseases and injuries 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380(9859):2163–2196. doi: 10.1016/S0140-6736(12)61729-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WADA H, GOTO H, HAGIWARA S, YAMAMOTO Y. Redox status of coenzyme Q10 is associated with chronological age. J Am Geriatr Soc. 2007;55:1141–1142. doi: 10.1111/j.1532-5415.2007.01209.x. [DOI] [PubMed] [Google Scholar]
- WADSWORTH TL, BISHOP JA, PAPPU AS, WOLTJER RL, QUINN JF. Evaluation of coenzyme Q as an antioxidant strategy for Alzheimer’s disease. J Alzheimers Dis. 2008;14:225–234. doi: 10.3233/jad-2008-14210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WAINWRIGHT L, HARGREAVES IP, GEORGIAN AR, TURNER C, DALTON RN, ABBOTT NJ, HEALES SJR, PRESTON JE. CoQ10 deficient endothelial cell culture model for the investigation of CoQ10 blood-brain barrier transport. J Clin Med. 2020;9:3236. doi: 10.3390/jcm9103236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WALTER L, MIYOSHI H, LEVERVE X, BERNARD P, FONTAINE E. Regulation of the mitochondrial permeability transition pore by ubiquinone analogs. A progress report. Free Radic Res. 2002;36:405–412. doi: 10.1080/10715760290021252. [DOI] [PubMed] [Google Scholar]
- WANG Y, HEKIMI S. Molecular genetics of ubiquinone biosynthesis in animals. Crit Rev Biochem Mol Biol. 2013;48:69–88. doi: 10.3109/10409238.2012.741564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WANG W, ZHAO F, MA X, PERRY G, ZHU X. Mitochondria dysfunction in the pathogenesis of Alzheimer’s disease: recent advances. Mol Neurodegener. 2020;15:30. doi: 10.1186/s13024-020-00376-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WANG Z, BAI Z, QIN X, CHENG Y. Aberrations in oxidative stress markers in amyotrophic lateral sclerosis: A systematic review and meta-analysis. Oxid Med Cell Longev. 2019;2019:1712323. doi: 10.1155/2019/1712323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- WATMOUGH NJ, FRERMAN FE. The electron transfer flavoprotein: ubiquinone oxidoreductases. Biochim Biophys Acta. 2010;1797:1910–1916. doi: 10.1016/j.bbabio.2010.10.007. [DOI] [PubMed] [Google Scholar]
- WEBER C, BYSTED A, HØLMER G. Coenzyme Q10 in the diet--daily intake and relative bioavailability. Mol Aspects Med. 1997a;18(Suppl):S251–254. doi: 10.1016/S0098-2997(97)00003-4. [DOI] [PubMed] [Google Scholar]
- WEBER C, BYSTED A, HØLMER G. The coenzyme Q10 content of the average Danish diet. Int J Vitam Nutr Res. 1997b;67:123–129. [PubMed] [Google Scholar]
- WITTE ME, NIJLAND PG, DREXHAGE JA, GERRITSEN W, GEERTS D, VAN HET HOF B, REIJERKERK A, DE VRIES HE, VAN DER VALK P, VAN HORSSEN J. Reduced expression of PGC-1α partly underlies mitochondrial changes and correlates with neuronal loss in multiple sclerosis cortex. Acta Neuropathol. 2013;125:231–243. doi: 10.1007/s00401-012-1052-y. [DOI] [PubMed] [Google Scholar]
- WOLDEAMANUEL YW, COWAN RP. Migraine affects 1 in 10 people worldwide featuring recent rise: A systematic review and meta-analysis of community-based studies involving 6 million participants. J Neurol Sci. 2017;372:307–315. doi: 10.1016/j.jns.2016.11.071. [DOI] [PubMed] [Google Scholar]
- WU KL, CHAO YM, TSAY SJ, CHEN CH, CHAN SH, DOVINOVA I, CHAN JY. Role of nitric oxide synthase uncoupling at rostral ventrolateral medulla in redox-sensitive hypertension associated with metabolic syndrome. Hypertension. 2014;64:815–824. doi: 10.1161/HYPERTENSIONAHA.114.03777. [DOI] [PubMed] [Google Scholar]
- XI Y, FENG D, TAO K, WANG R, SHI Y, QIN H, MURPHY MP, YANG Q, ZHAO G. MitoQ protects dopaminergic neurons in a 6-OHDA induced PD model by enhancing Mfn2-dependent mitochondrial fusion via activation of PGC-1α. Biochim Biophys Acta Mol Basis Dis. 2018;1864:2859–2870. doi: 10.1016/j.bbadis.2018.05.018. [DOI] [PubMed] [Google Scholar]
- YANG L, CALINGASAN NY, WILLE EJ, CORMIER K, SMITH K, FERRANTE RJ, BEAL MF. Combination therapy with coenzyme Q10 and creatine produces additive neuroprotective effects in models of Parkinson’s and Huntington’s diseases. J Neurochem. 2009;109:1427–1439. doi: 10.1111/j.1471-4159.2009.06074.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- YANG X, YANG Y, LI G, WANG J, YANG ES. Coenzyme Q10 attenuates β-amyloid pathology in the aged transgenic mice with Alzheimer presenilin 1 mutation. J Mol Neurosci. 2008;34:165–171. doi: 10.1007/s12031-007-9033-7. [DOI] [PubMed] [Google Scholar]
- YOUNG ML, FRANKLIN JL. The mitochondria-targeted antioxidant MitoQ inhibits memory loss, neuropathology, and extends lifespan in aged 3xTg-AD mice. Mol Cell Neurosci. 2019;101:103409. doi: 10.1016/j.mcn.2019.103409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- YORITAKA A, KAWAJIRI S, YAMAMOTO Y, NAKAHARA T, ANDO M, HASHIMOTO K, NAGASE M, SAITO Y, HATTORI N. Randomized, double-blind, placebo-controlled pilot trial of reduced coenzyme Q10 for Parkinson’s disease. Parkinsonism Relat Disord. 2015;21:911–916. doi: 10.1016/j.parkreldis.2015.05.022. [DOI] [PubMed] [Google Scholar]
- YORNS WR, JR, HARDISON HH. Mitochondrial dysfunction in migraine. Semin Pediatr Neurol. 2013;20:188–193. doi: 10.1016/j.spen.2013.09.002. [DOI] [PubMed] [Google Scholar]
- YU M, BEGA D. A review of the clinical evidence for complementary and alternative medicine in Huntington’s disease. Tremor Other Hyperkinet Mov (N Y) 2019 Aug 26;9 doi: 10.7916/tohm.v0.678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- YU MH, TSANG MH, LAI S, HO MS, TSE DML, WILLIS B, KWONG AK, CHOU YY, LIN SP, QUINZII CM, HWU WL, CHIEN YH, KUO PL, CHAN VC, TSOI C, CHONG SC, RODENBURG RJT, SMEITINK J, MAK CC, YEUNG KS, FUNG JL, LAM W, HUI J, LEE NC, FUNG CW, CHUNG BH. Primary coenzyme Q10 deficiency-7: expanded phenotypic spectrum and a founder mutation in southern Chinese. NPJ Genom Med. 2019;4:18. doi: 10.1038/s41525-019-0091-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- YUBERO D, MONTERO R, MARTÍN MA, MONTOYA J, RIBES A, GRAZINA M, TREVISSON E, RODRIGUEZ-AGUILERA JC, HARGREAVES IP, SALVIATI L, NAVAS P, ARTUCH R, JOU C, JIMENEZ-MALLEBRERA C, NASCIMENTO A, PÉREZ-DUEÑAS B, ORTEZ C, RAMOS F, COLOMER J, O’CALLAGHAN M, PINEDA M, GARCÍA-CAZORLA A, ESPINÓS C, RUIZ A, MACAYA A, MARCÉ-GRAU A, GARCIA-VILLORIA J, ARIAS A, EMPERADOR S, RUIZ-PESINI E, LOPEZ-GALLARDO E, NEERGHEEN V, SIMÕES M, DIOGO L, BLÁZQUEZ A, GONZÁLEZ-QUINTANA A, DELMIRO A, DOMÍNGUEZ-GONZÁLEZ C, ARENAS J, GARCÍA-SILVA MT, MARTÍN E, QUIJADA P, HERNÁNDEZ-LAÍN A, MORÁN M, RIVAS INFANTE E, ÁVILA POLO R, PARADAS LÓPE C, BAUTISTA LORITE J, MARTÍNEZ FERNÁNDEZ EM, CORTÉS AB, SÁNCHEZ-CUESTA A, CASCAJO MV, ALCÁZAR M, BREA-CALVO G. Secondary coenzyme Q10 deficiencies in oxidative phosphorylation (OXPHOS) and non-OXPHOS disorders. Mitochondrion. 2016;30:51–58. doi: 10.1016/j.mito.2016.06.007. [DOI] [PubMed] [Google Scholar]
- ZAHEDNASAB H, MESBAH-NAMIN SA, BALOOD M. Coenzyme Q10 supplementation and multiple sclerosis. Nutr Neurosci. 2015;18:192. doi: 10.1179/1476830514Y.0000000126. [DOI] [PubMed] [Google Scholar]
- ZENG Z, LI Y, LU S, HUANG W, DI W. Efficacy of CoQ10 as supplementation for migraine. A meta-analysis. Acta Neurol Scand. 2019;139:284–293. doi: 10.1111/ane.13051. [DOI] [PubMed] [Google Scholar]
- 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. doi: 10.1371/journal.pone.0170172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- ZHANG Y, ABERG F, APPELKVIST EL, DALLNER G, ERNSTER L. Uptake of dietary coenzyme Q supplement is limited in rats. J Nutr. 1995;125:446–453. doi: 10.1093/jn/125.3.446. [DOI] [PubMed] [Google Scholar]
- ZHANG Y, APPELKVIST EL, KRISTENSSON K, DALLNER G. The lipid compositions of different regions of rat brain during development and aging. Neurobiol Aging. 1996;17:869–875. doi: 10.1016/s0197-4580(96)00076-0. [DOI] [PubMed] [Google Scholar]
- ZHU ZG, SUN MX, ZHANG WL, WANG WW, JIN YM, XIE CL. The efficacy and safety of coenzyme Q10 in Parkinson’s disease: a meta-analysis of randomized controlled trials. Neurol Sci. 2017;38:215–224. doi: 10.1007/s10072-016-2757-9. [DOI] [PubMed] [Google Scholar]
- ZS-NAGY I. Chemistry, toxicology, pharmacology and pharmacokinetics of idebenone: a review. Arch Gerontol Geriatr. 1990;11:177–186. doi: 10.1016/0167-4943(90)90063-c. [DOI] [PubMed] [Google Scholar]
- ZULIANI C, BARONI L . COQ DEFICIENCY STUDY GROUP. Antioxidants for the prevention and treatment of multiple sclerosis: An overview. In: Watson R, Preedy V, editors. Bioactive Nutraceuticals and Dietary Supplements in Neurological and Brain Disease. Elsevier; 2015. pp. 341–353. [DOI] [Google Scholar]