Next Article in Journal
Exploration of Polyphenols Extracted from Cytisus Plants and Their Potential Applications: A Review
Previous Article in Journal
Redox Imbalance in Nasal Epithelial Cells of Primary Ciliary Dyskinesia Patients
Previous Article in Special Issue
Spatial and Temporal Protein Modules Signatures Associated with Alzheimer Disease in 3xTg-AD Mice Are Restored by Early Ubiquinol Supplementation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CoQ10 and Mitochondrial Dysfunction in Alzheimer’s Disease

by
Zdeněk Fišar
* and
Jana Hroudová
Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague, Czech Republic
*
Author to whom correspondence should be addressed.
Antioxidants 2024, 13(2), 191; https://doi.org/10.3390/antiox13020191
Submission received: 31 December 2023 / Revised: 30 January 2024 / Accepted: 1 February 2024 / Published: 2 February 2024
(This article belongs to the Special Issue CoQ10 and Aging and Age-Related Diseases—2nd Edition)

Abstract

:
The progress in understanding the pathogenesis and treatment of Alzheimer’s disease (AD) is based on the recognition of the primary causes of the disease, which can be deduced from the knowledge of risk factors and biomarkers measurable in the early stages of the disease. Insights into the risk factors and the time course of biomarker abnormalities point to a role for the connection of amyloid beta (Aβ) pathology, tau pathology, mitochondrial dysfunction, and oxidative stress in the onset and development of AD. Coenzyme Q10 (CoQ10) is a lipid antioxidant and electron transporter in the mitochondrial electron transport system. The availability and activity of CoQ10 is crucial for proper mitochondrial function and cellular bioenergetics. Based on the mitochondrial hypothesis of AD and the hypothesis of oxidative stress, the regulation of the efficiency of the oxidative phosphorylation system by means of CoQ10 can be considered promising in restoring the mitochondrial function impaired in AD, or in preventing the onset of mitochondrial dysfunction and the development of amyloid and tau pathology in AD. This review summarizes the knowledge on the pathophysiology of AD, in which CoQ10 may play a significant role, with the aim of evaluating the perspective of the pharmacotherapy of AD with CoQ10 and its analogues.

1. Introduction

The neurodegeneration and aging of the brain are influenced by genetic and epigenetic factors, external and internal environment, lifestyle, trauma and diseases. Impaired synaptic and structural neuroplasticity is associated with pathophysiological, functional, and morphological changes in the brain that may serve as biomarkers of brain aging and neurodegeneration [1]. Due to the key role of mitochondria in bioenergetics, oxidative stress, metabolism, neuroinflammation, neuroplasticity, and apoptosis [2,3,4], attention has long been paid to mitochondrial dysfunction in aging and age-related neurodegenerative diseases such as Alzheimer’s disease (AD) [5,6,7]. Amyloid beta (Aβ)- and tau-induced neurotoxicity, which is linked to mitochondrial dysfunction and oxidative stress, have been postulated to play critical roles in the pathophysiology of AD [8].
In general, mitochondrial dysfunction is associated with reduced ATP production, increased production of reactive oxygen species (ROS), release of proapoptotic factors, and disturbed calcium homeostasis. The reduced production of ATP is mainly caused by the impaired function of the oxidative phosphorylation system (OXPHOS), which includes a series of redox reactions ending in oxygen, during which the mitochondrial electron transport system (ETS) transfers electrons between the complexes of the respiratory chain with the formation of the proton motive force, but also of superoxide [9]. The essential carrier of electrons from complex I or from complex II to complex III is coenzyme Q (CoQ), especially CoQ10 in humans. The availability and activity of CoQ10 and the Q-cycle play a significant role in the effectiveness of the OXPHOS system. In addition, CoQ10 functions as a lipid antioxidant and a necessary factor for controlling protein uncoupling and opening mitochondrial permeability transition pores (mPTPs).
CoQ10 is endogenously synthesized in every cell; it is also partially absorbed through food, which can affect its availability and activity. Supplementation of CoQ10 and its analogs is tested in the treatment of diseases related to oxidative stress and disruption of cellular energy, including AD and other neurodegenerative diseases [10].
This review summarizes the findings on the role of oxidative stress and mitochondrial dysfunction in the pathophysiology of AD, with a focus on the role of CoQ10. The aim of this review is to show that the understanding of the connection of specific Aβ and tau pathology in AD with mitochondrial dysfunction and oxidative stress, together with the consideration of the function of CoQ10, give the potential for further research into the role of CoQ10 and its analogues in the treatment of AD.

2. Alzheimer’s Disease

AD is a neurodegenerative disease defined as dementia confirmed by neuropathological observation of brain neuritic plaques formed by Aβ and neurofibrillary tangles (NFTs) composed of paired helical filaments of hyperphosphorylated tau protein (P-tau) [11,12]. The cause of the onset of AD, and how the neurotoxicity of Aβ and tau leads to the development of AD pathophysiology, are not yet sufficiently understood. In addition to amyloidopathy and tauopathy, mitochondrial dysfunction, oxidative stress, metabolic disturbances, neuroinflammation, disruption of neurotransmission, accumulation of transition metals, chronic hypoperfusion of brain tissue, and alterations in neurotrophic factors contribute to AD pathogenesis. The multifactorial nature of AD is captured in various interrelated biological hypotheses of AD (Figure 1) [13].
The main pathophysiological feature of AD is impaired proteostasis of pathways involved in the synthesis, folding, post-translational modifications, aggregation, targeting, and degradation of Aβ and tau protein in the brain. Mitochondria, mitochondria-associated membranes (MAMs), and endoplasmic reticulum (ER), which are connected to the ubiquitin proteasome system, autophagy system, ROS production, regulation of free cytosolic calcium, and apoptosis, are significantly involved in these processes [14,15,16,17].
The targets of new potential AD drugs are mainly processes related to Aβ and P-tau neurotoxicity, mitochondrial dysfunction, oxidative stress, metabolic disorders, and neuroinflammation [18,19,20].

2.1. Risk Factors

The exact cause of AD onset remains controversial. Progress is expected from longitudinal studies allowing for the identification of risk factors and early biomarkers of AD detectable in peripheral blood long before the onset of clinical symptoms of the disease [8,21]. The progress in understanding the pathophysiology of AD is linked to progress in the understanding of the mechanisms of aging-related neurodegeneration.
The main risk factor for the sporadic form of AD is age. Aging is a complex event, with gradual increase in cellular dysfunctions caused by the accumulation of protein, lipid, and nucleic acid dysfunctions. In general, age-related neurodegeneration is influenced by environmental factors (diet, exercise, and lifestyle), metabolic and oxidative stress, mitochondrial dysfunction, genetics and epigenetics, cerebrovascular dysfunction, blood-brain barrier dysfunction, neurotoxicity, and neuroinflammation [22,23].
Genetic variation significantly contributes to the risk of AD. Late-onset AD shows a heritability of 58–79% [24,25]. Occurrence of the ε4 allele of the gene APOE that encodes apolipoprotein E4 (ApoE4) is a major genetic risk factor for late-onset sporadic AD [26,27], as ApoE4 increases the neurotoxicity of Aβ and tau, which have a role in AD pathology [28]. Epigenetic changes, including changes in mitochondrial DNA (mtDNA), have been shown to be important in the pathogenesis of AD [29]. The autosomal dominant (familial) form of AD, which is defined as pathologically confirmed dominantly inherited AD, occurs in less than 1% of all cases [30]. However, all forms of AD are thought to share similar pathophysiological processes.
Other risk factors for AD are female gender [31,32], other genetic and epigenetic variations [26,33], brain injury [34], and internal and external environmental factors and stressors [35,36], including low levels of education [37], lifestyle [38], infection [39], cardiovascular disease [40], and metabolic dysregulation [41] such as type 2 diabetes mellitus (T2DM) [42]. The most significant environmental risk factors for the development of AD are late-life depression and T2DM [36].

2.2. Biomarkers

Validated biochemical biomarkers of AD are low concentrations of Aβ42 in cerebrospinal fluid (CSF), which reflect Aβ deposition in the brain, and increased tau in CSF, which is a marker of neuronal degeneration or damage. Tau biomarkers include both total tau (T-tau) and phosphorylated tau (P-tau) [43]. Neuroimaging biomarkers [44,45,46,47], proteomic and metabolomic markers [48], and biomarkers of oxidative stress [49], mitochondrial dysfunction [50,51], and neuroinflammation [52] are currently being sought and studied in AD.
The time course of measurable pathophysiological biomarkers in relation to the clinical course of AD can be used as a basis for the development of new drugs in AD that target pathophysiological processes in the early stages of the disease. A hypothetical time course of biomarker abnormalities and pathological changes in AD is proposed based on the longitudinal measurements of (i) Aβ and P-tau in CSF or in the brain by positron emission tomography (PET), (ii) neurodegeneration using fluorodeoxyglucose (FDG)-PET (hypometabolism) and magnetic resonance imaging (MRI) (hippocampal atrophy), (iii) synaptic dysfunction using FDG-PET and functional MRI (fMRI), (iv) neuroinflammatory changes, and (v) mitochondrial dysfunction [8,30,45,53,54]. According to this model, the onset of AD (even decades before the recognition of clinical symptoms of the disease) is associated with age-related mitochondrial dysfunction and the neurotoxicity of Aβ oligomers, while neurodegeneration and progression of AD is more associated with the neurotoxicity of P-tau oligomers and NFTs.

2.3. Mitochondrial Hypothesis

The mitochondrial cascade hypothesis of AD posits that mitochondrial dysfunction determines the initiation and development of this disease. According to this hypothesis, primary changes in mitochondrial function may induce a cascade of processes that lead to AD-specific neuropathological changes. Aβ and tau pathology/neurotoxicity may also be potentiated by interaction with mitochondrial proteins and membranes. According to the original mitochondrial cascade hypothesis [55], the basic level of mitochondrial function is genetically determined, and the decline of mitochondrial function is determined by aging processes, genetic factors, and environmental influences. If the decline in mitochondrial function exceeds a certain threshold, then the histological and pathophysiological changes specific to AD are triggered.
According to the mitochondrial cascade hypothesis, AD neuropathology arises secondary to mitochondrial dysfunction when the age-associated decline in mitochondrial function reaches a point where compensatory mechanisms are no longer effective [8,56,57]. The primary cause of the disease may not only be mitochondrial dysfunction, amyloidopathy or tauopathy, but also changes in the activity of factors that can cause them and which are localized in mitochondria, such as ApoE4 [58], glycogen synthase kinase 3 [59,60], and monoamine oxidase [61]. It appears that AD may have multiple initiating pathological factors that interact with each other.
Amyloid and tau pathology are specific for AD, but due to the mutual interactions and feedback effects of the aforementioned processes, it is not yet clear what triggers AD. According to the integrative amyloid-tau-mitochondrial hypothesis [8], the interaction of risk factors and biomarkers and their mutual synergy, rather than the primary effects of one particular factor, are decisive for the development of AD.
Mitochondrial dysfunction in AD includes bioenergetic impairment, increased ROS production, impaired mitochondrial dynamics and trafficking, and DNA mutations [62,63]. Mitochondrial abnormalities, including impaired function of mitochondrial ETS complexes and ATP production, have been described in AD [64]. One possibility to regulate the processes associated with neurodegeneration in AD is the regulation of the OXPHOS system through the availability and activity of CoQ10 using metabolic modulators, drugs, diet, and exercise [65,66].
The disruption of synaptic plasticity is one of the first steps in the neurodegeneration process associated with aging and the development of neurodegenerative diseases such as AD. Early deficits in synaptic mitochondria in AD include increased Aβ accumulation, mitochondrial dysfunction, increased mPTPs opening, decreased mitochondrial respiration, and decreased complex IV activity [67,68]. At the same time, Aβ and tau pathologies are in a reciprocal relationship with mitochondrial dysfunction and oxidative stress in AD [8,69,70,71]. Mitochondrial dysfunction associated with Aβ and tau pathology in AD includes decreased ATP production, mitophagy, biogenesis (decreased expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha, PGC-1α), activity of components of the OXPHOS system and other enzymes (e.g., 17β-hydroxysteroid dehydrogenase type 10, HSD-10), mitochondrial membrane potential (Δψm), and import of mitochondrial proteins, imbalance of mitochondrial dynamics (upregulation of dynamin-like protein-1, DRP-1), impairment of intracellular Ca2+ homeostasis, membrane damage, interaction with adenine nucleotide translocator 1 (ANT1) and voltage-dependent anion channel 1 (VDAC1), and increased ROS production, apoptosis (increased cytochrome c release), and mPTP opening [8,63,68,72,73,74] (Figure 2).
In a mouse model of AD, the disruption of mitochondrial bioenergetics has been shown to precede the development of AD pathology [75]. Aβ accumulates in mitochondria and reduces the enzymatic activity of complex II and IV, reduces mitochondrial respiration, and impairs mitochondrial dynamics [73,76,77]. Damage to mitochondrial bioenergetics in AD was demonstrated both by measurements of the brains of AD transgenic mice and by PET neuroimaging in human AD brains [63]. Aβ and tau appear to act synergistically to damage the OXPHOS system, with tau damaging complex I and Aβ damaging complex IV [78].

2.4. Oxidative Stress Hypothesis

According to the oxidative stress hypothesis, the cause of the development of AD is oxidative stress, where damage to brain cells by ROS contributes to neurodegeneration and cognitive decline. Both mitochondrial dysfunction and Aβ and P-tau pathology contribute to increased ROS production [79]. ROS can trigger an inflammatory response and, conversely, inflammation induces oxidative stress [80]. Damage to synapses and brain cells due to oxidative stress may be both a consequence and a cause of Aβ and tau neurotoxicity in AD [81].
The oxidative stress hypothesis is based on the observation that oxidative stress, i.e., an imbalance between the production and elimination of ROS, occurs in neurodegeneration associated with AD. Increased ROS production and/or reduced antioxidant defenses lead to the accumulation of dysfunctional proteins, lipids and nucleic acids (including mtDNA) and impaired mitophagy.
In neurons, superoxide is mainly produced in the mitochondrial matrix during electron transfer in ETS (mainly generated by the mitochondrial complexes I and III). Superoxide is converted directly in the matrix by superoxide dismutase to less reactive hydrogen peroxide, which, however, passes through membranes and can be converted to a very reactive hydroxyl radical in the cytosol [9]. This radical can initiate increased peroxidation of membrane lipids in AD [82]. Lipophilic antioxidants such as CoQ10 have a protective effect on the peroxidation of membrane lipids; thus, administration of CoQ10 and/or regulation of CoQ10 activity in the ETS is being tested in AD therapy.
Mitochondrial dysfunction can lead to both oxidative and nitrosative stress, as impaired electron transfer in the mitochondrial ETS leads to increased production of ROS, and impaired transport of calcium ions into the mitochondrial matrix induces increased production of nitric oxide (NO) and the formation of reactive peroxynitrite.

3. Coenzyme Q10

CoQ10 (ubiquinone, 2,3 dimethoxy-5-methyl-6-decaprenyl-1,4-benzoquinone) is the most common form of CoQ in humans [83]; it is found in all cell membranes, where it acts as an electron carrier and antioxidant. The internal synthesis of CoQ10 starts from the amino acid tyrosine (benzoquinone ring) and from the mevalonate pathway (isoprenoid side chain) (Figure 3) [84,85]. Current knowledge about CoQ10 biosynthesis is described in detail in a series of reviews [86,87,88]. A collection of enzymes that produce CoQ10 (termed as complex Q) are localized in the inner mitochondrial membrane (IMM) and in the ER [89,90].
The main cellular functions of CoQ10 include (i) electron transport in the mitochondrial ETS, with a key role in the formation of ATP, (ii) antioxidant action, including protection against lipid peroxidation, participation in the reduction/recycling of other antioxidant molecules (α-tocopherol and ascorbate), and stabilization of the plasma membrane and cell redox balance, (iii) apoptosis and modulation of mPTPs, (iv) signaling modulation of gene expression, including anti-inflammatory effects, (v) maintenance of the proton gradient on the lysosomal membrane, and (vi) activation of mitochondrial uncoupling proteins [90,91,92,93,94,95,96,97].
CoQ10 is present in mitochondria as part of the electron transport chain. CoQ10 is a lipophilic molecule located in cell membranes, mainly in the IMM. It is crucial for electron transfer between complex I or II and complex III of the respiratory chain, and also for electron capture from dehydrogenases [10,98,99] localized on the outer or inner surface of the IMM. Moreover, CoQ10 and its redox state could indirectly modulate a number of mitochondrial and non-mitochondrial metabolic pathways, such as sulfide metabolism, one-carbon metabolism, glutathione, and ferroptosis; therefore, it can be included in the pathophysiology of some metabolic diseases [99].
CoQ10 occurs in three redox forms: as an oxidized form (ubiquinone, C59H90O4), a reduced form (ubiquinol, C59H92O4), and a semi-oxidized form (semiquinone, C59H91O4; under physiological conditions it occurs as an ion C59H90O4–●) (Figure 3). The reduced form of CoQ10(H2) (ubiquinol) acts as an antioxidant and scavenger of free radicals, thereby preventing oxidative damage to lipids, proteins and nucleic acids. On the other hand, CoQ10 can also have a pro-oxidant role, as it can participate in the formation of superoxide and hydrogen peroxide [100].

3.1. CoQ10 in OXPHOS System

High-energy electrons enter the mitochondrial ETS via complex I (from the reduced coenzyme nicotinamide adenine dinucleotide, NADH) or via complex II (from the reduced coenzyme flavin adenine dinucleotide, FADH2). The product of redox reactions in the complex I is the transfer of 4 protons per 1 NADH molecule into the intermembrane space. Electrons are transferred from complex I or from complex II to complex III (coenzyme Q:cytochrome c—oxidoreductase, sometimes called the cytochrome bc1 complex) by ubiquinol (reduced CoQ10(H2)) and enter the protonmotive Q-cycle of the complex III. In the Q-cycle mechanism, protons are translocated across the IMM as a result of the reoxidation of ubiquinol at the Qo-site on the outer side of the IMM, and the reduction of ubiquinone (CoQ10) at the Qi-site on the matrix side of the IMM (Figure 4). The respiration subunits in the Q-cycle are cytochrome c1 (cyt c1), cyt b (with low (bL) and high (bH) potential hemes), and Rieske protein. The substrates of redox reactions catalyzed by complex III are ubiquinol and two molecules of ferricytochrome c, and the products are ubiquinone, two molecules of ferrocytochrome c, and four protons (released into the intermembrane space and used by ATP synthase during ATP biosynthesis) [92,101,102,103]. Total mitochondrial ETS activity is most easily measured as the kinetics of mitochondrial oxygen consumption [104,105]. Note that the Qo-site of the complex III is an important site for generation of superoxide, and thus has a role in oxidative damage during aging and neurodegeneration.
The efficiency of the OXPHOS system, especially the transfer of electrons by means of CoQ10 and the Q-cycle, is influenced by the existence and organization of respiratory supercomplexes [106,107]. The most common supercomplexes (respirasomes) observed are complex I/III, complex I/III/IV, and complex III/IV. Supercomplex assembly is dynamic, and their formation and stabilization depend on the lipid composition of the IMM, especially on the presence of cardiolipin [108]; the initiation of their formation can be associated with the membrane potential. The formation of supercomplexes can significantly increase the efficiency of the OXPHOS system by optimizing the utilization of ETS substrates, stabilizing complex I and reducing the formation of ROS [109,110]. A constitutive part of respirasomes is CoQ10, which exists in different CoQ10 pools in the IMM [111].
The dependence between respiratory complexes, supercomplex assembly dynamics, and the existence of CoQ10 pools in the effectiveness of the ETS system is intensively studied [112]. According to the classic fluid model, respiratory complexes I–IV are randomly distributed in the IMM, and electron transfer between them is realized by electron carriers and CoQ10 located inside the IMM and cyt c located on the external surface of the IMM (Figure 4). The random collision model [113] assumes that mitochondrial electron transport is a process of random collisions based on the diffusion of individual components in the fluid IMM. The discovery of the existence of supercomplexes in the respiratory chain [114] and the recognition of their function [115] led to the modification of the fluid model to the plasticity model, which adds to the fluid model a new view of the structural and functional complexity in the transfer of electrons in the mitochondrial ETS, as well as the function of the CoQ10 pool in the IMM [111,116,117].
The plasticity model assumes that respiratory complexes can function either individually or as components of supercomplexes. It has long been assumed that free CoQ10 resides in the IMM in a homogeneous pool [118]. With the discovery of respirasomes and their function, it is shown that there is a segmentation of CoQ10 molecules, into a pool attached by supercomplex I/III (CoQNADH pool) and into a pool available for complex II and other enzymes (CoQFADH pool) [109], while the two pools interact with each other. The majority of CoQ that receives electrons from FADH2 and the majority of cyt c apparently remain unbound to supercomplexes.

3.2. CoQ10 in AD Pathophysiology

CoQ10 acts primarily as an antioxidant and bioenergetic modulator (Figure 2). Its antioxidant, neuroprotective, and anti-inflammatory effects can improve oxidative stress and neuroinflammation associated with AD [119]; its effects on the efficiency of the mitochondrial ETS system [120] can contribute to the elimination or prevention of mitochondrial dysfunction in AD.
It is accepted that mitochondrial dysfunctions in the pathophysiology of AD involve the altered activities of several mitochondrial enzymes, including those in the OXPHOS system, mainly in the reduced activity of complex IV [121,122,123]. The role of CoQ10 in bioenergetics led to the assumption that the availability and activity of CoQ10 may contribute to impaired energy metabolism in AD.
One of the mitochondrial parameters that characterizes the efficiency of the OXPHOS system (including the efficiency of CoQ10) is the kinetics of oxygen consumption. Mitochondrial respiratory capacity in platelets from AD patients was confirmed to be significantly reduced and negatively correlated with cognitive decline, as measured using the Mini Mental State Examination (MMSE) score [50,124]. At the same time, an association between MMSE score and plasma CoQ10 concentration in AD patients was revealed [51].
CoQ10 may play a role in specific AD pathological processes. This is supported by protective effects of CoQ10 against Aβ neurotoxicity [125] and the observation that serum CoQ10 levels may be inversely associated with dementia risk [126]. A role for CoQ10 in AD is supported by a study in which the in vitro induction of tau aggregation by CoQ and its presence in paired helical filaments was observed [127].
It is supposed that the determination of serum CoQ10 levels could be useful for predicting the development of AD. A systematic review and meta-analysis of studies measuring CoQ10 levels in AD patients and other dementias, and a review of the therapeutic effects of CoQ10 in humans with AD, was recently published [128] and concluded that that CoQ10 deficiency has not been sufficiently demonstrated in peripheral blood of AD patients.
Oxidative stress is also thought to contribute to AD progression by inducing Aβ overexpression and accumulation, while it is unclear what is the primary cause of disease development. Oxidative stress modulates proteostasis, which is strongly impaired in AD [129]. Therefore, antioxidants are among the new clinically tested potential drugs in AD [20]. The reduced form of CoQ10(H2) is a known antioxidant that also affects Aβ pathology [130]. The protective effects of CoQ10 and other antioxidants against Aβ accumulation in the brain, and against neuroinflammation and hypoxia, have been reported [131].
The therapeutic importance of CoQ10 in the treatment of AD has mainly been demonstrated by studies in cellular and animal models of AD, but has not been confirmed in humans. The neuroprotective effect of CoQ10 on cognitive impairments and oxidative damage of specific areas of the brain of a rat was established [132]. A study with a mouse model of AD showed that there is a relationship between changes in the hippocampus and cerebral cortex and oxidative stress, proteostasis, and bioenergetics, and that CoQ10 may act preventively [133]. Cell culture studies have shown a protective effect of CoQ10 on Aβ neurotoxicity [125,130,134].
The administration of CoQ10 as an antioxidant had no significant effect on biomarkers associated with amyloid and tau pathology in AD measured in CSF (Aβ42, tau, and P-tau), nor on cognitive function [135]. Also, the administration of CoQ10 did not have a significant therapeutic effect in other neurodegenerative diseases such as Parkinson’s disease and Huntington’s disease [136]. Currently, the changes in CoQ10 levels in AD patients, and their effects on improving neuropsychological assessments, are not clearly confirmed, and the exact role of CoQ10 in the pathogenesis of AD and the treatment of neurodegenerative diseases is yet to be clarified [91,128]. Studies that provide clinical and mechanistic data regarding the efficacy of CoQ10 in the treatment of AD-related cognitive decline are lacking. The results so far indicate a lower risk of cognitive impairment in those with normal cognition who consumed CoQ10 daily; therefore, supplementation of CoQ10 can be recommended to slow cognitive decline in AD [137].

4. AD Treatment

4.1. Targets of Novel AD Drugs

The drugs currently approved or recommended for the treatment of AD belong to the category of agents targeting neurotransmitter receptors (cholinesterase inhibitors and N-methyl-d-aspartate receptor antagonists) and Aβ (monoclonal antibodies directed against Aβ plaques, protofibrils, and oligomers). In clinical use, there are drugs aimed at suppressing or alleviating the symptoms of AD (donepezil, rivastigmine, galantamine, memantine and the memantine/donepezil combination). Two disease-modifying drugs (DMDs) targeting Aβ pathology (aducanumab and lecanemab) have recently received accelerated approval from the United States Food and Drug Administration (FDA). Attention is mainly devoted to the development of new effective and specific DMDs. However, there is also ongoing testing of symptomatic substances aimed at improving cognitive functions and neuropsychiatric symptoms in AD, and these are often drugs already approved for the treatment of other diseases or substances used in alternative and complementary medicine [138].
The categories of biological processes and AD drug targets that are closely related to mitochondrial function (metabolism and bioenergetics, synaptic plasticity/neuroprotection, and cell death) and oxidative stress are included in the CADRO classification system (“Common Alzheimer’s and Related Dementias Research Ontology”). According to the periodic annual review of AD drug development [20], a total of 141 agents were tested for the treatment of AD and mild cognitive impairment (MCI) in phase 1, 2, or 3 clinical trials at the beginning of 2023. They mainly targeted inflammation (17.0%), Aβ (15.6%), synaptic plasticity/neuroprotection (12.9%), tau (9.2%), metabolism and bioenergetics (7.1%), and oxidative stress (5.0%).
In addition to new DMDs, drugs targeting the primary causes of AD onset and appropriate combinations of approved drugs with adjuvant agents are also being sought and tested. These supplements, such as CoQ10, ω-3 fatty acids, soy, ginkgo biloba, B vitamins, vitamin D plus calcium, vitamin C, and β-carotene have not yet been shown to prevent cognitive dysfunction in AD [128,139]. It can be expected that if adjuvants have anti-amyloid, anti-tau, neurochemical, mitochondrial, antioxidant or anti-inflammatory effects [125,140], they may also have the therapeutic potential to moderate the progression of cognitive impairment in AD.
Drugs in phase 2 or 3 clinical trials that target oxidative stress include hydralazine, icosapent ethyl, polyunsaturated fatty acids (PUFA), omega-3, edavarone, and Flos gossypii flavonoids. Note that, although oxidative stress is accepted as a key modulator of the biological processes in neurodegenerative diseases [141], therapeutic interventions must also take into account the useful endogenous mechanisms regulated by ROS [142]. Metabolism and bioenergetics are targeted by metformin, tricaprilin, insulin intranasal (+empagliflozin), dapagliflozin, empagliflozin, semaglutide, T3D-959, Chinese traditional medicine, and obicetrapib [20].
Currently, antioxidants are used as supplements to approved AD drugs. The lack of effectiveness of antioxidants in AD therapy can be explained by their non-specific intervention in the balance between ROS activity and the activity of the antioxidant system, which can suppress the useful and necessary role of free radicals in certain areas of the brain. Nevertheless, the testing of CoQ10 and its analogues as a supportive therapy for AD remains important, primarily in the context of suppressing excessive peroxidation of membrane lipids and enhancing bioenergetics.
A suitable target for substances that restore or increase mitochondrial function is the stimulation of mitochondrial biogenesis. Mitochondrial biogenesis is associated not only with cell division, but also with the response to oxidative stress (that is, with the demand for increased cellular energy consumption), exercise, hormones, electrical stimulation, etc. Mitochondrial biogenesis has become the target of new drugs for diseases associated with mitochondrial dysfunction, including neurodegenerative disorders such as AD. Pathways associated with mitochondrial biogenesis, and activated in response to energy deficit, include the activation of the peroxisome PGC-1α axis, activation of the AMP-activated protein kinase (AMPK), and Sirtuin 1 activation [63].
In summary, the main approach in the search for new DMDs for AD is the targeting of the pathophysiological processes that cause the onset of the disease, which, according to current AD hypotheses, are Aβ and tau pathology, mitochondrial dysfunction, oxidative stress, neuroinflammation, disturbed neurotransmission, and disorders of brain metabolism. If we assume that age is the main risk factor for AD, then we can focus on the regulation of processes and biomarkers discussed in the oxidative stress hypothesis and in the mitochondrial hypothesis.

4.2. CoQ10 in AD Treatment

Substances that act as antioxidants or increase mitochondrial bioenergetics have potential for the treatment of neurodegenerative diseases such as AD. From the perspective of the mitochondrial hypothesis of AD and the possibility of pharmacologically influencing mitochondrial dysfunction, the synaptic plasticity and metabolism of brain cells, CoQ10, and mitochondrial proteins and lipids interacting with it also appear to be potential targets for new AD drugs. These substances are capable of regulating the availability and activity of CoQ10, and thus the function of the OXPHOS system.
CoQ10 is mainly formed endogenously, so the cause of a deficiency in CoQ10 can mainly be its impaired biosynthesis, increased degradation, or increased usage. In mitochondrial diseases, CoQ10 deficiency can primarily be caused by mutations in the genes responsible for CoQ10 biosynthesis, or secondarily by defects in other genes [143]. Endogenous metabolites, which are also involved in cholesterol production, may be involved in the regulation of CoQ10 biosynthesis [90]. By upregulating the synthesis of CoQ10, not only its concentration but also its appropriate mitochondrial localization can be achieved. However, dietary intake can also contribute to CoQ10 availability, especially when its endogenous production is reduced with aging or with genetic mutations primarily or secondarily involved in CoQ10 biosynthesis.
Supplemental CoQ10 is well tolerated, but because it is almost insoluble in water, its therapeutic use is limited. To minimize absorption and increase plasma levels of CoQ10, it is recommended to administer it in multiple doses and while dissolved in an oil matrix [144]. To improve the bioavailability of CoQ10, solubilized formulations, including water-soluble CoQ10 analogues, have been developed [145,146,147,148,149,150]. On the periphery, CoQ10 is transported in lipoproteins [151]. The bioavailability and efficacy of oxidized and reduced forms of CoQ10 are similar [152]. CoQ10 has been confirmed to cross the blood-brain barrier in rats and mice, and its oral or intravenous administration leads to increased levels of CoQ10 in the brain [151,153,154], brain mitochondria [155], and an increase in the number of mitochondria in the brain [156], which result in an increase in cellular antioxidant potential [157] and bioenergetics [158].
CoQ10 and other antioxidants or bioenergetics stimulators may be potentially effective in the treatment of neurodegenerative diseases [159,160]. CoQ10 is well characterized as a neuroprotective antioxidant in animal models and in human studies of neurodegenerative disorders. CoQ10 has been shown to reduce oxidative stress and amyloid pathology in a mouse model of AD [161]. Age-related decline in mitochondrial function has been shown in a mouse model to be accompanied by decreased levels of mitochondrial CoQ, and exogenous administration of water-soluble CoQ can lead to the restoration of mitochondrial function [148]. A number of studies have confirmed the significant neuroprotective effects of CoQ10 in experimental biological models, but the suitability of CoQ10 as a biomarker or drug has not been confirmed in AD patients [128].
Given the role of CoQ10 in bioenergetics and antioxidant activity, and the observation that CoQ10 has protective effects against Aβ-induced cell toxicity and impaired synaptic plasticity [134,162], CoQ10, and the processes regulated by it, have strong therapeutic potential in AD [163]. Although studies in animal models of AD show significant improvements in cognition, clinical trials have not been very successful. Therefore, upregulation of brain CoQ10 biosynthesis appears to be more suitable for the treatment of neurodegenerative disorders. An increase in CoQ10 biosynthesis is possible in a physiological way (cold adaptation and exercise) [84], or by targeting of isoprenoid regulation within mevalonate pathway [164], e.g., by the administration of substances, such as epoxidized all-trans polyisoprenoids [90].
The reduced availability of CoQ10 can be eliminated by the dietary administration of this substance. The process of CoQ10 absorption and bioavailability is complex and strongly depends on the formulation of the preparation [152]. CoQ10 and its analogues, idebenone and mitoquinone (MitoQ), are used in the treatment of mitochondrial disorders and in the supportive treatment of neurodegenerative diseases associated with mitochondrial dysfunction, such as AD [85]. CoQ10 is well tolerated and safe, but not approved for the treatment of AD [165].
The synthetic analogue of CoQ10 idebenone (hydroxydyecylubiquinone) acts as an antioxidant and an electron carrier in ETS. Idebenone has protective effects against many toxins [166], but inhibits complex I [167]. In some studies it showed therapeutic effects on AD progression [168], but in other studies no effect of idebenone on cognitive decline in AD [169] or on biomarkers related to Aβ and tau pathology in AD was demonstrated [135].
The administration of MitoQ, which passes through membranes more easily and concentrates in the mitochondria thanks to the attached triphenylphosphonium to ubiquinone, seems promising. MitoQ has protective effects against mitochondrial damage [170] and appears promising in suppressing AD symptoms. In a Caenorhabditis elegans model, the administration of MitoQ extends lifespan and shows protective effects against Aβ toxicity [171]. In experiments with cell cultures and a mouse model of AD, MitoQ was found to increase synaptic connectivity and neurite outgrowth, prevented Aβ-induced oxidative stress and improved memory retention [172,173,174].
In summary, the mitochondria-targeted CoQ10 analogs appear promising for AD therapy. To evaluate the effect of CoQ10 on the activity of the OXPHOS system, or on mitochondrial dysfunction associated with impaired electron transport in the ETS in AD, data from in vivo measurements are not yet available. However, in vitro measurements using isolated mitochondria suggest that exogenously supplied CoQ10 can increase the mitochondrial respiratory rate. It can be hypothesized that the antioxidant activity of CoQ10, which protects mitochondrial membranes from oxidative damage, and the increase in electron transfer efficiency due to the incorporation of CoQ10 into the IMM, contribute to this increase in ETS efficiency. From this point of view, it appears as a perspective synthesis and testing of (i) analogues of CoQ10 with good bioavailability in brain mitochondria, (ii) substances aimed at regulating CoQ10 biosynthesis, and (iii) substances aimed at increasing the activity of CoQ10 in the Q cycle, including those that affect the formation of supercomplexes containing CoQ10 and complex III.

5. Discussion and Conclusions

Based on the findings on which the mitochondrial hypothesis of AD and the oxidative stress hypothesis are based, mitochondrial ETS, especially CoQ10 and the processes mediated by it, appear to be a promising target for new AD drugs. The uptake of dietary CoQ10 into tissues is limited, as CoQ10 is localized in the membranes of the central hydrophobic part of the lipid bilayer; thus, the space available for CoQ10 and similar lipophilic compounds is limited. CoQ10 diffusion in the lipid bilayer may represent the rate-limiting step of electron transfer [175].
CoQ10 concentration decreases with aging, with the availability and redox status of CoQ10 playing a role in the oxidative stress associated with aging [176,177,178,179]. The increased availability of CoQ10 may have neuroprotective effects through antioxidant and bioenergetic effects. However, a systematic review did not confirm a decrease in plasma CoQ10 in AD patients [128]. Also, the decrease in mitochondrial respiratory rate in platelets with age was not different in AD patients compared to age-matched healthy controls [180]. These results indicate that mitochondrial dysfunction (potentially associated with reduced availability and activity of CoQ10) in AD is associated with the onset of the disease rather than its progression. CoQ10 supplementation may then slow down disease-related neurodegeneration.
When we tested in vitro the effect of CoQ10 and other antioxidants on mitochondrial respiration using a model of isolated brain mitochondria, only the addition of CoQ10 caused an increase in the respiratory rate [120]. These results demonstrate that it is possible to regulate OXPHOS activity by CoQ10. It can be hypothesized that the increase in ETS activity may be achieved by the direct incorporation of CoQ10 into the IMM, rather than the antioxidant action of CoQ10. It can be assumed that the improvement of mitochondrial function in AD is possible by increased CoQ10 biosynthesis and increased mitochondrial biogenesis, rather than by dietary CoQ10 supplementation, which does not ensure an increase in CoQ10 in IMM of brain mitochondria. To increase the availability of CoQ10 in the brain and in brain mitochondria, its exogenous administration and stimulation of biosynthesis can be combined. Exogenous supplementation of CoQ10 is safe and does not affect its endogenous biosynthesis [181].
Age-associated mitochondrial dysfunction (measurable as a decrease in ETS capacity or a decrease in respiratory reserve), together with the effects of ApoE4, may be at the start of Aβ and tau pathology, oxidative stress, metabolic dysregulation, and neuroinflammation in the late-onset sporadic form of AD (Figure 2). Targeting new AD drugs on the activity and efficiency of ETS, specifically on the availability and activity of CoQ10 in the IMM and on the regulation of redox processes in the ETS associated with CoQ10, can therefore be considered a promising research approach. The direct in vitro effects of CoQ10 on increasing mitochondrial respiration [120] suggest that the regulation of CoQ10 biosynthesis could be a promising direction in the development of new AD drugs.
In conclusion, the specific pathophysiology of AD is primarily associated with Aβ and tau pathology. According to the mitochondrial hypothesis and the oxidative stress hypothesis, mitochondrial dysfunction and oxidative stress are involved in the pathogenesis of AD, which can be both the initiating and accompanying processes in the pathophysiology of AD and the development of neurodegenerative processes in AD. The approach of targeting new AD drugs to the availability and activity of CoQ10 is underpinned by the role of CoQ10 in the cellular antioxidant system and in mitochondrial bioenergetics. Increasing the availability and activity of CoQ10 is possible by its exogenous administration. Using biological models of isolated brain mitochondria, cell cultures, and animal models of AD, both the antioxidant and bioenergetic effects of CoQ10 have been demonstrated. However, the effects of antioxidants are shown to be insufficiently effective in AD therapy in humans, and exogenous administration of CoQ10 does not yet allow its reliable increased utilization by brain mitochondria. From this point of view, targeting new AD drugs to increase mitochondrial bioenergetics by regulating mitochondrial biogenesis or CoQ10 biosynthesis in the brain appears to be a more appropriate pharmacological strategy. The regulation of the activity of the OXPHOS system through increasing the efficiency of electron transfer in the ETS using CoQ10 and cyt c appears promising. However, targeted pharmacological intervention in mitochondrial electron transfer requires a deeper understanding of the normal and pathological processes in the OXPHOS system, including those associated with the assembly of respiratory complexes into respirasomes and the function of supercomplexes and CoQ10 in electron transport efficiency.
Considering the role of CoQ10 in bioenergetics and lipid peroxidation, it is advisable to continue studying the possibilities of AD therapy by regulating the activity of CoQ10 in the OXPHOS system. In the early stages of AD development, the stimulation of mitochondrial bioenergetics and the antioxidant action of CoQ10 could prevent the development of Aβ and tau neurotoxicity. However, even in the later stages of the disease, the effects of CoQ10 on mitochondrial bioenergetics could slow the progression of the disease. Insights into the possible role of CoQ10 in the pathophysiology of AD show that the regulation of mitochondrial function using CoQ10 and its analogues is a promising approach in the development of new AD drugs and protection against Aβ- and tau-induced neurotoxicity.

Author Contributions

Conceptualization, Z.F. and J.H.; writing—original draft preparation, Z.F.; writing—review and editing, Z.F. and J.H. All authors have read and agreed to the published version of the manuscript.

Funding

Supported by Charles University, Prague, Czech Republic (project Cooperatio, research area Neurosciences) and by the projects of Ministry of Health, Czech Republic (grant number MH CZ-DRO VFN64165 and grant number AZV CR NU23-04-00032).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Mattson, M.P.; Arumugam, T.V. Hallmarks of Brain Aging: Adaptive and Pathological Modification by Metabolic States. Cell Metab. 2018, 27, 1176–1199. [Google Scholar] [CrossRef]
  2. Zia, A.; Pourbagher-Shahri, A.M.; Farkhondeh, T.; Samarghandian, S. Molecular and cellular pathways contributing to brain aging. Behav. Brain Funct. 2021, 17, 6. [Google Scholar] [CrossRef]
  3. Tanaka, M.; Vecsei, L. Editorial of Special Issue ‘Dissecting Neurological and Neuropsychiatric Diseases: Neurodegeneration and Neuroprotection’. Int. J. Mol. Sci. 2022, 23, 6991. [Google Scholar] [CrossRef]
  4. Picca, A.; Calvani, R.; Coelho-Junior, H.J.; Landi, F.; Bernabei, R.; Marzetti, E. Mitochondrial Dysfunction, Oxidative Stress, and Neuroinflammation: Intertwined Roads to Neurodegeneration. Antioxidants 2020, 9, 647. [Google Scholar] [CrossRef]
  5. Hroudová, J.; Singh, N.; Fišar, Z. Mitochondrial dysfunctions in neurodegenerative diseases: Relevance to Alzheimer’s disease. BioMed Res. Int. 2014, 2014, 175062. [Google Scholar] [CrossRef]
  6. Grimm, A.; Eckert, A. Brain aging and neurodegeneration: From a mitochondrial point of view. J. Neurochem. 2017, 143, 418–431. [Google Scholar] [CrossRef] [PubMed]
  7. Wu, Y.; Chen, M.; Jiang, J. Mitochondrial dysfunction in neurodegenerative diseases and drug targets via apoptotic signaling. Mitochondrion 2019, 49, 35–45. [Google Scholar] [CrossRef] [PubMed]
  8. Fišar, Z. Linking the Amyloid, Tau, and Mitochondrial Hypotheses of Alzheimer’s Disease and Identifying Promising Drug Targets. Biomolecules 2022, 12, 1676. [Google Scholar] [CrossRef] [PubMed]
  9. Hroudová, J.; Fišar, Z. Control mechanisms in mitochondrial oxidative phosphorylation. Neural Regen. Res. 2013, 8, 363–375. [Google Scholar] [CrossRef] [PubMed]
  10. Rauchova, H. Coenzyme Q10 effects in neurological diseases. Physiol. Res. 2021, 70 (Suppl. 4), S683–S714. [Google Scholar] [CrossRef] [PubMed]
  11. Pereira, C.F.; Santos, A.E.; Moreira, P.I.; Pereira, A.C.; Sousa, F.J.; Cardoso, S.M.; Cruz, M.T. Is Alzheimer’s disease an inflammasomopathy? Ageing Res. Rev. 2019, 56, 100966. [Google Scholar] [CrossRef] [PubMed]
  12. 2023 Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2023, 19, 1598–1695. [CrossRef] [PubMed]
  13. Liu, P.P.; Xie, Y.; Meng, X.Y.; Kang, J.S. History and progress of hypotheses and clinical trials for Alzheimer’s disease. Signal Transduct. Target. Ther. 2019, 4, 29. [Google Scholar] [CrossRef] [PubMed]
  14. Penke, B.; Szucs, M.; Bogar, F. New Pathways Identify Novel Drug Targets for the Prevention and Treatment of Alzheimer’s Disease. Int. J. Mol. Sci. 2023, 24, 5383. [Google Scholar] [CrossRef]
  15. Penke, B.; Bogar, F.; Fulop, L. beta-Amyloid and the Pathomechanisms of Alzheimer’s Disease: A Comprehensive View. Molecules 2017, 22, 1692. [Google Scholar] [CrossRef]
  16. Schon, E.A.; Area-Gomez, E. Mitochondria-associated ER membranes in Alzheimer disease. Mol. Cell. Neurosci. 2013, 55, 26–36. [Google Scholar] [CrossRef] [PubMed]
  17. Eysert, F.; Kinoshita, P.F.; Mary, A.; Vaillant-Beuchot, L.; Checler, F.; Chami, M. Molecular Dysfunctions of Mitochondria-Associated Membranes (MAMs) in Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 9521. [Google Scholar] [CrossRef]
  18. Jack, C.R., Jr.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement. 2018, 14, 535–562. [Google Scholar] [CrossRef]
  19. McKhann, G.M.; Knopman, D.S.; Chertkow, H.; Hyman, B.T.; Jack, C.R., Jr.; Kawas, C.H.; Klunk, W.E.; Koroshetz, W.J.; Manly, J.J.; Mayeux, R.; et al. The diagnosis of dementia due to Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s Dement. 2011, 7, 263–269. [Google Scholar] [CrossRef]
  20. Cummings, J.; Zhou, Y.; Lee, G.; Zhong, K.; Fonseca, J.; Cheng, F. Alzheimer’s disease drug development pipeline: 2023. Alzheimer’s Dement. 2023, 9, e12385. [Google Scholar] [CrossRef]
  21. Jack, C.R., Jr.; Knopman, D.S.; Jagust, W.J.; Shaw, L.M.; Aisen, P.S.; Weiner, M.W.; Petersen, R.C.; Trojanowski, J.Q. Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet Neurol. 2010, 9, 119–128. [Google Scholar] [CrossRef]
  22. Wareham, L.K.; Liddelow, S.A.; Temple, S.; Benowitz, L.I.; Di Polo, A.; Wellington, C.; Goldberg, J.L.; He, Z.; Duan, X.; Bu, G.; et al. Solving neurodegeneration: Common mechanisms and strategies for new treatments. Mol. Neurodegener. 2022, 17, 23. [Google Scholar] [CrossRef] [PubMed]
  23. Rysz, J.; Franczyk, B.; Rysz-Gorzynska, M.; Gluba-Brzozka, A. Ageing, Age-Related Cardiovascular Risk and the Beneficial Role of Natural Components Intake. Int. J. Mol. Sci. 2021, 23, 183. [Google Scholar] [CrossRef]
  24. Avramopoulos, D. Genetics of Alzheimer’s disease: Recent advances. Genome Med. 2009, 1, 34. [Google Scholar] [CrossRef] [PubMed]
  25. Sims, R.; Hill, M.; Williams, J. The multiplex model of the genetics of Alzheimer’s disease. Nat. Neurosci. 2020, 23, 311–322. [Google Scholar] [CrossRef]
  26. Troutwine, B.R.; Hamid, L.; Lysaker, C.R.; Strope, T.A.; Wilkins, H.M. Apolipoprotein E and Alzheimer’s disease. Acta Pharm. Sin. B 2022, 12, 496–510. [Google Scholar] [CrossRef]
  27. Corder, E.H.; Saunders, A.M.; Strittmatter, W.J.; Schmechel, D.E.; Gaskell, P.C.; Small, G.W.; Roses, A.D.; Haines, J.L.; Pericak-Vance, M.A. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 1993, 261, 921–923. [Google Scholar] [CrossRef]
  28. Shi, Y.; Yamada, K.; Liddelow, S.A.; Smith, S.T.; Zhao, L.; Luo, W.; Tsai, R.M.; Spina, S.; Grinberg, L.T.; Rojas, J.C.; et al. ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature 2017, 549, 523–527. [Google Scholar] [CrossRef] [PubMed]
  29. Gao, X.; Chen, Q.; Yao, H.; Tan, J.; Liu, Z.; Zhou, Y.; Zou, Z. Epigenetics in Alzheimer’s Disease. Front. Aging Neurosci. 2022, 14, 911635. [Google Scholar] [CrossRef]
  30. Bateman, R.J.; Xiong, C.; Benzinger, T.L.; Fagan, A.M.; Goate, A.; Fox, N.C.; Marcus, D.S.; Cairns, N.J.; Xie, X.; Blazey, T.M.; et al. Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. N. Engl. J. Med. 2012, 367, 795–804. [Google Scholar] [CrossRef]
  31. van der Flier, W.M.; Scheltens, P. Epidemiology and risk factors of dementia. J. Neurol. Neurosurg. Psychiatry 2005, 76 (Suppl. 5), v2–v7. [Google Scholar] [CrossRef]
  32. Demetrius, L.A.; Eckert, A.; Grimm, A. Sex differences in Alzheimer’s disease: Metabolic reprogramming and therapeutic intervention. Trends Endocrinol. Metab. 2021, 32, 963–979. [Google Scholar] [CrossRef]
  33. Andrews, S.J.; Fulton-Howard, B.; Goate, A. Interpretation of risk loci from genome-wide association studies of Alzheimer’s disease. Lancet Neurol. 2020, 19, 326–335. [Google Scholar] [CrossRef]
  34. Ramos-Cejudo, J.; Wisniewski, T.; Marmar, C.; Zetterberg, H.; Blennow, K.; de Leon, M.J.; Fossati, S. Traumatic Brain Injury and Alzheimer’s Disease: The Cerebrovascular Link. EBioMedicine 2018, 28, 21–30. [Google Scholar] [CrossRef]
  35. Gaugler, J.; James, B.; Johnson, T.; Reimer, J.; Solis, M.; Weuve, J.; Buckley, R.F.; Hohman, T.J. 2022 Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2022, 18, 700–789. [Google Scholar] [CrossRef]
  36. Bellou, V.; Belbasis, L.; Tzoulaki, I.; Middleton, L.T.; Ioannidis, J.P.A.; Evangelou, E. Systematic evaluation of the associations between environmental risk factors and dementia: An umbrella review of systematic reviews and meta-analyses. Alzheimer’s Dement. 2017, 13, 406–418. [Google Scholar] [CrossRef] [PubMed]
  37. Evans, D.A.; Hebert, L.E.; Beckett, L.A.; Scherr, P.A.; Albert, M.S.; Chown, M.J.; Pilgrim, D.M.; Taylor, J.O. Education and other measures of socioeconomic status and risk of incident Alzheimer disease in a defined population of older persons. Arch. Neurol. 1997, 54, 1399–1405. [Google Scholar] [CrossRef]
  38. Flicker, L. Modifiable lifestyle risk factors for Alzheimer’s disease. J. Alzheimer’s Dis. 2010, 20, 803–811. [Google Scholar] [CrossRef] [PubMed]
  39. Douros, A.; Santella, C.; Dell’Aniello, S.; Azoulay, L.; Renoux, C.; Suissa, S.; Brassard, P. Infectious Disease Burden and the Risk of Alzheimer’s Disease: A Population-Based Study. J. Alzheimer’s Dis. 2021, 81, 329–338. [Google Scholar] [CrossRef]
  40. Luchsinger, J.A.; Reitz, C.; Honig, L.S.; Tang, M.X.; Shea, S.; Mayeux, R. Aggregation of vascular risk factors and risk of incident Alzheimer disease. Neurology 2005, 65, 545–551. [Google Scholar] [CrossRef]
  41. Yan, X.; Hu, Y.; Wang, B.; Wang, S.; Zhang, X. Metabolic Dysregulation Contributes to the Progression of Alzheimer’s Disease. Front. Neurosci. 2020, 14, 530219. [Google Scholar] [CrossRef]
  42. Patel, V.N.; Chorawala, M.R.; Shah, M.B.; Shah, K.C.; Dave, B.P.; Shah, M.P.; Patel, T.M. Emerging Pathophysiological Mechanisms Linking Diabetes Mellitus and Alzheimer’s Disease: An Old Wine in a New Bottle. J. Alzheimer’s Dis. Rep. 2022, 6, 349–357. [Google Scholar] [CrossRef]
  43. Baldeiras, I.; Santana, I.; Leitao, M.J.; Vieira, D.; Duro, D.; Mroczko, B.; Kornhuber, J.; Lewczuk, P. Erlangen Score as a tool to predict progression from mild cognitive impairment to dementia in Alzheimer’s disease. Alzheimer’s Res. Ther. 2019, 11, 2. [Google Scholar] [CrossRef]
  44. Marquez, F.; Yassa, M.A. Neuroimaging Biomarkers for Alzheimer’s Disease. Mol. Neurodegener. 2019, 14, 21. [Google Scholar] [CrossRef]
  45. Leuzy, A.; Chiotis, K.; Lemoine, L.; Gillberg, P.G.; Almkvist, O.; Rodriguez-Vieitez, E.; Nordberg, A. Tau PET imaging in neurodegenerative tauopathies-still a challenge. Mol. Psychiatry 2019, 24, 1112–1134. [Google Scholar] [CrossRef]
  46. Vlassenko, A.G.; Benzinger, T.L.; Morris, J.C. PET amyloid-beta imaging in preclinical Alzheimer’s disease. Biochim. Biophys. Acta 2012, 1822, 370–379. [Google Scholar] [CrossRef]
  47. Gordon, B.A.; Blazey, T.M.; Su, Y.; Hari-Raj, A.; Dincer, A.; Flores, S.; Christensen, J.; McDade, E.; Wang, G.; Xiong, C.; et al. Spatial patterns of neuroimaging biomarker change in individuals from families with autosomal dominant Alzheimer’s disease: A longitudinal study. Lancet Neurol. 2018, 17, 241–250. [Google Scholar] [CrossRef]
  48. Hampel, H.; Nistico, R.; Seyfried, N.T.; Levey, A.I.; Modeste, E.; Lemercier, P.; Baldacci, F.; Toschi, N.; Garaci, F.; Perry, G.; et al. Omics sciences for systems biology in Alzheimer’s disease: State-of-the-art of the evidence. Ageing Res. Rev. 2021, 69, 101346. [Google Scholar] [CrossRef]
  49. Butterfield, D.A.; Reed, T.; Newman, S.F.; Sultana, R. Roles of amyloid beta-peptide-associated oxidative stress and brain protein modifications in the pathogenesis of Alzheimer’s disease and mild cognitive impairment. Free Radic. Biol. Med. 2007, 43, 658–677. [Google Scholar] [CrossRef]
  50. Fišar, Z.; Hroudová, J.; Hansiková, H.; Spáčilová, J.; Lelková, P.; Wenchich, L.; Jirák, R.; Zvěřová, M.; Zeman, J.; Martásek, P.; et al. Mitochondrial respiration in the platelets of patients with Alzheimer’s disease. Curr. Alzheimer Res. 2016, 13, 930–941. [Google Scholar] [CrossRef]
  51. 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. [Google Scholar] [CrossRef] [PubMed]
  52. Zheng, C.; Zhou, X.W.; Wang, J.Z. The dual roles of cytokines in Alzheimer’s disease: Update on interleukins, TNF-alpha, TGF-beta and IFN-gamma. Transl. Neurodegener. 2016, 5, 7. [Google Scholar] [CrossRef] [PubMed]
  53. Sperling, R.A.; Aisen, P.S.; Beckett, L.A.; Bennett, D.A.; Craft, S.; Fagan, A.M.; Iwatsubo, T.; Jack, C.R., Jr.; Kaye, J.; Montine, T.J.; et al. Toward defining the preclinical stages of Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s Dement. 2011, 7, 280–292. [Google Scholar] [CrossRef] [PubMed]
  54. Long, J.M.; Holtzman, D.M. Alzheimer Disease: An Update on Pathobiology and Treatment Strategies. Cell 2019, 179, 312–339. [Google Scholar] [CrossRef] [PubMed]
  55. Swerdlow, R.H.; Khan, S.M. A “mitochondrial cascade hypothesis” for sporadic Alzheimer’s disease. Med. Hypotheses 2004, 63, 8–20. [Google Scholar] [CrossRef] [PubMed]
  56. Swerdlow, R.H.; Burns, J.M.; Khan, S.M. The Alzheimer’s disease mitochondrial cascade hypothesis. J. Alzheimer’s Dis. 2010, 20 (Suppl. 2), S265–S279. [Google Scholar] [CrossRef]
  57. Swerdlow, R.H. Mitochondria and Mitochondrial Cascades in Alzheimer’s Disease. J. Alzheimer’s Dis. 2018, 62, 1403–1416. [Google Scholar] [CrossRef]
  58. Pires, M.; Rego, A.C. Apoe4 and Alzheimer’s Disease Pathogenesis-Mitochondrial Deregulation and Targeted Therapeutic Strategies. Int. J. Mol. Sci. 2023, 24, 778. [Google Scholar] [CrossRef]
  59. Yang, K.; Chen, Z.; Gao, J.; Shi, W.; Li, L.; Jiang, S.; Hu, H.; Liu, Z.; Xu, D.; Wu, L. The Key Roles of GSK-3beta in Regulating Mitochondrial Activity. Cell. Physiol. Biochem. 2017, 44, 1445–1459. [Google Scholar] [CrossRef] [PubMed]
  60. Beurel, E.; Grieco, S.F.; Jope, R.S. Glycogen synthase kinase-3 (GSK3): Regulation, actions, and diseases. Pharmacol. Ther. 2015, 148, 114–131. [Google Scholar] [CrossRef] [PubMed]
  61. Behl, T.; Kaur, D.; Sehgal, A.; Singh, S.; Sharma, N.; Zengin, G.; Andronie-Cioara, F.L.; Toma, M.M.; Bungau, S.; Bumbu, A.G. Role of Monoamine Oxidase Activity in Alzheimer’s Disease: An Insight into the Therapeutic Potential of Inhibitors. Molecules 2021, 26, 3724. [Google Scholar] [CrossRef] [PubMed]
  62. Lakatos, A.; Derbeneva, O.; Younes, D.; Keator, D.; Bakken, T.; Lvova, M.; Brandon, M.; Guffanti, G.; Reglodi, D.; Saykin, A.; et al. Association between mitochondrial DNA variations and Alzheimer’s disease in the ADNI cohort. Neurobiol. Aging 2010, 31, 1355–1363. [Google Scholar] [CrossRef] [PubMed]
  63. Chaturvedi, R.K.; Flint Beal, M. Mitochondrial diseases of the brain. Free Radic. Biol. Med. 2013, 63, 1–29. [Google Scholar] [CrossRef] [PubMed]
  64. Macdonald, R.; Barnes, K.; Hastings, C.; Mortiboys, H. Mitochondrial abnormalities in Parkinson’s disease and Alzheimer’s disease: Can mitochondria be targeted therapeutically? Biochem. Soc. Trans. 2018, 46, 891–909. [Google Scholar] [CrossRef] [PubMed]
  65. Shen, X.; Sun, P.; Zhang, H.; Yang, H. Mitochondrial quality control in the brain: The physiological and pathological roles. Front. Neurosci. 2022, 16, 1075141. [Google Scholar] [CrossRef] [PubMed]
  66. Lee, D.; Jo, M.G.; Kim, S.Y.; Chung, C.G.; Lee, S.B. Dietary Antioxidants and the Mitochondrial Quality Control: Their Potential Roles in Parkinson’s Disease Treatment. Antioxidants 2020, 9, 1056. [Google Scholar] [CrossRef]
  67. Du, H.; Guo, L.; Yan, S.; Sosunov, A.A.; McKhann, G.M.; Yan, S.S. Early deficits in synaptic mitochondria in an Alzheimer’s disease mouse model. Proc. Natl. Acad. Sci. USA 2010, 107, 18670–18675. [Google Scholar] [CrossRef]
  68. Du, H.; Yan, S.S. Mitochondrial permeability transition pore in Alzheimer’s disease: Cyclophilin D and amyloid beta. Biochim. Biophys. Acta 2010, 1802, 198–204. [Google Scholar] [CrossRef]
  69. Calkins, M.J.; Reddy, P.H. Amyloid beta impairs mitochondrial anterograde transport and degenerates synapses in Alzheimer’s disease neurons. Biochim. Biophys. Acta 2011, 1812, 507–513. [Google Scholar] [CrossRef]
  70. Pavlov, P.F.; Hansson Petersen, C.; Glaser, E.; Ankarcrona, M. Mitochondrial accumulation of APP and Abeta: Significance for Alzheimer disease pathogenesis. J. Cell. Mol. Med. 2009, 13, 4137–4145. [Google Scholar] [CrossRef]
  71. Pagani, L.; Eckert, A. Amyloid-Beta interaction with mitochondria. Int. J. Alzheimer’s Dis. 2011, 2011, 925050. [Google Scholar] [CrossRef] [PubMed]
  72. Supnet, C.; Bezprozvanny, I. The dysregulation of intracellular calcium in Alzheimer disease. Cell Calcium 2010, 47, 183–189. [Google Scholar] [CrossRef] [PubMed]
  73. Manczak, M.; Calkins, M.J.; Reddy, P.H. Impaired mitochondrial dynamics and abnormal interaction of amyloid beta with mitochondrial protein Drp1 in neurons from patients with Alzheimer’s disease: Implications for neuronal damage. Hum. Mol. Genet. 2011, 20, 2495–2509. [Google Scholar] [CrossRef] [PubMed]
  74. Manczak, M.; Reddy, P.H. Abnormal interaction of VDAC1 with amyloid beta and phosphorylated tau causes mitochondrial dysfunction in Alzheimer’s disease. Hum. Mol. Genet. 2012, 21, 5131–5146. [Google Scholar] [CrossRef]
  75. Yao, J.; Irwin, R.W.; Zhao, L.; Nilsen, J.; Hamilton, R.T.; Brinton, R.D. Mitochondrial bioenergetic deficit precedes Alzheimer’s pathology in female mouse model of Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2009, 106, 14670–14675. [Google Scholar] [CrossRef]
  76. Manczak, M.; Anekonda, T.S.; Henson, E.; Park, B.S.; Quinn, J.; Reddy, P.H. Mitochondria are a direct site of A beta accumulation in Alzheimer’s disease neurons: Implications for free radical generation and oxidative damage in disease progression. Hum. Mol. Genet. 2006, 15, 1437–1449. [Google Scholar] [CrossRef]
  77. Caspersen, C.; Wang, N.; Yao, J.; Sosunov, A.; Chen, X.; Lustbader, J.W.; Xu, H.W.; Stern, D.; McKhann, G.; Yan, S.D. Mitochondrial Aβ: A potential focal point for neuronal metabolic dysfunction in Alzheimer’s disease. FASEB J. 2005, 19, 2040–2041. [Google Scholar] [CrossRef]
  78. Rhein, V.; Song, X.; Wiesner, A.; Ittner, L.M.; Baysang, G.; Meier, F.; Ozmen, L.; Bluethmann, H.; Drose, S.; Brandt, U.; et al. Amyloid-beta and tau synergistically impair the oxidative phosphorylation system in triple transgenic Alzheimer’s disease mice. Proc. Natl. Acad. Sci. USA 2009, 106, 20057–20062. [Google Scholar] [CrossRef]
  79. Padurariu, M.; Ciobica, A.; Lefter, R.; Serban, I.L.; Stefanescu, C.; Chirita, R. The oxidative stress hypothesis in Alzheimer’s disease. Psychiatr. Danub. 2013, 25, 401–409. [Google Scholar]
  80. Miles, E.A.; Calder, P.C. Effects of Citrus Fruit Juices and Their Bioactive Components on Inflammation and Immunity: A Narrative Review. Front. Immunol. 2021, 12, 712608. [Google Scholar] [CrossRef]
  81. Plascencia-Villa, G.; Perry, G. Roles of Oxidative Stress in Synaptic Dysfunction and Neuronal Cell Death in Alzheimer’s Disease. Antioxidants 2023, 12, 1628. [Google Scholar] [CrossRef]
  82. Mattson, M.P. Roles of the lipid peroxidation product 4-hydroxynonenal in obesity, the metabolic syndrome, and associated vascular and neurodegenerative disorders. Exp. Gerontol. 2009, 44, 625–633. [Google Scholar] [CrossRef]
  83. Aberg, F.; Appelkvist, E.L.; Dallner, G.; Ernster, L. Distribution and redox state of ubiquinones in rat and human tissues. Arch. Biochem. Biophys. 1992, 295, 230–234. [Google Scholar] [CrossRef]
  84. Dallner, G.; Sindelar, P.J. Regulation of ubiquinone metabolism. Free Radic. Biol. Med. 2000, 29, 285–294. [Google Scholar] [CrossRef] [PubMed]
  85. Pradhan, N.; Singh, C.; Singh, A. Coenzyme Q10 a mitochondrial restorer for various brain disorders. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2021, 394, 2197–2222. [Google Scholar] [CrossRef] [PubMed]
  86. Staiano, C.; Garcia-Corzo, L.; Mantle, D.; Turton, N.; Millichap, L.E.; Brea-Calvo, G.; Hargreaves, I. Biosynthesis, Deficiency, and Supplementation of Coenzyme Q. Antioxidants 2023, 12, 1469. [Google Scholar] [CrossRef] [PubMed]
  87. Fernandez-Del-Rio, L.; Clarke, C.F. Coenzyme Q Biosynthesis: An Update on the Origins of the Benzenoid Ring and Discovery of New Ring Precursors. Metabolites 2021, 11, 385. [Google Scholar] [CrossRef] [PubMed]
  88. Stefely, J.A.; Pagliarini, D.J. Biochemistry of Mitochondrial Coenzyme Q Biosynthesis. Trends Biochem. Sci. 2017, 42, 824–843. [Google Scholar] [CrossRef] [PubMed]
  89. Barcelos, I.P.; Haas, R.H. CoQ10 and Aging. Biology 2019, 8, 28. [Google Scholar] [CrossRef] [PubMed]
  90. Bentinger, M.; Tekle, M.; Dallner, G. Coenzyme Q–biosynthesis and functions. Biochem. Biophys. Res. Commun. 2010, 396, 74–79. [Google Scholar] [CrossRef] [PubMed]
  91. Manzar, H.; Abdulhussein, D.; Yap, T.E.; Cordeiro, M.F. Cellular Consequences of Coenzyme Q10 Deficiency in Neurodegeneration of the Retina and Brain. Int. J. Mol. Sci. 2020, 21, 9299. [Google Scholar] [CrossRef] [PubMed]
  92. Ernster, L.; Dallner, G. Biochemical, physiological and medical aspects of ubiquinone function. Biochim. Biophys. Acta 1995, 1271, 195–204. [Google Scholar] [CrossRef] [PubMed]
  93. Bentinger, M.; Brismar, K.; Dallner, G. The antioxidant role of coenzyme Q. Mitochondrion 2007, 7, S41–S50. [Google Scholar] [CrossRef] [PubMed]
  94. Eriksson, E.K.; Agmo Hernandez, V.; Edwards, K. Effect of ubiquinone-10 on the stability of biomimetic membranes of relevance for the inner mitochondrial membrane. Biochim. Biophys. Acta Biomembr. 2018, 1860, 1205–1215. [Google Scholar] [CrossRef] [PubMed]
  95. Echtay, K.S.; Winkler, E.; Klingenberg, M. Coenzyme Q is an obligatory cofactor for uncoupling protein function. Nature 2000, 408, 609–613. [Google Scholar] [CrossRef] [PubMed]
  96. 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. [Google Scholar] [CrossRef]
  97. 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-kappaB signaling pathway after spinal cord injury in rats. Am. J. Transl. Res. 2019, 11, 6544–6552. [Google Scholar]
  98. Genova, M.L.; Lenaz, G. New developments on the functions of coenzyme Q in mitochondria. Biofactors 2011, 37, 330–354. [Google Scholar] [CrossRef]
  99. Hidalgo-Gutierrez, A.; Gonzalez-Garcia, P.; Diaz-Casado, M.E.; Barriocanal-Casado, E.; Lopez-Herrador, S.; Quinzii, C.M.; Lopez, L.C. Metabolic Targets of Coenzyme Q10 in Mitochondria. Antioxidants 2021, 10, 520. [Google Scholar] [CrossRef]
  100. Linnane, A.W.; Kios, M.; Vitetta, L. Coenzyme Q10—Its role as a prooxidant in the formation of superoxide anion/hydrogen peroxide and the regulation of the metabolome. Mitochondrion 2007, 7, S51–S61. [Google Scholar] [CrossRef]
  101. Hunte, C.; Palsdottir, H.; Trumpower, B.L. Protonmotive pathways and mechanisms in the cytochrome bc1 complex. FEBS Lett. 2003, 545, 39–46. [Google Scholar] [CrossRef] [PubMed]
  102. Crofts, A.R. The cytochrome bc1 complex: Function in the context of structure. Annu. Rev. Physiol. 2004, 66, 689–733. [Google Scholar] [CrossRef]
  103. Barragan, A.M.; Crofts, A.R.; Schulten, K.; Solov’yov, I.A. Identification of ubiquinol binding motifs at the Qo-site of the cytochrome bc1 complex. J. Phys. Chem. B 2015, 119, 433–447. [Google Scholar] [CrossRef] [PubMed]
  104. Fišar, Z.; Hroudová, J. Measurement of Mitochondrial Respiration in Platelets. Methods Mol. Biol. 2021, 2277, 269–276. [Google Scholar] [CrossRef]
  105. Hroudová, J.; Fišar, Z. Assessment of the Effects of Drugs on Mitochondrial Respiration. Methods Mol. Biol. 2021, 2277, 133–142. [Google Scholar] [CrossRef] [PubMed]
  106. Enriquez, J.A. Supramolecular Organization of Respiratory Complexes. Annu. Rev. Physiol. 2016, 78, 533–561. [Google Scholar] [CrossRef]
  107. Genova, M.L.; Lenaz, G. Functional role of mitochondrial respiratory supercomplexes. Biochim. Biophys. Acta 2014, 1837, 427–443. [Google Scholar] [CrossRef]
  108. Zhang, M.; Mileykovskaya, E.; Dowhan, W. Gluing the respiratory chain together. Cardiolipin is required for supercomplex formation in the inner mitochondrial membrane. J. Biol. Chem. 2002, 277, 43553–43556. [Google Scholar] [CrossRef]
  109. Lapuente-Brun, E.; Moreno-Loshuertos, R.; Acin-Perez, R.; Latorre-Pellicer, A.; Colas, C.; Balsa, E.; Perales-Clemente, E.; Quiros, P.M.; Calvo, E.; Rodriguez-Hernandez, M.A.; et al. Supercomplex assembly determines electron flux in the mitochondrial electron transport chain. Science 2013, 340, 1567–1570. [Google Scholar] [CrossRef]
  110. Lenaz, G.; Genova, M.L. Supramolecular organisation of the mitochondrial respiratory chain: A new challenge for the mechanism and control of oxidative phosphorylation. Adv. Exp. Med. Biol. 2012, 748, 107–144. [Google Scholar] [CrossRef]
  111. Moreno-Loshuertos, R.; Enriquez, J.A. Respiratory supercomplexes and the functional segmentation of the CoQ pool. Free Radic. Biol. Med. 2016, 100, 5–13. [Google Scholar] [CrossRef]
  112. Hernansanz-Agustin, P.; Enriquez, J.A. Functional segmentation of CoQ and cyt c pools by respiratory complex superassembly. Free Radic. Biol. Med. 2021, 167, 232–242. [Google Scholar] [CrossRef] [PubMed]
  113. Hackenbrock, C.R.; Chazotte, B.; Gupte, S.S. The random collision model and a critical assessment of diffusion and collision in mitochondrial electron transport. J. Bioenerg. Biomembr. 1986, 18, 331–368. [Google Scholar] [CrossRef] [PubMed]
  114. Schagger, H.; Pfeiffer, K. Supercomplexes in the respiratory chains of yeast and mammalian mitochondria. EMBO J. 2000, 19, 1777–1783. [Google Scholar] [CrossRef] [PubMed]
  115. Cogliati, S.; Cabrera-Alarcon, J.L.; Enriquez, J.A. Regulation and functional role of the electron transport chain supercomplexes. Biochem. Soc. Trans. 2021, 49, 2655–2668. [Google Scholar] [CrossRef]
  116. Enriquez, J.A.; Lenaz, G. Coenzyme q and the respiratory chain: Coenzyme q pool and mitochondrial supercomplexes. Mol. Syndromol. 2014, 5, 119–140. [Google Scholar] [CrossRef]
  117. Acin-Perez, R.; Enriquez, J.A. The function of the respiratory supercomplexes: The plasticity model. Biochim. Biophys. Acta 2014, 1837, 444–450. [Google Scholar] [CrossRef] [PubMed]
  118. Lenaz, G.; Genova, M.L. Kinetics of integrated electron transfer in the mitochondrial respiratory chain: Random collisions vs. solid state electron channeling. Am. J. Physiol. Cell Physiol. 2007, 292, C1221–C1239. [Google Scholar] [CrossRef]
  119. Sheykhhasan, M.; Amini, R.; Soleimani Asl, S.; Saidijam, M.; Hashemi, S.M.; Najafi, R. Neuroprotective effects of coenzyme Q10-loaded exosomes obtained from adipose-derived stem cells in a rat model of Alzheimer’s disease. Biomed. Pharmacother. 2022, 152, 113224. [Google Scholar] [CrossRef]
  120. Fišar, Z.; Hroudová, J.; Singh, N.; Kopřivová, A.; Macečková, D. Effect of Simvastatin, Coenzyme Q10, Resveratrol, Acetylcysteine and Acetylcarnitine on Mitochondrial Respiration. Folia Biol. 2016, 62, 53–66. [Google Scholar]
  121. Cardoso, S.; Carvalho, C.; Correia, S.C.; Seica, R.M.; Moreira, P.I. Alzheimer’s Disease: From Mitochondrial Perturbations to Mitochondrial Medicine. Brain Pathol. 2016, 26, 632–647. [Google Scholar] [CrossRef]
  122. Bosetti, F.; Brizzi, F.; Barogi, S.; Mancuso, M.; Siciliano, G.; Tendi, E.A.; Murri, L.; Rapoport, S.I.; Solaini, G. Cytochrome c oxidase and mitochondrial F1F0-ATPase (ATP synthase) activities in platelets and brain from patients with Alzheimer’s disease. Neurobiol. Aging 2002, 23, 371–376. [Google Scholar] [CrossRef]
  123. Cardoso, S.M.; Santana, I.; Swerdlow, R.H.; Oliveira, C.R. Mitochondria dysfunction of Alzheimer’s disease cybrids enhances Abeta toxicity. J. Neurochem. 2004, 89, 1417–1426. [Google Scholar] [CrossRef]
  124. Fišar, Z.; Jirák, R.; Zvěřová, M.; Setnička, V.; Habartová, L.; Hroudová, J.; Vaníčková, Z.; Raboch, J. Plasma amyloid beta levels and platelet mitochondrial respiration in patients with Alzheimer’s disease. Clin. Biochem. 2019, 72, 71–80. [Google Scholar] [CrossRef]
  125. Choi, H.; Park, H.H.; Koh, S.H.; Choi, N.Y.; Yu, H.J.; Park, J.; Lee, Y.J.; Lee, K.Y. Coenzyme Q10 protects against amyloid beta-induced neuronal cell death by inhibiting oxidative stress and activating the P13K pathway. Neurotoxicology 2012, 33, 85–90. [Google Scholar] [CrossRef]
  126. Yamagishi, K.; Ikeda, A.; Moriyama, Y.; Chei, C.L.; Noda, H.; Umesawa, M.; Cui, R.; Nagao, M.; Kitamura, A.; Yamamoto, Y.; et al. Serum coenzyme Q10 and risk of disabling dementia: The Circulatory Risk in Communities Study (CIRCS). Atherosclerosis 2014, 237, 400–403. [Google Scholar] [CrossRef] [PubMed]
  127. Santa-Mara, I.; Santpere, G.; MacDonald, M.J.; Gomez de Barreda, E.; Hernandez, F.; Moreno, F.J.; Ferrer, I.; Avila, J. Coenzyme q induces tau aggregation, tau filaments, and Hirano bodies. J. Neuropathol. Exp. Neurol. 2008, 67, 428–434. [Google Scholar] [CrossRef] [PubMed]
  128. Jimenez-Jimenez, F.J.; Alonso-Navarro, H.; Garcia-Martin, E.; Agundez, J.A.G. Coenzyme Q10 and Dementia: A Systematic Review. Antioxidants 2023, 12, 533. [Google Scholar] [CrossRef]
  129. Hampel, H.; Hardy, J.; Blennow, K.; Chen, C.; Perry, G.; Kim, S.H.; Villemagne, V.L.; Aisen, P.; Vendruscolo, M.; Iwatsubo, T.; et al. The Amyloid-beta Pathway in Alzheimer’s Disease. Mol. Psychiatry 2021, 26, 5481–5503. [Google Scholar] [CrossRef] [PubMed]
  130. Frontinan-Rubio, J.; Rabanal-Ruiz, Y.; Duran-Prado, M.; Alcain, F.J. The Protective Effect of Ubiquinone against the Amyloid Peptide in Endothelial Cells Is Isoprenoid Chain Length-Dependent. Antioxidants 2021, 10, 1806. [Google Scholar] [CrossRef] [PubMed]
  131. Cummings, J.; Aisen, P.S.; DuBois, B.; Frolich, L.; Jack, C.R., Jr.; Jones, R.W.; Morris, J.C.; Raskin, J.; Dowsett, S.A.; Scheltens, P. Drug development in Alzheimer’s disease: The path to 2025. Alzheimer’s Res. Ther. 2016, 8, 39. [Google Scholar] [CrossRef]
  132. Ishrat, T.; Khan, M.B.; Hoda, M.N.; Yousuf, S.; Ahmad, M.; Ansari, M.A.; Ahmad, A.S.; Islam, F. Coenzyme Q10 modulates cognitive impairment against intracerebroventricular injection of streptozotocin in rats. Behav. Brain Res. 2006, 171, 9–16. [Google Scholar] [CrossRef]
  133. Llanos-Gonzalez, E.; Sancho-Bielsa, F.J.; Frontinan-Rubio, J.; Rabanal-Ruiz, Y.; Garcia-Carpintero, S.; Chicano, E.; Ubeda-Banon, I.; Flores-Cuadrado, A.; Gimenez-Llort, L.; Alcain, F.J.; et al. Spatial and Temporal Protein Modules Signatures Associated with Alzheimer Disease in 3xTg-AD Mice Are Restored by Early Ubiquinol Supplementation. Antioxidants 2023, 12, 747. [Google Scholar] [CrossRef] [PubMed]
  134. Duran-Prado, M.; Frontinan, J.; Santiago-Mora, R.; Peinado, J.R.; Parrado-Fernandez, C.; Gomez-Almagro, M.V.; Moreno, M.; Lopez-Dominguez, J.A.; Villalba, J.M.; Alcain, F.J. Coenzyme Q10 protects human endothelial cells from beta-amyloid uptake and oxidative stress-induced injury. PLoS ONE 2014, 9, e109223. [Google Scholar] [CrossRef] [PubMed]
  135. Galasko, D.R.; Peskind, E.; Clark, C.M.; Quinn, J.F.; Ringman, J.M.; Jicha, G.A.; Cotman, C.; Cottrell, B.; Montine, T.J.; Thomas, R.G.; et al. Antioxidants for Alzheimer disease: A randomized clinical trial with cerebrospinal fluid biomarker measures. Arch. Neurol. 2012, 69, 836–841. [Google Scholar] [CrossRef]
  136. Testai, L.; Martelli, A.; Flori, L.; Cicero, A.F.G.; Colletti, A. Coenzyme Q(10): Clinical Applications beyond Cardiovascular Diseases. Nutrients 2021, 13, 1697. [Google Scholar] [CrossRef] [PubMed]
  137. Jiang, X.; Guo, Y.; Cui, L.; Huang, L.; Guo, Q.; Huang, G. Study of Diet Habits and Cognitive Function in the Chinese Middle-Aged and Elderly Population: The Association between Folic Acid, B Vitamins, Vitamin D, Coenzyme Q10 Supplementation and Cognitive Ability. Nutrients 2023, 15, 1243. [Google Scholar] [CrossRef]
  138. Kleinová, L.; Cerman, J.; Hlávka, J.; Hort, J. New pharmacological options in the treatment of Alzheimer’s disease (Nové farmakologické možnosti v léčbě Alzheimerovy nemoci). Ceska a Slovenska Neurologie a Neurochirurgie 2022, 85, 462–469. [Google Scholar] [CrossRef]
  139. Butler, M.; Nelson, V.A.; Davila, H.; Ratner, E.; Fink, H.A.; Hemmy, L.S.; McCarten, J.R.; Barclay, T.R.; Brasure, M.; Kane, R.L. Over-the-Counter Supplement Interventions to Prevent Cognitive Decline, Mild Cognitive Impairment, and Clinical Alzheimer-Type Dementia: A Systematic Review. Ann. Intern. Med. 2018, 168, 52–62. [Google Scholar] [CrossRef]
  140. Cenini, G.; Voos, W. Mitochondria as Potential Targets in Alzheimer Disease Therapy: An Update. Front. Pharmacol. 2019, 10, 902. [Google Scholar] [CrossRef]
  141. Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative Stress: A Key Modulator in Neurodegenerative Diseases. Molecules 2019, 24, 1583. [Google Scholar] [CrossRef]
  142. Ristow, M.; Schmeisser, S. Extending life span by increasing oxidative stress. Free Radic. Biol. Med. 2011, 51, 327–336. [Google Scholar] [CrossRef]
  143. DiMauro, S.; Quinzii, C.M.; Hirano, M. Mutations in coenzyme Q10 biosynthetic genes. J. Clin. Investig. 2007, 117, 587–589. [Google Scholar] [CrossRef]
  144. Singh, R.B.; Niaz, M.A.; Kumar, A.; Sindberg, C.D.; Moesgaard, S.; Littarru, G.P. Effect on absorption and oxidative stress of different oral Coenzyme Q10 dosages and intake strategy in healthy men. Biofactors 2005, 25, 219–224. [Google Scholar] [CrossRef]
  145. Ehrenhaus Masotta, N.; Hocht, C.; Contin, M.; Lucangioli, S.; Rojas, A.M.; Tripodi, V.P. Bioavailability of coenzyme Q(10) loaded in an oleogel formulation for oral therapy: Comparison with a commercial-grade solid formulation. Int. J. Pharm. 2020, 582, 119315. [Google Scholar] [CrossRef]
  146. Pastor-Maldonado, C.J.; Suarez-Rivero, J.M.; Povea-Cabello, S.; Alvarez-Cordoba, M.; Villalon-Garcia, I.; Munuera-Cabeza, M.; Suarez-Carrillo, A.; Talaveron-Rey, M.; Sanchez-Alcazar, J.A. Coenzyme Q(10): Novel Formulations and Medical Trends. Int. J. Mol. Sci. 2020, 21, 8432. [Google Scholar] [CrossRef] [PubMed]
  147. Pravst, I.; Rodriguez Aguilera, J.C.; Cortes Rodriguez, A.B.; Jazbar, J.; Locatelli, I.; Hristov, H.; Zmitek, K. Comparative Bioavailability of Different Coenzyme Q10 Formulations in Healthy Elderly Individuals. Nutrients 2020, 12, 784. [Google Scholar] [CrossRef]
  148. Takahashi, M.; Takahashi, K. Water-soluble CoQ10 as A Promising Anti-aging Agent for Neurological Dysfunction in Brain Mitochondria. Antioxidants 2019, 8, 61. [Google Scholar] [CrossRef] [PubMed]
  149. Bhagavan, H.N.; Chopra, R.K. Plasma coenzyme Q10 response to oral ingestion of coenzyme Q10 formulations. Mitochondrion 2007, 7, S78–S88. [Google Scholar] [CrossRef] [PubMed]
  150. Suarez-Rivero, J.M.; Pastor-Maldonado, C.J.; Povea-Cabello, S.; Alvarez-Cordoba, M.; Villalon-Garcia, I.; Munuera-Cabeza, M.; Suarez-Carrillo, A.; Talaveron-Rey, M.; Sanchez-Alcazar, J.A. Coenzyme Q(10) Analogues: Benefits and Challenges for Therapeutics. Antioxidants 2021, 10, 236. [Google Scholar] [CrossRef]
  151. Bhagavan, H.N.; Chopra, R.K. Coenzyme Q10: Absorption, tissue uptake, metabolism and pharmacokinetics. Free Radic. Res. 2006, 40, 445–453. [Google Scholar] [CrossRef] [PubMed]
  152. Mantle, D.; Dybring, A. Bioavailability of Coenzyme Q(10): An Overview of the Absorption Process and Subsequent Metabolism. Antioxidants 2020, 9, 386. [Google Scholar] [CrossRef] [PubMed]
  153. Smith, K.M.; Matson, S.; Matson, W.R.; Cormier, K.; Del Signore, S.J.; Hagerty, S.W.; Stack, E.C.; Ryu, H.; Ferrante, R.J. Dose ranging and efficacy study of high-dose coenzyme Q10 formulations in Huntington’s disease mice. Biochim. Biophys. Acta 2006, 1762, 616–626. [Google Scholar] [CrossRef]
  154. Belousova, M.; Tokareva, O.G.; Gorodetskaya, E.; Kalenikova, E.I.; Medvedev, O.S. 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. [Google Scholar] [CrossRef] [PubMed]
  155. Kamzalov, S.; Sumien, N.; Forster, M.J.; Sohal, R.S. Coenzyme Q intake elevates the mitochondrial and tissue levels of Coenzyme Q and alpha-tocopherol in young mice. J. Nutr. 2003, 133, 3175–3180. [Google Scholar] [CrossRef] [PubMed]
  156. Matthews, R.T.; Yang, L.; Browne, S.; Baik, M.; Beal, M.F. Coenzyme Q10 administration increases brain mitochondrial concentrations and exerts neuroprotective effects. Proc. Natl. Acad. Sci. USA 1998, 95, 8892–8897. [Google Scholar] [CrossRef]
  157. Kwong, L.K.; Kamzalov, S.; Rebrin, I.; Bayne, A.C.; Jana, C.K.; Morris, P.; Forster, M.J.; Sohal, R.S. 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. [Google Scholar] [CrossRef]
  158. Acosta, M.J.; Vazquez Fonseca, L.; Desbats, M.A.; Cerqua, C.; Zordan, R.; Trevisson, E.; Salviati, L. Coenzyme Q biosynthesis in health and disease. Biochim. Biophys. Acta 2016, 1857, 1079–1085. [Google Scholar] [CrossRef]
  159. Beal, M.F. Mitochondrial dysfunction and oxidative damage in Alzheimer’s and Parkinson’s diseases and coenzyme Q10 as a potential treatment. J. Bioenerg. Biomembr. 2004, 36, 381–386. [Google Scholar] [CrossRef]
  160. Ebrahimi, A.; Kamyab, A.; Hosseini, S.; Ebrahimi, S.; Ashkani-Esfahani, S. Involvement of Coenzyme Q10 in Various Neurodegenerative and Psychiatric Diseases. Biochem. Res. Int. 2023, 2023, 5510874. [Google Scholar] [CrossRef]
  161. Dumont, M.; Kipiani, K.; Yu, F.; Wille, E.; Katz, M.; Calingasan, N.Y.; Gouras, G.K.; Lin, M.T.; Beal, M.F. Coenzyme Q10 decreases amyloid pathology and improves behavior in a transgenic mouse model of Alzheimer’s disease. J. Alzheimer’s Dis. 2011, 27, 211–223. [Google Scholar] [CrossRef]
  162. Komaki, H.; Faraji, N.; Komaki, A.; Shahidi, S.; Etaee, F.; Raoufi, S.; Mirzaei, F. Investigation of protective effects of coenzyme Q10 on impaired synaptic plasticity in a male rat model of Alzheimer’s disease. Brain Res. Bull. 2019, 147, 14–21. [Google Scholar] [CrossRef]
  163. Mantle, D.; Heaton, R.A.; Hargreaves, I.P. Coenzyme Q10, Ageing and the Nervous System: An Overview. Antioxidants 2021, 11, 2. [Google Scholar] [CrossRef]
  164. Hooff, G.P.; Wood, W.G.; Muller, W.E.; Eckert, G.P. Isoprenoids, small GTPases and Alzheimer’s disease. Biochim. Biophys. Acta 2010, 1801, 896–905. [Google Scholar] [CrossRef]
  165. Raizner, A.E. Coenzyme Q(10). Methodist. Debakey Cardiovasc. J. 2019, 15, 185–191. [Google Scholar] [CrossRef] [PubMed]
  166. Gueven, N.; Ravishankar, P.; Eri, R.; Rybalka, E. Idebenone: When an antioxidant is not an antioxidant. Redox Biol. 2021, 38, 101812. [Google Scholar] [CrossRef]
  167. Rauchova, 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. [Google Scholar] [CrossRef]
  168. 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. [Google Scholar] [CrossRef] [PubMed]
  169. Thal, L.J.; Grundman, M.; Berg, J.; Ernstrom, K.; Margolin, R.; Pfeiffer, E.; Weiner, M.F.; Zamrini, E.; Thomas, R.G. Idebenone treatment fails to slow cognitive decline in Alzheimer’s disease. Neurology 2003, 61, 1498–1502. [Google Scholar] [CrossRef]
  170. Murphy, M.P.; Smith, R.A. Targeting antioxidants to mitochondria by conjugation to lipophilic cations. Annu. Rev. Pharmacol. Toxicol. 2007, 47, 629–656. [Google Scholar] [CrossRef] [PubMed]
  171. Ng, L.F.; Gruber, J.; Cheah, I.K.; Goo, C.K.; Cheong, W.F.; Shui, G.; Sit, K.P.; Wenk, M.R.; Halliwell, B. The mitochondria-targeted antioxidant MitoQ extends lifespan and improves healthspan of a transgenic Caenorhabditis elegans model of Alzheimer disease. Free Radic. Biol. Med. 2014, 71, 390–401. [Google Scholar] [CrossRef] [PubMed]
  172. Manczak, M.; Mao, P.; Calkins, M.J.; Cornea, A.; Reddy, A.P.; Murphy, M.P.; Szeto, H.H.; Park, B.; Reddy, P.H. Mitochondria-targeted antioxidants protect against amyloid-beta toxicity in Alzheimer’s disease neurons. J. Alzheimer’s Dis. 2010, 20 (Suppl. 2), S609–S631. [Google Scholar] [CrossRef] [PubMed]
  173. McManus, M.J.; Murphy, M.P.; Franklin, J.L. 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. [Google Scholar] [CrossRef]
  174. Young, M.L.; Franklin, J.L. The mitochondria-targeted antioxidant MitoQ inhibits memory loss, neuropathology, and extends lifespan in aged 3xTg-AD mice. Mol. Cell. Neurosci. 2019, 101, 103409. [Google Scholar] [CrossRef] [PubMed]
  175. Lenaz, G.; Fato, R.; Di Bernardo, S.; Jarreta, D.; Costa, A.; Genova, M.L.; Parenti Castelli, G. Localization and mobility of coenzyme Q in lipid bilayers and membranes. Biofactors 1999, 9, 87–93. [Google Scholar] [CrossRef] [PubMed]
  176. 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. [Google Scholar] [CrossRef]
  177. Onur, S.; Niklowitz, P.; Fischer, A.; Metges, C.C.; Grune, T.; Menke, T.; Rimbach, G.; Doring, F. A comparative study into alterations of coenzyme Q redox status in ageing pigs, mice, and worms. Biofactors 2014, 40, 346–354. [Google Scholar] [CrossRef]
  178. Niklowitz, P.; Onur, S.; Fischer, A.; Laudes, M.; Palussen, M.; Menke, T.; Doring, 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. [Google Scholar] [CrossRef]
  179. Nagase, M.; Yamamoto, Y.; Mitsui, J.; Tsuji, S. Simultaneous detection of reduced and oxidized forms of coenzyme Q10 in human cerebral spinal fluid as a potential marker of oxidative stress. J. Clin. Biochem. Nutr. 2018, 63, 205–210. [Google Scholar] [CrossRef]
  180. Fišar, Z.; Hroudová, J.; Zvěřová, M.; Jirák, R.; Raboch, J.; Kitzlerová, E. Age-Dependent Alterations in Platelet Mitochondrial Respiration. Biomedicines 2023, 11, 1564. [Google Scholar] [CrossRef]
  181. Hidaka, T.; Fujii, K.; Funahashi, I.; Fukutomi, N.; Hosoe, K. Safety assessment of coenzyme Q10 (CoQ10). Biofactors 2008, 32, 199–208. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Biological hypotheses of Alzheimer’s disease. Solid arrow shows direct effect, dotted arrow shows indirect effect of coenzyme Q10 (CoQ10).
Figure 1. Biological hypotheses of Alzheimer’s disease. Solid arrow shows direct effect, dotted arrow shows indirect effect of coenzyme Q10 (CoQ10).
Antioxidants 13 00191 g001
Figure 2. Simplified scheme of Alzheimer’s disease (AD) pathophysiology, with focus on the role of mitochondrial dysfunction. AD pathophysiology is associated with amyloid beta (Aβ) pathology (neurotoxicity of Aβ oligomers and plaques), tau pathology (neurotoxicity of tau oligomers and neurofibrillary tangles), mitochondrial dysfunction, oxidative stress, neuroinflammation, and loss of proteostasis. All these processes are interrelated and result in cellular and synaptic dysfunction, impaired neuroplasticity and neurochemistry, neurodegeneration (synaptic and neuronal loss and brain atrophy), cognitive decline, and AD dementia. Mitochondrial dysfunction associated with Aβ and tau pathology in AD includes decreased ATP production, mitophagy, biogenesis (peroxisome PGC-1α), activity of components of the OXPHOS system and other enzymes, mitochondrial membrane potential (Δψm), and import of mitochondrial proteins, imbalance of mitochondrial dynamics (DRP-1), impaired of intracellular Ca2+ homeostasis, membrane damage, interaction with ANT1 and VDAC1, and increased ROS production, apoptosis, and mPTP opening. Coenzyme Q10 (CoQ10), as a unique endogenous antioxidant and electron transporter in the OXPHOS system, may have a significant role in the pathophysiology and treatment of AD, primarily through the regulation of mitochondrial function. ANT1—adenine nucleotide translocator 1; ApoE4—apolipoprotein E4; APP—amyloid precursor protein; DRP-1—dynamin-like protein-1; HSD10—17β-hydroxysteroid dehydrogenase type 10; mPTP—mitochondrial permeability transition pore; NFTs—neurofibrillary tangles; OXPHOS—oxidative phosphorylation; PGC-1α—peroxisome proliferator-activated receptor gamma coactivator 1-alpha; P-tau—phosphorylated tau; ROS—reactive oxygen species; VDAC1—voltage-dependent anion channel 1.
Figure 2. Simplified scheme of Alzheimer’s disease (AD) pathophysiology, with focus on the role of mitochondrial dysfunction. AD pathophysiology is associated with amyloid beta (Aβ) pathology (neurotoxicity of Aβ oligomers and plaques), tau pathology (neurotoxicity of tau oligomers and neurofibrillary tangles), mitochondrial dysfunction, oxidative stress, neuroinflammation, and loss of proteostasis. All these processes are interrelated and result in cellular and synaptic dysfunction, impaired neuroplasticity and neurochemistry, neurodegeneration (synaptic and neuronal loss and brain atrophy), cognitive decline, and AD dementia. Mitochondrial dysfunction associated with Aβ and tau pathology in AD includes decreased ATP production, mitophagy, biogenesis (peroxisome PGC-1α), activity of components of the OXPHOS system and other enzymes, mitochondrial membrane potential (Δψm), and import of mitochondrial proteins, imbalance of mitochondrial dynamics (DRP-1), impaired of intracellular Ca2+ homeostasis, membrane damage, interaction with ANT1 and VDAC1, and increased ROS production, apoptosis, and mPTP opening. Coenzyme Q10 (CoQ10), as a unique endogenous antioxidant and electron transporter in the OXPHOS system, may have a significant role in the pathophysiology and treatment of AD, primarily through the regulation of mitochondrial function. ANT1—adenine nucleotide translocator 1; ApoE4—apolipoprotein E4; APP—amyloid precursor protein; DRP-1—dynamin-like protein-1; HSD10—17β-hydroxysteroid dehydrogenase type 10; mPTP—mitochondrial permeability transition pore; NFTs—neurofibrillary tangles; OXPHOS—oxidative phosphorylation; PGC-1α—peroxisome proliferator-activated receptor gamma coactivator 1-alpha; P-tau—phosphorylated tau; ROS—reactive oxygen species; VDAC1—voltage-dependent anion channel 1.
Antioxidants 13 00191 g002
Figure 3. Basic steps in coenzyme Q10 biosynthesis and three redox isoforms. HMG-CoA—β-Hydroxy β-methylglutaryl-coenzyme A.
Figure 3. Basic steps in coenzyme Q10 biosynthesis and three redox isoforms. HMG-CoA—β-Hydroxy β-methylglutaryl-coenzyme A.
Antioxidants 13 00191 g003
Figure 4. A simplified diagram of the mitochondrial electron transport system (ETS) with electron transfer in the Q-cycle of complex III. Electrons enter the ETS via complex I or via complex II and are transferred by coenzyme Q10 (CoQ10) to complex III. CoQ10 can also transfer electrons from dehydrogenases (DH) located on the outer or inner surface of the inner mitochondrial membrane (IMM). In the Qo-site complex III near the outer side of the IMM, two electrons from one ubiquinol (CoQ10(H2)), and subsequently two more electrons from the second ubiquinol, pass into two bifurcated transfer chains: (1) The acceptor of the two electrons is the iron –sulfur cluster (Fe2S2 center) of the Rieske protein, which passes electrons via cytochrome c1 (cyt c1) to cytochrome c (cyt c) on the outer surface of the IMM. Cyt c transfers electrons to complex IV (cytochrome c oxidase), where oxygen is finally reduced to water. (2) The acceptor of the second two electrons in the Q-cycle is cytochrome b containing low (cyt bL) and high (cyt bH) potential hemes. This chain supplies electrons to the Qi-site at the matrix side of the IMM, where they reduce one ubiquinone (CoQ10) to semiquinone and then to ubiquinol. Complexes I and III (Qo-site) are the sources of superoxide (O2•–).
Figure 4. A simplified diagram of the mitochondrial electron transport system (ETS) with electron transfer in the Q-cycle of complex III. Electrons enter the ETS via complex I or via complex II and are transferred by coenzyme Q10 (CoQ10) to complex III. CoQ10 can also transfer electrons from dehydrogenases (DH) located on the outer or inner surface of the inner mitochondrial membrane (IMM). In the Qo-site complex III near the outer side of the IMM, two electrons from one ubiquinol (CoQ10(H2)), and subsequently two more electrons from the second ubiquinol, pass into two bifurcated transfer chains: (1) The acceptor of the two electrons is the iron –sulfur cluster (Fe2S2 center) of the Rieske protein, which passes electrons via cytochrome c1 (cyt c1) to cytochrome c (cyt c) on the outer surface of the IMM. Cyt c transfers electrons to complex IV (cytochrome c oxidase), where oxygen is finally reduced to water. (2) The acceptor of the second two electrons in the Q-cycle is cytochrome b containing low (cyt bL) and high (cyt bH) potential hemes. This chain supplies electrons to the Qi-site at the matrix side of the IMM, where they reduce one ubiquinone (CoQ10) to semiquinone and then to ubiquinol. Complexes I and III (Qo-site) are the sources of superoxide (O2•–).
Antioxidants 13 00191 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fišar, Z.; Hroudová, J. CoQ10 and Mitochondrial Dysfunction in Alzheimer’s Disease. Antioxidants 2024, 13, 191. https://doi.org/10.3390/antiox13020191

AMA Style

Fišar Z, Hroudová J. CoQ10 and Mitochondrial Dysfunction in Alzheimer’s Disease. Antioxidants. 2024; 13(2):191. https://doi.org/10.3390/antiox13020191

Chicago/Turabian Style

Fišar, Zdeněk, and Jana Hroudová. 2024. "CoQ10 and Mitochondrial Dysfunction in Alzheimer’s Disease" Antioxidants 13, no. 2: 191. https://doi.org/10.3390/antiox13020191

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

Article Metrics

Back to TopTop