Skip to main content
Current Neuropharmacology logoLink to Current Neuropharmacology
. 2023 Apr 12;21(5):1165–1183. doi: 10.2174/1570159X20666220830112408

Mitochondrial Medicine: A Promising Therapeutic Option Against Various Neurodegenerative Disorders

Mohannad A Almikhlafi 1,#, Mohammed M Karami 2,#, Ankit Jana 3, Thamer M Alqurashi 4, Mohammed Majrashi 5, Badrah S Alghamdi 6,7,8,*, Ghulam Md Ashraf 9,*
PMCID: PMC10286591  PMID: 36043795

Abstract

Abnormal mitochondrial morphology and metabolic dysfunction have been observed in many neurodegenerative disorders (NDDs). Mitochondrial dysfunction can be caused by aberrant mitochondrial DNA, mutant nuclear proteins that interact with mitochondria directly or indirectly, or for unknown reasons. Since mitochondria play a significant role in neurodegeneration, mitochondria-targeted therapies represent a prosperous direction for the development of novel drug compounds that can be used to treat NDDs. This review gives a brief description of how mitochondrial abnormalities lead to various NDDs such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and amyotrophic lateral sclerosis. We further explore the promising therapeutic effectiveness of mitochondria-directed antioxidants, MitoQ, MitoVitE, MitoPBN, and dimebon. We have also discussed the possibility of mitochondrial gene therapy as a therapeutic option for these NDDs.

Keywords: Alzheimer’s disease, amyotrophic lateral sclerosis, gene therapy, Huntington’s disease, mitochondrial dysfunction, Parkinson’s disease

1. INTRODUCTION

The brain, despite accounting for just a small portion of our total body weight, is the greatest source of power, responsible for roughly 20% of total oxygen metabolism. Out of this, neurons are reported to utilize between 75-80% [1]. This energy is mostly used at the synapse, with a major percentage going towards restoring the depolarized neuronal membrane potentials. This high energy demand is constant, and even short durations of oxygen or glucose deprivation can cause neuronal demise [2].

Mitochondria not only provide energy to cells, but they also play an important role in cell signaling that is critical for cellular function. They can directly dictate cell survival by controlling physiological processes such as calcium homeostasis, cell proliferation, differentiation, cell cycle, protein synthesis, amino acids metabolism, and apoptosis signaling [3, 4]. Mitochondria, along with producing ATP, conduct critical metabolic activities and are decisive of cell death and survival. They serve as a nexus for apoptosis signals generated together by external and internal signals. As a result, the mitochondria occupy dominant importance in the cellular organelle hierarchy, allowing them to either encourage or conclude the cell's healthy life [5-7]. Mitochondria are required for neuronal activity due to neurons' inadequate glycolytic capability, which renders them extremely reliant on aerobic oxidative phosphorylation (OXPHOS) for energy [2, 8, 9]. OXPHOS, on the other hand, is a key source of hazardous endogenous free radicals such as hydrogen peroxide (H2O2), hydroxyl radical (.OH), and superoxide (O2.-) radical, which are all produced during regular cell respiration. When the electron transport chain (ETC) is blocked, electrons pile up in complex I and coenzyme Q, where they are transferred effectively to molecular oxygen to produce O2.-, which can then be detoxified by mitochondrial manganese superoxide dismutase (MnSOD) to produce H2O2, which in turn is transformed to H2O by glutathione peroxidase (GPx). Nevertheless, O2.- in the company of nitric oxide (NO.) produced by nitric oxide synthase (NOS) during the conversion of arginine to citrulline can result in the creation of peroxynitrite (ONOO-). Moreover, Fenton and/or Haber–Weiss reactions can turn H2O2 hazardous .OH in the vicinity of reduced transition metals. The accumulation of potentially harmful quantities of reactive oxygen species (ROS) is an outcome of normal mitochondrial respiration and homeostasis [10]. ROS and reactive nitrogen species (RNS) are well understood to serve a dual role as they can be useful as well as destructive to biological systems [11]. Oxidative stress arises when the level of free radical species generated exceeds the cells' ability to counteract them, resulting in mitochondrial malfunction and neurological damage. Mitochondrial reactive species have a variety of cellular targets, such as mitochondrial elements (lipids, proteins, and DNA). Because mitochondrial DNA (mtDNA) lacks histones and has a limited ability for DNA repair, it is an extremely sensitive target for oxidative stress [12]. Several recent reports have suggested that mitochondrially generated ROS may have a role in the initiation and advancement of neurodegenerative disorders (NDDs) in the elderly population [13-18]. The emerging recognition that mitochondria are at the crossroads of a cell's life and death, as evidenced by the role of mitochondrial injury in a variety of diseases, has made mitochondria an attractive choice for drug development. The intimate link between mitochondrial dysfunction and neurodegeneration has been discussed in this review. Moreover, we have explored some mitochondria-targeted therapies namely redox therapy, mitochondrial gene therapy, triphenylphosphonium (TPP) cation-based antioxidants, MitoQ, MitoVitE, MitoPBN, dimebon, etc.

2. MITOCHONDRIAL BIOGENESIS

Biogenesis of the mitochondria is the course through which new mitochondria are generated from pre-existing mitochondria since they cannot be generated de novo. Mitochondrial biogenesis includes the synthesis of mitochondrial membranes, the synthesis of mitochondrial proteins, the import of nucleus-encoded proteins, and mtDNA replication [19, 20]. In the brain, mitochondrial biogenesis takes place in the soma of the neurons, and it is regulated and controlled by multiple nuclear-encoded proteins such as peroxisome proliferator-activated receptor gamma (PPARγ) coactivator-1 alpha (PGC1α), which is believed to be the key regulator of the process. The interaction between PGC1α and nuclear respiratory factor 1 and 2 (NRF1 and NRF2) activates and overexpresses NRF1 and NRF2 leading to the activation of mitochondrial transcription factor A (Tfam), which is a critical protein involved in the transcription of mitochondrial ETC component genes by adhering to their promoter regions [21].

3. MITOCHONDRIA IN NEURAL CELLS: BIOENERGETICS AND DYNAMICS

Mitochondria are the home of major enzymes responsible for ATP production through the oxidation of sugars, fats, and proteins. Cells require a substantial quantity of ATP to perform their biological function. The balance between ATP supply/demand is linked to mitochondrial dynamics through fission and fusion [22].

Because of the nature of the metabolic activity of the brain, neurons consume around 20% of body glucose, which needs nonstop delivery of oxygen and nutrients through blood circulation [23, 24]. Once glucose is uptaken by the cell, it is metabolized into pyruvate molecules that are metabolized with oxygen via mitochondria tricarboxylic acid (TCA) cycle to feed the bioenergetic engine. Mitochondrial bioenergetic machinery contains transmembrane respiratory chain complexes protein complexes (I-IV) forming the ETC. Moreover, the proper assembly of these complexes with F1F0ATP synthase (known also as complex V) is necessary for ATP production throughout OXPHOS (Fig. 1) [25, 26].

Fig. (1).

Fig. (1)

Structure and OXPHOS of the mitochondria. (A) Mitochondria is a cellular organelle that play role in multiple cellular processes. Structurally, mitochondria have two membranes the outer and inner membrane. Both membranes are separated by an intermembrane space. The outer membrane separates the mitochondria from the cell cytoplasm while the inner membrane separates the matrix from intermembrane space. Inner membrane is differentiated and extend into the matrix forming cristae. (B) Membrane bound electron transport chain in addition to complex V are responsible for ATP production through OXPHOS. The process involves electron flow from complex I, complex II, complex III, complex VI, and complex V. Created with BioRender.com.

Mitochondria are extremely dynamic cellular components, which undergo constant remodeling by altering their size and number via two opposing controlled processes: fusion and fission, determined by dynamin-related GTPases (Fig. 2) [27]. Mitochondrial fusion is regulated by optic atrophy protein 1 (OPA1) and mitofusins 1 and 2 (MFN1 and MFN2). There are two forms of OPA1, the long isoform that anchors to the inner membrane and the short and soluble isoform that maintains the shape of the cristae [28]. Mitochondrial outer membrane GTPase, MFN1, and MFN2, facilitate the tethering of the two neighboring mitochondria by forming homo and heteroligomeric complexes [29, 30]. While MFNs are important for outer mitochondrial membrane fusion, the GTPase OPA1 is critical for inner mitochondrial membrane fusion. Evidence also proposed that OPA1 has a function in maintaining the shape of the cristae and the loss or mutation in OPA1 may result in an alteration in mitochondrial network morphology [31]. Interestingly, OPA1 requires only one (MFN1) of the two mitofusins to function normally [32].

Fig. (2).

Fig. (2)

Mitochondrial dynamics. Mitochondria are very dynamic organelle that go through fission and fusion. Fission or mitochondria division can occur once Drp1 forms a ring around the mitochondria once recruited by Fis1, MID49, and MID51. Fusion or fuse of two mitochondria together occurs once multiple OMM proteins MFN1/2 and OPA1 are recruited to the surface of the mitochondria. Created with BioRender.com.

Mitochondrial fission depends mainly on a large family of dynamin-like GTPase including cytosolic dynamin-related protein (Drp1). Drp1 may self-assemble into multimeric spiral-like structures on the outer mitochondrial membrane, which are essential for mitochondrial fission to occur [33]. Overexpression of Drp1 was found to cause fragmentation of the mitochondria, while dominant-negative mutation was linked to aberrant brain development because of a defect in fission in the mitochondria and the peroxisomes [33-35]. In addition to Drp1, fission is promoted by several other proteins that work on the recruitment of Drp1, such as the mitochondrial fission protein 1 (Fis1), mitochondrial fission factor (Mff), and mitochondrial dynamics proteins of 49 and 51 kDa (MiD49 and MiD51, respectively) [36, 37].

Because healthy mitochondria are critical for cellular survival, even little changes in mitochondrial homeostasis can have a major influence on the cell's function and integrity. Proteinopathies is the formation of misfolded and unfolded proteins that has a great impact on cell survival, which was found in many neurodegenerative diseases [38]. The existence of a quality control machinery is critical to overcome any change in mitochondrial homeostasis, which works on different levels: molecular, organellar, and cellular. If mitochondrial function declines, molecular chaperons are activated by mitochondrial unfolded protein response (UPR) that promotes repair and recovery of the mitochondrial network and maintains normal cellular function [39, 40]. In response to UPR, damaged proteins are refolded or removed from the mitochondria [39]. Despite the capacity of chaperons to restore protein folding equilibrium, cells poorly adapt to prolong UPR since the cell becomes in a persistent mitochondria recovery leading to an increase in the accumulation of damaged mtDNA, which significantly contributes to aging-associated neurodegenerative disorders [41-45]. In these conditions, the change in intracellular nutrients, functional mitochondria, and ROS level that compromises the integrity of the proteome is influenced by vitagenes that encode for heat shock proteins (Hsp), thioredoxin, thioredoxin reductase, heme oxygenase 1, and sirtunin, all of which can be upregulated by Nuclear erythroid 2-related factor 2 (Nrf2) [45-47].

4. MITOCHONDRIAL PATHOLOGY IN NDDs

4.1. Mitochondrial Respiratory Complex Defects

Defects in OXPHOS complexes may cause disturbance in the electron passage and proton pumping through the complexes, resulting in decreased mitochondrial function and hence reduced ATP synthesis [48-50]. First mitochondrial dysfunction was discovered in Sweden in 1962 when Rolf Luft of the University of Stockholm studied a case of a woman with severe fatigue and muscle weakness with significantly elevated body temperature. Generally, when the ADP amount is low, mitochondrial substrates are not oxidized. However, in the case of this woman, mitochondria were overactive, generating heat rather than cellular energy despite the low ADP level [51]. Since Rolf Luft`s report, various diseases have been linked to mitochondrial defects, which are mainly affecting muscle [52] and brain [53, 54] tissues, both of which require a huge amount of ATP.

Altered expression of encoded OXPHOS complexes subunits genes of mtDNA and nuclear DNA contribute to the oxidation metabolism defects in several diseases, including Alzheimer’s disease (AD) and schizophrenia [55]. Downregulation of mtDNA genes of complex I subunits, such as ND4 and ND15, was detected in the temporal cortex [56] and a decrease in its enzyme activity in AD patients [57, 58]. Differential expression was also detected in complex III and IV in the hippocampus and inferior parietal lobule of AD patients [55, 59].

According to several investigations, there is a link between the inhibition of the activity of complex I and neuronal cell apoptosis [60] after generating mouse models with specific complex I activity inhibition [61-63]. These models have been generated by using pharmacological inhibitors of complex I; MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine), rotenone, or Annonaceous acetogenins [64-66]. Using MPTP caused deterioration of dopaminergic neurons of the substantia nigra, which is a manifestation of Parkinson’s disease (PD) [67]. These models showed a significant decrease for different mtDNA gens, including complex 1 and ATP synthase subunits [68].

In addition to the defect in ATP production, ROS production can be increased via scaping protons from defective complex I and III as they are the primary sources of ROS in both physiological and pathological conditions [69]. Changing the expression of subunit genes of OXPHOS can alter the efficiency of the ETC in eliminating the excess of ROS, and subsequently resulting in the accumulation of ROS [70] owing to brain functional changes during aging and defects in the mitochondrial respiration chain, in which complexes I, III, and IV appear to be the most affected [71]. In skin fibroblasts, an anticorrelation was reported between the severity of complex I assembly and enzyme defects and increased ROS production [72, 73]. In addition, it was found that complex III dysfunction was also associated with ROS production in isolated lymphocytes’ mitochondria from complex III deficiency patients [74]. Another study was conducted on six complex III-deficient patients with BCS1L mutations [75]. It was found that superoxide production was increased as OXPHOS complexes I, III, and IV defects severity raised in parallel with decreased production of antioxidants [38].

4.2. Decreased Mitochondrial Free Radical Clearing Ability

A minimal amount of ROS is vital for different physiological activities [76]. It is involved in the maintenance of essential neural progenitors [40], redox signaling [41], and the immune system by directly killing pathogens [76]. However, an imbalance between overproduction and insufficient clearance of ROS [77, 78] has been shown to cause NDDs such as AD [79] and PD [80]. Overproduction of ROS can directly damage DNA, proteins, and lipids and subsequently impairs mitochondrial functions [78]. Therefore, the antioxidative defense mechanism by which mitochondria clear ROS and protect its component is very efficient and regulated.

The antioxidative defense mechanism exerts its function in three different pathways, all of which are executed in the mitochondria. The first pathway involves superoxide dismutase (SOD)-2 and catalase, which are capable of neutralizing ROS activity. The second pathway involves peroxiredoxins 3 and 5 (Prx3 and Prx5) enzymes. They rely for their regeneration on thioredoxin (Trx) and thioredoxin reductase (TRx2). The third pathway involves GPx1, GPx4 and glutaredoxins, which rely on GSH and glutathione reductase (GR) to renew GSH [81]. There are other antioxidants, which help in ROS clearance, such as NADPH, which depends on four enzymes located in the mitochondrial matrix for its regeneration [82] on intermembrane cytochrome c that removes superoxide to make it available for oxidative phosphorylation of ADP [83].

Defects of mitochondrial complexes and mitochondrial ROS clearance ability have been documented in NDDs. Mutations of complex I subunits are correlated with 40% of all mitochondrial defects [84]. In a normal state, dopamine is oxidized by monoamine oxidase to produce hydrogen peroxidase [85]. However, in a state of ROS overproduction, dopamine is non-enzymatically oxidized by superoxide, resulting in the generation of toxic oxidants [86]. Mutations in complex II subunits and their correlation with ROS production have also been documented. Qp of complex II is a vital site for electron transfer between ubiquinone and ubisemiquinone radical. Mutations in the district Qp site were shown to be involved in ubisemiquinone destabilization, resulting in scaping an electron to interact with false acceptors such as molecular oxygen, producing highly reactive ROS [87]. Dysfunction of brain mitochondrial complex II is a characteristic of Huntington's disease (HD) and different NDDs. It was shown that inhibiting the activity of complex II by using Nitropropionic acid can mimic HD-like pathology and symptoms [88] and increase the production of ROS in neurons [89].

The ROS clearance ability of mitochondria has been compromised in many NDDs. In AD, the defects are thought to be caused by a combination of high iron levels, low GSH levels, and mitochondrial complex I defect [90]. MPP+ treatment of dopaminergic PC12 cells reduced the expression of both antioxidant enzymes Trx1, Trx2, and Trx5 [91]. In addition, the aggregation of amyloid-β-peptide (Aβ) is correlated with mitochondrial antioxidant system defects in AD [92]. Aβ is responsible for H2O2 accumulation and contribution to mitochondrial defects [93]. It was found that SOD2 downregulation was correlated with the accumulation of brain Aβ levels in human amyloid precursor protein (hAPP) transgenic mice [94]. Other NDDs have also shown a decrease in Prx3 expressions, such as post-mortem brains of Down syndrome (DS) patients [95, 96] and in the motor neurons of familial amyotrophic lateral sclerosis (ALS) patients [97]. Selective mGSH depletion accelerates the onset of Huntington's disease (HD) symptoms in mice after in vivo injection of 3-nitropropionic acid [98].

The above discussed findings show a possible important role of mitochondrial antioxidant ability in the prevention of NDDs. Hence, boosting the antioxidant ability could be a potential preventive or therapeutic approach in the future.

4.3. mtDNA Lesions

MtDNA, circular DNA, can make its own RNAs and proteins since it has its own genetic material and machinery. MtDNA encodes 13 mitochondria-associated polypeptides, two rRNAs, and 22 tRNAs that are involved in mitochondrial protein synthesis [99, 100]. Even though mtDNA is highly protected by the antioxidants from the low level of OS, it is still sensitive to OS-induced mutations because of the low efficiency of mtDNA repair enzymes, and the physical closeness of mtDNA to free radical formation hotspots [101]. It has been shown that mutations of mtDNA resulted in insufficiency of mitochondrial complex activities, resulting in mitochondrial OXPHOS defects, ROS overproduction, and subsequently, cell apoptosis [102, 103] as observed in aging and a wide variety of NDDs, such as AD, PD, HD and ALS [104-107].

Genetic studies revealed mtDNA mutations in PD and AD to be associated with mitochondrial-specific OXPHOS complexes defects [108]. For example, specific knockout in mtDNA resulted in complex I deficiency of PD- affected neurons [109, 110]. In AD brains, it was found that cytochrome c oxidase-deficient neurons obtain a greater level of mtDNA mutations compared with age-matched controls [111]. In ALS, mutant SOD1 was found to significantly reduce voltage-dependent anion-selective channel protein 1 (VDAC1) activity, resulting in a significant decrease in energy production in mitochondria [112, 113]. Likewise, in transgenic ALS mice’ brain, levels of mtDNA damage were found to be 30-fold higher in the motor cortex as compared to spinal motor neurons [114, 115].

4.4. Mitochondrial Calcium Dyshomeostasis and Mitochondrial Permeability Transition Pore (mPTP)

Calcium signalling is involved in a variety of physiological processes, including muscle contraction, neuron excitability, and cell migration [116, 117]. The main storage sites of calcium are ER and mitochondria. Not surprisingly, therefore, mitochondria are essentially involved in calcium homeostasis maintenance through calcium buffering, which keeps calcium levels between 50 and 500 nM in numerous types of normal cells, and its interactions with other channels or organelles, such as ER [118-120]. Dysregulation of calcium may lead to defects in mitochondrial dynamics, function, and metabolism [121, 122].

Disturbance of calcium buffering capability of mitochondria results in an overload of calcium, which is one of the main features of mitochondrial abnormalities in NDDs. In AD, PD, HD, and ALS patients, calcium overload has been detected in affected regions, and similar results were obtained in the animal and/or cellular models of these diseases [123-126]. Calcium overload was found to cause ROS overproduction and activation of mPTP formation to enhance calcium efflux and ROS accumulation. Consequently, mitochondrial respiration complexes are damaged, cytoplasm is flooded with pro-apoptotic chemicals, mitochondria are swelled up and its membrane gets ruptured [127-129]. The main components of mPTP are voltage-dependent anion channel (VDAC), adenine nucleotide translocase (ANT), and cyclophilin D (CypD), which are embedded in the outer mitochondrial membrane (OMM), inner mitochondrial membrane (IMM), and matrix, respectively [130]. Translocation of CypD to IMM for binding with ANT is the initial step of the formation of mPTP [131-133]. The formation of mPTP in neurons causes apoptosis and cell death [138], which is widely detected in affected regions of different NDDs. CypD levels were found to be considerably higher in AD-affected areas, temporal pole, and hippocampi. Likewise, CypD was overexpressed in the brains of transgenic AD mice (including hippocampus and cortex) as well aged mice [127, 134]. Another study screened the expression CypD in different brain regions of rats with different NDDs. It was reported to exhibit an increase in the CypD expression levels in the disease-affected brain regions compared to normal healthy rats. This finding suggested the possible involvement of CypD in the etiology of different NDDs [135].

4.5. Mitochondrial Impairments in Brain Aging: Insight into the Role of Estrogen

Neurodegeneration is a progressive disorder involving brain aging. This is primarily characterized by a cognitive decline as well as a decline in physical functioning. The reason for this decline is not well understood. However, several hypotheses, causes, and factors have been proposed to explain the deterioration in cognitive and physical function [136]. One potential cause is the poor production of mitochondrial proteins due to the impairment of cellular energy production [137]. A link between the deterioration in mitochondrial function and aging has been suggested [138]. Moreover, mitochondrial impairment is believed to be one of the main reasons for neuronal cell death [139, 140]. Multiple shreds of evidence also revealed that NDDs may be caused by mitochondrial dysfunction [141]. In the past few years, studies with a focus on age-related NDDs such as AD, PD, and HD have established the link between mitochondrial dysfunctions and NDDs [142-145]. Several in vivo investigations have found that mitochondrial function in the brains of AD patients is impaired [146, 147]. Aging was found to cause a decline in mitophagy, which functions to remove defective proteins including dysfunctional mitochondria. The reduction of mitophagy may lead to deterioration in mitochondrial function because of the accumulation of defective proteins, and mutations along with oxidative injury. As a result, mtDNA volume, integrity, and functionality are reduced, which impairs mitochondria and may manifest decreased oxidative capacity and ATP production along with a substantial rise in ROS production [148]. Moreover, recent studies have described that mitochondria serve as targets for estrogen effect as well as essential intermediaries of steroid hormones’ biogenesis including estrogen [149]. Estrogen regulates mitochondrial structure and function by increasing the expression of respiratory complexes, antioxidant particles, and anti-apoptotic factors [150]. Predominantly, estrogen is produced in the ovaries and adrenal glands, but it is also produced by several tissues including adipose, breast stromal, and brain tissues. Estrogens exert their effects by acting on estrogen receptors α and β (ERα and ERβ), along with G-protein coupled estrogen receptor 1 (GPER1 also known as GPR30) [151]. ERα and ERβ are considered to be transcription regulators which influence gene transcription by binding to genomic and mtDNA. Additionally, ERα and ERβ initiate intracellular signaling pathways leading to the modification of transcriptional reactions, involving mitochondrial structure and function by interacting with plasma membrane-associated signaling proteins. Estrogens have different forms such as estriol and estradiol, which is the main female sex hormone that is engaged in mitochondrial function control [150]. Estradiol acts via the activation of the transcription factor NRF-1 that reacts with PGC-1α to regulate mitochondrial genes. Estrogen shows neuroprotective, neurotrophic, and antioxidant effects in the brain [152]. Several studies have indicated that during aging in women, estrogen production is decreased, thus precipitating the susceptibility of women to brain degeneration and aging diseases. The decrease in estrogen production was also correlated with significant impairment in brain mitochondrial function [153]. Aging is associated with not only sex steroid deficiency but also an augmentation in free radical generation causing damage to mitochondria. Impaired mitochondria have additional susceptibility to generate more free radicals that can damage cellular function including estrogen biogenesis [138]. Hence, mitochondrial therapeutics and the improvement of mitochondrial function using estrogen may be potential future targets and/or tools to fight several critical NDDs.

5. MITOCHONDRIAL MEDICINE FOR NEUROLOGICAL DISORDERS

5.1. Mitochondria-based Interventional Medicine

Mitochondrial dysfunction has a role in the onset of a plethora of diseases. Therefore, mitochondria are currently considered a prominent pharmacological target. A vast number of experiments have confirmed the beneficial outcome of mitochondria-targeting compounds (MTCs) in the management of various neurological disorders such as AD, PD, HD, and MS [154-157]. Although no criteria exist to characterize MTCs, Zinovkin et al. proposed that for any chemical to be classified as MTC 90% of the total quantity injected into the cells should get accumulated in the mitochondria [158]. In 1995, Thiobutyl-TPP bromide was identified as the first mitochondrial targeted compound by Murphy et al. [159]. Since then, dozens of MTCs have been involved in in vitro and in vivo experiments.

In contrast to other organelles inside the cells, the mitochondria have some distinctive features that can be utilized to synthesize/identify a MTC compound. For example, the mitochondria have negative charges in their cores and high transmembrane potential between the matrix and the intermembrane space (average value of 180 mV) [160]. The mitochondrial membrane potential is physiologically crucial to generate ATP molecules. However, it can be utilized to target the mitochondria via the utilization of cations, which will be directed to the negatively charged part of the mitochondria (the matrix) [161]. Another mitochondrial unique feature is the presence of phospholipid cardiolipin in their inner membrane. This phospholipid reinforces respiratory chain complexes but it can also be used to deliver compounds into the mitochondria [162]. Additionally, the mitochondria possess a specialized protein import system that binds to specific amino acid sequences [163]. This system can be exploited to deliver compounds inside the mitochondria.

Once inside the mitochondria, possible effects of MTCs include the decoupling of the mitochondrial membrane potential and its usage for mitochondria-dependent ATP generation (also called mitochondrial uncoupling), the induction of mitochondria-dependent programmed cell death, and the reduction of ROS level. Moreover, MTCs can be designed as a sensor to detect ROS level. MTCs may exert one or a combination of these effects. MTCs often have other concomitant effects because of the complex mitochondrial processes. Specific MTCs can lower ROS levels while acting as a mitochondrial uncoupler at certain doses [164]. Other MTCs can even induce ROS production and decreased ATP levels when used at high concentrations [165, 166]. Therefore, extensive in vivo/in vitro research is required to investigate the effect of any newly identified MTC.

5.2. Redox Therapy

A redox reaction is a chemical process that involves the movement of electrons between two molecules. In an oxidation-reduction redox reaction, the oxidation number of a molecule varies by acquiring or releasing an electron. While molecular oxygen (O2) is essential for life, its univalent reduction within the body causes the generation of various ROS species. Cells can clear ROS and protect themselves from their deleterious effects via the use of antioxidant compounds [167]. These antioxidants are either from endogenous or exogenous sources. Antioxidants include enzymes such as SOD, catalase, and GPx/GR, minerals such as copper and zinc, vitamins such as vitamin C and E, and other chemicals such as bilirubin and uric acid [168]. Lipid-soluble antioxidants like carotenoids, quinones, and certain polyphenols, as well as water-soluble antioxidants like ascorbic acid, are all examples of dietary antioxidants [168].

The balance between the rate of ROS generation and clearance is critical for OS. If the generation of ROS increases above the capacity of endogenous antioxidants or if endogenous free radical clearing ability was diminished, nutritional and exogenous sources of antioxidants might play a significant role to maintain redox balance and decrease stress levels. In fact, the beneficial effect of antioxidants has been reported in various in vitro/in vivo research (Table 1). Therefore, redox therapy is a hopeful approach to treating NDDs.

Table 1.

Examples of anti-oxidant substances and their beneficial effects in vivo / in vitro in neurodegenerative diseases.

Substance Extracted
Compound/s
Clinical Beneficial Effect Pass BBB Disease Molecular Beneficial Effect References
Ginko biloba EGb761 Cognition
Memory
Attention
- AD Reduce amyloid beta aggregation and
toxicity.
[228, 229]
Selenium Selenoproteins
glutathione
peroxidase (GPx)
Cognitive + AD
PD
MS
Protects against amyloid beta and iron/
hydrogen peroxide-mediated neuron death.
[230, 231]
Tumeric Curcumin Cognition + AD
PD
Decrease amyloid beta levels, neutralize ROS and peroxynitrite, increase GSH formation, inhibit transcription factor NF-kB*. [232]
Cannabinoids Dronabinol
Cannabidiol
Cognition
Memory
Sympathomimetic effect,
motor manifestation of PD
Neuropathic pain
- PD
AD
ALS
MS
Antioxidant effect, antinflammatory effect, inhibit transcription factor NF-kB, protect dopaminergic ncurons, ncrease trophic
factors, romotes, neuroglia survival,
decrease demyelination.
[233-236]

There has been a tremendous effort to identify nutraceutical antioxidants as novel therapies for neurodegenerative diseases. One of the natural product families that showed promising results in various studies is plant polyphenols. This family includes a number of flavonoids and non-flavonoids compounds, which differs according to the number of hydroxyl groups, and the presence of other substituents [169]. The list of the studied natural phenols includes curcumin, epigallocatechin-3-gallate (EGCG) (the flavanol found in green tea) and resveratrol (a stilbene found in grapes and in red wine) [170].

The beneficial effect of plant polyphenols has been an emerging research focus in many neurological diseases and has been more evident in conditions where oxidative stress is implicated in the pathophysiology such as AD and PD [171]. The antioxidant properties of polyphenols, are demonstrated by their ability to scavenge reactive radicals. Some polyphenols neutralize ROS by trapping chain-propagating free radicals, either via hydrogen atoms transfer or electron transfer [172]. Other polyphenols prevent the deleterious effect of ROS on a substrate via the inhibition of oxidation promoters (such as metal ions, dioxygen and pro-oxidative enzymes), thus, reducing their redox potentials [172]. Nonetheless, the exact mechanism/s of action of polyphenols is/are not fully understood. Studies suggested that polyphenols might have multiple mechanisms of action and that they are capable of modifying gene expression, miRNA and proteins [173]. In line with that, emerging evidence shows that the polyphenols – mediated neuroprotection is likely achieved via the activation of vitagene signaling pathways [171].

The term “vitagenes” is referred to a group of genes involved in preserving cellular homeostasis during either physiological or pathological stressful conditions [174]. Research found that the expression of numerous vitagenes decreases with age. Specific supplement intake positively affects vitagenes expression and ameliorates the unwanted effect of aging [174]. Notably, the number of genes that may be classified as vitagenes rises if aging was associated with disease [174]. Some important vitagenes are listed in Table 2. Notably, they are all directly or indirectly linked to the mitochondria.

Table 2.

Some important vitagenes and their function in the cell.

Vitagene Protein Function
BCL2 Bcl-2 Protect against of mitochondrial-dependent apoptosis [237]
CREB1 CREB-1 Regulate mitochondrial synthesis [238]
GSS Glutathione synthetase Defense against reactive oxygen species [239]
HMOX1 Heme oxigenase-1 Regulate mitochondrial synthesis [239]
HSP70 Hsp70 Post translational modification of mitochondrial proteins [240]
SIRT1 - 4 Sirtuin-1 - 4 Post translational modification of mitochondrial proteins, regulation of mitochondrial electron transport and oxidation, Regulate mitochondrial synthesis and dynamics [241, 242]
SOD2 Superoxide dismutase Defense against reactive oxygen species [243]

Research showed that polyphenols initiate phase 2 response, leading to the expression of various Nrf2-dependent antioxidant vitagenes, including the aforementioned Hsp70, HO-1, sirtuins system and many others [175]. Nrf2 is a key transcription factor that governs hundreds of cytoprotective genes. Notably, Nrf2 activation induces a mild stress response, which promotes cell survival and induces a healthy physiological steady state. However, prolonged Nrf2 activation may results in an adverse outcome, indicating that Nrf2 has a hormetic-like behavior [176, 177]. Hormesis has emerged as a central concept in biological and biomedical sciences. Hermetic dose-response may be established when a low certain dose leads to stimulation while a high dose of the same compounds leads to inhibition, that is, a biphasic dose-response relationship [178]. The principle of hormesis appears to be applicable to the downstream target of the Nrf2 pathway as well. Research showed that the antioxidant effect of HO-1 against oxidative and nitrosative stress is abolished upon excessive upregulation of HO-1 system [179-181]. A possible explanation is the accumulation of its by-products such as carbon monoxide, iron, and bilirubin [181].

An increasing number of studies support the usefulness of polyphenols, more evidently when used with other drugs [182]. Although some conflicting results have been reported, these could be attributed to various factors such as different experimental settings and clinical conditions. More work is needed to investigate these findings. However, the overall current body of the data strongly suggests the nutraceutical value of plant polyphenols.

5.3. Mitochondrial Permeability Transition Inhibition

Since the discovery of cyclosporin A (CsA) as the first inhibitor of mPTP four decades ago [183], cumulative evidence showed that mitochondrial dysfunction and mPTP opening is considered the primary mechanism of apoptosis in many NDDs [184]. Consequently, several CsA- related mPTP inhibitors have been identified and tested. Although these inhibitors showed promising results in preclinical research, we are still far from a complete understanding of their molecular mechanism. This is mainly attributed to the polypharmacology of CsA and its derivatives [184]. In addition, the process of mPTP opening itself is not completely understood. Various proteins such as IMM proteins: ANT and mitochondrial phosphate carrier (PiC), and OMM proteins: VDAC and BCL-2 family members BAK/BAX; have been suggested as key players in mPTP creation. Nonetheless, almost all of these candidate proteins did not show consistent results in knockout/over-expression genetic studies [184].

To date, the only protein that has consistently proved to serve a direct function in regulating mPTP opening is peptidyl prolyl cis-trans isomerase (PPIase), also known as CypD [185]. CypD possesses a Peptidyl-Proline Isomerases activity, which is crucial for mediating pore opening. Research showed that CypD inhibition decreases the susceptibility of the cells to Ca2+ and ROS via the inhibition of mPTP formation [186]. However, if Ca2+ concentration increases above a certain level, mPTP formation occurs regardless of CypD inhibition [186]. This suggests that CypD plays a regular role and is not structurally involved in the process. The scientists proposed that the mPTP formation process starts upon the binding of ANT to CypD in IMM. This binding along with the help of other proteins consequently mediate the formation of tunnel-like structures across both IMM and OMM [187].

The vast majority of known mPTP inhibitors are CypD-dependent as they target CypD specifically [188]. CsA is the most tested and best characterized CypD-dependant mPTP inhibitor. In mAPP mice models of AD (mice expressing a mutant form of hAPP), CsA treatment protected the neurons from oxidative damage via reducing the generation rate of ROS [189]. Moreover, CsA treatment attenuated Ca2+ imbalances and mitochondrial swelling [189]. CsA-treated mice had substantially improved scores in learning and memory tests [189]. Similar findings were observed in CypD-deficient mAPP mouse model [189]. The beneficial effect of CypD inhibition was also demonstrated in PD-related models. CypD-deficient mice treated with mPTP showed significant cytoprotection compared with control mice [190]. The isolated mitochondria of CypD-deficient mice demonstrated higher resistance to MPP+ treatment and lower ROS level in comparison to the wild type [190]. Interestingly, the expression of apoptotic markers in CypD-deficient mice was not increased, and the beneficial effect of CypD-deficiency was apparent only with an acute regimen of mPTP treatment [190].

Unfortunately, the beneficial effect of CsA mediated mPTP inhibition was difficult to be demonstrated clinically due to certain limitations like toxicity, inhibitory effect on the immune system, and limited bioavailability in the central nervous system (CNS) [191]. Therefore, researchers are currently working to synthesize and identify alternatives to CsA. One of the approaches used to identify new mPTP inhibitors includes virtual screening to identify the best fit synthetic, and semisynthetic molecules. In this approach, the qualified molecules can be further modified into novel more active analogs with the aid of modeling techniques such as in-silico QSAR modeling and molecular docking. For example, Valasani et al. used diastereomeric crystallization and pharmacophore modeling to generate multiple selective CypD inhibitors [192]. Likewise, Belkacem et al. synthesized a group of nonpeptidic cyclophilin inhibitors that are structurally different from CsA, and possess PPIase inhibitory activity [193]. In addition, it has been reported that some quinoxaline derivatives such as quinazoline urea analogs bind and inhibit CypD, and demonstrated strong inhibitory ability against Ca2+-dependent rat liver mitochondrial swelling [194]. These compounds are promising CsA alternatives if they prove to be safe clinically.

5.4. Mitochondrial Gene Therapy

Gene therapy for NDDs has progressed significantly over the past years. This is attributed not only to the discoveries related to the role of genetics in the etiology of these disorders but also to the advance in the technology that delivers the therapeutic DNA/RNA segments to the desired tissue. Gene therapy may result in gene silencing to control the gain of function mutations or gene overexpression to compensate for the loss of function mutations. Gene therapy has many advantages in comparison to traditional medical treatment. It has a more permanent effect and does not require repetitive doses. Gene therapy can also treat tissue that has been consistently unresponsive to medical treatment.

There are special viral/non-viral vectors that have been used to carry the transgene to their targets in the CNS. These transgenes might carry codes to express therapeutic proteins, cDNA for gene addition, Cas9/gRNA for gene editing, small interfering RNA (siRNA) etc. [195]. The most commonly used vector in NDDs is the Adeno-associated viruses (AAV) [196]. Dozens of AAV, which are classified in 13 serotypes, have been identified [197]. AAV2 in particular is commonly used clinically for gene therapy of NDDs as it is relatively safe and has consistent expression in neurons [197]. For example, AAV2-NGF (nerve growth factor) gene therapy via basal forebrain injection proved to be well tolerated in AD patients [198]. AAV2-NGF carries the codes for the NGF), which is an endogenous neurotrophic-factor protein that can protect degenerating cholinergic neurons. AAV2-NGF was used in the multicenter randomized clinical trial as a treatment for AD [198]. However, there was no improvement in cognition after 24 months of treatment [198].

Since mitochondrial dysfunction plays a significant role in the pathology of NDDs, mitochondrial gene therapy might have great potential as a treatment strategy. Gene therapy that mediates the productions of various regulatory factors of mitochondrial ROS and mitochondrial dynamics was able to protect neurons in vitro in PD and AD experimental models [199, 200]. However, clinical studies did not show promising results [197]. This could be because the patients in these studies were too mature for treatment because they had already experienced substantial neurodegeneration. Nonetheless, scientists still believe that mitochondria-targeting gene therapy for NDDs is a promising area of research, particularly for NDDs with strong evidence of mitochondrial dysfunctions such as PD.

5.5. TPP Cation-based Antioxidants

As mitochondria are the main course of ROS in the cells, antioxidant drug that targets ROS production needs to be accumulated in the mitochondria to mediate its effect. This could be achieved via the conjugation of the antioxidant molecule to a mitochondria-targeted peptide [201] which is an N-terminal mitochondrial targeting signal (MTS) peptide that builds amphipathic helical structures with positively charged residues. The positive charge allows electrostatic interactions with the negatively charged mitochondrial interface [201]. Subsequently, the molecule is transported inside the mitochondria. Alternatively, the antioxidant can be conjugated to a lipophilic cation that can diffuse through the phospholipid bilayer of the mitochondrial membrane [202]. The charged cations are functionally arranged on the hydrophobic surface of lipophilic cation molecules. This arrangement renders the lipophilic cation capable of diffusing through the mitochondrial membrane with minimal activation energy and without the need for pores or transporter proteins [202]. The most commonly used lipophilic cation to deliver antioxidants to the inside of the mitochondria is TPP [201]. TPP contains an intermediate positive charge of phosphorus which attracts the TPP toward the negatively charged mitochondrial matrix [201]. In the coming section, we will briefly discuss some examples of TPP cation-based antioxidants.

5.6. MitoQ

MitoQ is a mitochondria-directed compound made up of ubiquinone molecule (the oxidized form of CoQ10) that is covalently bonded to TPP molecule [203]. Thus, MitoQ can diffuse through the mitochondrial membrane and aggregate in the mitochondrial matrix. Inside the mitochondria, MitoQ detoxifies ROS and consequently reduces lipid peroxidation and mitochondrial injury [204]. Moreover, ubiquinol (the reduced form of CoQ10) can be oxidized back to ubiquinone and used continuously by complex II of the ETC, which makes MitoQ superior to other mitochondria-targeted antioxidants [201].

Since the discovery of MitoQ in the 1990s, it has been involved in multiple studies to investigate its potential in NDDs [205]. With regards to PD, MitoQ showed promising results in in vitro studies. MitoQ pre-treatment prevented mitochondrial translocation of Drp1 and the consequent mitochondrial fragmentation in 6-hydroxydopamine treated SH-SY5Y cells [206]. In the same study, MitoQ also prevented the trafficking of the pro-apoptotic factor, Bax, to the mitochondria and enhanced the survival of the SH-SY5Y cells [206]. In MPTP-induced mouse models of PD, MitoQ treatment improved motor deficit and increased dopamine levels and the expression of tyrosine hydroxylase in the substantia nigra [207]. Similar beneficial effects were also observed in AD-related research [207]. MitoQ reduced both Aβ accumulation and Aβ-induced OS. In the genetic murine model of AD, namely 3xTg-AD mouse, MitoQ delayed the onset of cognitive dysfunction in Morris Water Maze tests [208]. The obtained brain samples revealed evidence of the therapeutic effect at molecular levels such as decreased caspase 3 and 7 activity, Aβ immunoreactivity, and oxidative stress markers [208].

Although MitoQ passed phase I and phase II clinical trials with success as a treatment option for patients with Hepatitis C virus [209], it failed to demonstrate clinical benefits in PD patients. One possible reason is that the degree of dopaminergic neuronal impairment in PD patients had exceeded the protective capacity of MitoQ. More study is required to confirm the clinical benefit of MitoQ in NDDs given their robust encouraging findings in in vitro and in vivo investigations.

5.7. MitoVitE

Vitamin E is a collection of naturally occurring plant lipids known as tocopherols and tocotrienols [210]. The members of the vitamin E family differ in their methylation pattern. However, they all possess an electrophilic hydroxyl group that can extinguish carbon radicals effectively [210]. This hydroxyl group is responsible for the powerful antioxidant effect of vitamin E [210]. Physiologically, Vitamin E is highly linked to the nervous system. In fact, the manifestations of Vitamin E deficiency are mainly neurological [211]. Interestingly, it seems that the brain is specially adapted to vitamin E usage. Vitamin E’s biological life in the brain is gradual and varies with its concentration from region to region [212, 213], which also reflects the presence of tissue-specific regulatory mechanisms for vitamin E storage and usage.

Vitamin E appears to be useful in diseases in which OS serves a significant role in the pathogenesis [214]. Previous studies showed that a low concentration of vitamin E has a neuroprotective role in the case of glutamate excitotoxicity as well [215]. A recent questionnaire-based case-control study suggested that vitamin E intake might protect against PD. This study involved 100 PD patients and an equal number of healthy controls and showed that the quantity of vitamin E in one's diet was negatively correlated with PD incidence, regardless of sex and age [216]. This is in accordance with a recent meta-analysis research that looked at the link between PD and dietary consumption of vitamin C, β carotene, and vitamin E [217]. While there was no significant link between vitamin C and carotene, nutritional consumption of vitamin E was discovered to be protective against PD. More research is needed to investigate how MitoVitE can help elderly people and people with NDDs.

5.8. MitoPBN

MitoPBN is a compound that consists of coenzyme Q (quinone) and phenyl tertbutylnitrone molecules attached to TPP bromide [218]. MitoPBN was synthesized to protect against ROS-induced lipid peroxidation relying on the ability of N-tert-butyl-α-phenylnitrone (PBN) to neutralize carbon-centered radicals (R.) and peroxyl radicals (ROO.) [219].

A more complex nitrenium cation (LPBNAH) (scientific nomenclature: N-[4-(octa-O-acetylactobionamidomethylene) benzylidene]-N-[1,1-dimethyl-2-(N-octanoyl) amido]-ethyl-amine N-oxide), which is a derivative of PBN, seem to be more effective due to its high stability and selectivity [220]. This compound demonstrated a neuroprotective effect against OS and cell demise in neuroblastoma cells that were exposed to Aβ, hydrogen peroxide, and 3-hydroxykynurenine [220]. Nonetheless, there are very few studies have been done to explore the effect of MitoPBN and LPBNAH in the context of NDDs. More in vivo and in vitro research is required to investigate the potential of these promising compounds.

5.9. Dimebon

The antihistamine medicine Dimebon was initially utilized to manage allergies in Russia in the early 1980s. In recent times, Dimebon has been suggested as a treatment for NDDs [221]. The first crucial clinical trial of Dimebon in AD revealed that it reduced the disease's clinical manifestations [222]. In this randomized, double-blind, placebo-controlled trial of 183 individuals with mild-to-moderate AD, Dimebon showed statistically significant improvements in all essential areas of the disorder including memory, thinking, activities of daily living, behavior, and overall function. People treated with Dimebon did considerably better than placebo-treated individuals in all critical metrics of the disorder after 6 months and one year of treatment [222]. Dimebon was also found to be effective in a phase II trial done by Medivation and the Huntington Study Group with HD patients (DIMOND). Although there are highly hopeful outcomes in clinical trials, but mechanisms behind Dimebon's therapeutic effects are yet unknown. Dimebon has been shown to block NMDA receptors and voltage-gated Ca2+ channels in the past [223-226]. Dimebon also inhibits the entry of the mPTP caused by Aβ25–35 and MPP+, according to a prior study [227]. These findings imply that Dimebon's therapeutic effects may be attributed to its capacity to maintain neuronal calcium homeostasis and mitochondrial activity. Dimebon's mechanism of action in neurological disorders is currently being investigated by researchers.

As discussed in the aforementioned sections, the specific roles of each mitochondria-directed medicine have been illustrated in Fig. (3).

Fig. (3).

Fig. (3)

Mitochondrial medicines. Antioxidants maintain redox balance and decrease the stress level by avoiding damage of lipids, proteins, and DNA; Dimebon inhibits glutamate inflow by binding to NMDA receptors on the cell's surface. It also binds to Ca2+ channels, preventing a Ca2+ influx into the cell; Triphenylphosphonium cation-based antioxidants like MitoQ. MitoPBN etc. could be used to deliver antioxidants to the inside of the mitochondria; cyclosporin A (CsA) inhibits mitochondrial permeability transition pore (mPTP). Created with BioRender.com.

CONCLUSION AND FUTURE PERSPECTIVES

Numerous research reports indicate that mitochondrial abnormality and oxidative stress are important in the pathophysiology of many NDDs including AD, PD, HD and ALS. Mitochondria are the primary source of energy for brain cells to function normally. Higher production of ROS, aberrant protein-protein interactions, and decreased mitochondrial ATP synthesis have all been linked to mitochondrial abnormalities. In early-onset, inherited, and late-onset, non-inherited NDDs, increased generation of ROS with reduced mitochondrial activity has been demonstrated to harm neurons. As a result, developing strategies to combat or reduce mitochondrial abnormality could be therapeutically beneficial. Redox therapy, mitochondrial gene therapy, TPP cation-based antioxidants have been reported to be efficient in pre-clinical and clinical investigations demonstrating their potential. Furthermore, using antioxidants in combination maybe even be more efficient than using single compounds. Dimebon, a novel therapy candidate, can improve cognitive deterioration in AD and HD patients. However, broader clinical studies with a greater quantity of individuals are required to give more conclusive evidence of these compounds' therapeutic potential. Nevertheless, the molecular mechanism of Dimebon is unknown, indicating the necessity of additional investigation, especially in animal studies. Mitochondrial-targeted medicines will open long-term options for manipulating mitochondrial function, potentially protecting against NDDs.

ACKNOWLEDGEMENTS

The authors gratefully acknowledge technical and financial support from the Ministry of Education and King Abdulaziz University, DSR, Jeddah, Saudi Arabia.

LIST OF ABBREVIATIONS

AAV

Adeno-Associated Viruses

AD

Alzheimer’s Disease

ALS

Amyotrophic Lateral Sclerosis

ANT

Adenine Nucleotide Translocase

DS

Down Syndrome

ETC

Electron Transport Chain

GPx

Glutathione Peroxidase

GR

Glutathione Reductase

HD

Huntington's Disease

IMM

Inner Mitochondrial Membrane

Mff

Mitochondrial Fission Factor

MnSOD

Manganese Superoxide Dismutase

mPTP

Mitochondrial Permeability Transition Pore

MTS

Mitochondrial Targeting Signal

NDDs

Neurodegenerative Disorders

NOS

Nitric Oxide Synthase

OPA1

Optic Atrophy Protein 1

OXPHOS

Oxidative Phosphorylation

PD

Parkinson’s Disease

RNS

Reactive Nitrogen Species

ROS

Reactive Oxygen Species

siRNA

Small Interfering RNA

TCA

Tricarboxylic Acid

TPP

Triphenylphosphonium

UPR

Unfolded Protein Response

VDAC

Voltage-Dependent Anion Channel

CONSENT FOR PUBLICATION

Not applicable.

FUNDING

This research work was funded by the Institutional Fund Projects under grant no. (IFPDP-64-22).

CONFLICT OF INTEREST

The authors declare no conflict of interest, financial or otherwise.

REFERENCES

  • 1.Harris J.J., Jolivet R., Attwell D. Synaptic energy use and supply. Neuron. 2012;75(5):762–777. doi: 10.1016/j.neuron.2012.08.019. [DOI] [PubMed] [Google Scholar]
  • 2.Moreira P.I., Santos M.S., Oliveira C.R. Alzheimer’s disease: A lesson from mitochondrial dysfunction. Antioxid. Redox Signal. 2007;9(10):1621–1630. doi: 10.1089/ars.2007.1703. [DOI] [PubMed] [Google Scholar]
  • 3.Duchen M.R. Mitochondria and calcium: From cell signalling to cell death. J. Physiol. 2000;529(Pt 1):57–68. doi: 10.1111/j.1469-7793.2000.00057.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Susin S.A., Lorenzo H.K., Zamzami N., Marzo I., Snow B.E., Brothers G.M., Mangion J., Jacotot E., Costantini P., Loeffler M., Larochette N., Goodlett D.R., Aebersold R., Siderovski D.P., Penninger J.M., Kroemer G. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature. 1999;397(6718):441–446. doi: 10.1038/17135. [DOI] [PubMed] [Google Scholar]
  • 5.Budd S.L., Nicholls D.G. Mitochondria in the life and death of neurons. Essays Biochem. 1998;33:43–52. doi: 10.1042/bse0330043. [DOI] [PubMed] [Google Scholar]
  • 6.Finkel T. Radical medicine: Treating ageing to cure disease. Nat. Rev. Mol. Cell Biol. 2005;6(12):971–976. doi: 10.1038/nrm1763. [DOI] [PubMed] [Google Scholar]
  • 7.Fiskum G. Mitochondrial participation in ischemic and traumatic neural cell death. J. Neurotrauma. 2000;17(10):843–855. doi: 10.1089/neu.2000.17.843. [DOI] [PubMed] [Google Scholar]
  • 8.Moreira P.I., Duarte A.I., Santos M.S., Rego A.C., Oliveira C.R. An integrative view of the role of oxidative stress, mitochondria and insulin in Alzheimer’s disease. J. Alzheimers Dis. 2009;16(4):741–761. doi: 10.3233/JAD-2009-0972. [DOI] [PubMed] [Google Scholar]
  • 9.Sullivan P.G., Keller J.N., Mattson M.P., Scheff S.W. Traumatic brain injury alters synaptic homeostasis: Implications for impaired mitochondrial and transport function. J. Neurotrauma. 1998;15(10):789–798. doi: 10.1089/neu.1998.15.789. [DOI] [PubMed] [Google Scholar]
  • 10.Zorov D.B., Juhaszova M., Sollott S.J. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol. Rev. 2014;94(3):909–950. doi: 10.1152/physrev.00026.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Valko M., Leibfritz D., Moncol J., Cronin M.T.D., Mazur M., Telser J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007;39(1):44–84. doi: 10.1016/j.biocel.2006.07.001. [DOI] [PubMed] [Google Scholar]
  • 12.Moreira P.I., Nunomura A., Nakamura M., Takeda A., Shenk J.C., Aliev G., Smith M.A., Perry G. Nucleic acid oxidation in Alzheimer disease. Free Radic. Biol. Med. 2008;44(8):1493–1505. doi: 10.1016/j.freeradbiomed.2008.01.002. [DOI] [PubMed] [Google Scholar]
  • 13.Islam Md. T. Oxidative stress and mitochondrial dysfunction-linked neurodegenerative disorders. Neurol. Res. 2017;39(1):73–82. doi: 10.1080/01616412.2016.1251711. [DOI] [PubMed] [Google Scholar]
  • 14.Singh A., Kukreti R., Saso L., Kukreti S. Oxidative stress: A key modulator in neurodegenerative diseases. Molecules. 2019;24(8):1583. doi: 10.3390/molecules24081583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Reddy P.H. Amyloid precursor protein-mediated free radicals and oxidative damage: Implications for the development and progression of Alzheimer’s disease. J. Neurochem. 2006;96(1):1–13. doi: 10.1111/j.1471-4159.2005.03530.x. [DOI] [PubMed] [Google Scholar]
  • 16.Schapira A.H.V. Mitochondrial disease. Lancet. 2006;368(9529):70–82. doi: 10.1016/S0140-6736(06)68970-8. [DOI] [PubMed] [Google Scholar]
  • 17.Swerdlow R.H., Burns J.M., Khan S.M. The Alzheimer’s disease mitochondrial cascade hypothesis. J. Alzheimers Dis. 2010;20(Suppl. 2):S265–S279. doi: 10.3233/JAD-2010-100339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wallace D.C. A mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: A dawn for evolutionary medicine. Annu. Rev. Genet. 2005;39(1):359–407. doi: 10.1146/annurev.genet.39.110304.095751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ventura-Clapier R., Garnier A., Veksler V. Transcriptional control of mitochondrial biogenesis: The central role of PGC-1. Cardiovasc. Res. 2008;79(2):208–217. doi: 10.1093/cvr/cvn098. [DOI] [PubMed] [Google Scholar]
  • 20.Zhu J., Wang K.Z.Q., Chu C.T. After the banquet. Autophagy. 2013;9(11):1663–1676. doi: 10.4161/auto.24135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Uittenbogaard M., Chiaramello A. Mitochondrial biogenesis: A therapeutic target for neurodevelopmental disorders and neurodegenerative diseases. Curr. Pharm. Des. 2014;20(35):5574–5593. doi: 10.2174/1381612820666140305224906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Twig G., Elorza A., Molina A.J.A., Mohamed H., Wikstrom J.D., Walzer G., Stiles L., Haigh S.E., Katz S., Las G., Alroy J., Wu M., Py B.F., Yuan J., Deeney J.T., Corkey B.E., Shirihai O.S. Fission and selective fusion govern mitochondrial segregation and elimination by autophagy. EMBO J. 2008;27(2):433–446. doi: 10.1038/sj.emboj.7601963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Siesjö B.K. Brain metabolism and anaesthesia. Acta Anaesthesiol. Scand. Suppl. 1978;70:56–59. [PubMed] [Google Scholar]
  • 24.Rolfe D.F., Brown G.C. Cellular energy utilization and molecular origin of standard metabolic rate in mammals. Physiol. Rev. 1997;77(3):731–758. doi: 10.1152/physrev.1997.77.3.731. [DOI] [PubMed] [Google Scholar]
  • 25.Guo R., Zong S., Wu M., Gu J., Yang M. Architecture of human mitochondrial respiratory megacomplex I2III2IV2. Cell. 2017;170(6):1247–1257.e12. doi: 10.1016/j.cell.2017.07.050. [DOI] [PubMed] [Google Scholar]
  • 26.Iwata S., Lee J.W., Okada K., Lee J.K., Iwata M., Rasmussen B., Link T.A., Ramaswamy S., Jap B.K. Complete structure of the 11-subunit bovine mitochondrial cytochrome bc1 complex. Science. 1998;281(5373):64–71. doi: 10.1126/science.281.5373.64. [DOI] [PubMed] [Google Scholar]
  • 27.Westermann B. Mitochondrial fusion and fission in cell life and death. Nat. Rev. Mol. Cell Biol. 2010;11(12):872–884. doi: 10.1038/nrm3013. [DOI] [PubMed] [Google Scholar]
  • 28.Lee H., Smith S.B., Yoon Y. The short variant of the mitochondrial dynamin OPA1 maintains mitochondrial energetics and cristae structure. J. Biol. Chem. 2017;292(17):7115–7130. doi: 10.1074/jbc.M116.762567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ishihara N., Eura Y., Mihara K. Mitofusin 1 and 2 play distinct roles in mitochondrial fusion reactions via GTPase activity. J. Cell Sci. 2004;117(26):6535–6546. doi: 10.1242/jcs.01565. [DOI] [PubMed] [Google Scholar]
  • 30.Koshiba T., Detmer S.A., Kaiser J.T., Chen H., McCaffery J.M., Chan D.C. Structural basis of mitochondrial tethering by mitofusin complexes. Science. 2004;305(5685):858–862. doi: 10.1126/science.1099793. [DOI] [PubMed] [Google Scholar]
  • 31.Chan D.C. Mitochondrial dynamics and its involvement in disease. Annu. Rev. Pathol. 2020;15(1):235–259. doi: 10.1146/annurev-pathmechdis-012419-032711. [DOI] [PubMed] [Google Scholar]
  • 32.Cipolat S., de Brito O.M., Dal Zilio B., Scorrano L. OPA1 requires mitofusin 1 to promote mitochondrial fusion. Proc. Natl. Acad. Sci. USA. 2004;101(45):15927–15932. doi: 10.1073/pnas.0407043101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Smirnova E., Griparic L., Shurland D.L., van der Bliek A.M. Dynamin-related protein Drp1 is required for mitochondrial division in mammalian cells. Mol. Biol. Cell. 2001;12(8):2245–2256. doi: 10.1091/mbc.12.8.2245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Waterham H.R., Koster J., van Roermund C.W.T., Mooyer P.A.W., Wanders R.J.A., Leonard J.V. A lethal defect of mitochondrial and peroxisomal fission. N. Engl. J. Med. 2007;356(17):1736–1741. doi: 10.1056/NEJMoa064436. [DOI] [PubMed] [Google Scholar]
  • 35.Lee Y., Jeong S.Y., Karbowski M., Smith C.L., Youle R.J. Roles of the mammalian mitochondrial fission and fusion mediators Fis1, Drp1, and Opa1 in apoptosis. Mol. Biol. Cell. 2004;15(11):5001–5011. doi: 10.1091/mbc.e04-04-0294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Palmer C.S., Osellame L.D., Laine D., Koutsopoulos O.S., Frazier A.E., Ryan M.T. MiD49 and MiD51, new components of the mitochondrial fission machinery. EMBO Rep. 2011;12(6):565–573. doi: 10.1038/embor.2011.54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Jakobs S., Martini N., Schauss A.C., Egner A., Westermann B., Hell S.W. Spatial and temporal dynamics of budding yeast mitochondria lacking the division component Fis1p. J. Cell Sci. 2003;116(10):2005–2014. doi: 10.1242/jcs.00423. [DOI] [PubMed] [Google Scholar]
  • 38.Sweeney P., Park H., Baumann M., Dunlop J., Frydman J., Kopito R., McCampbell A., Leblanc G., Venkateswaran A., Nurmi A., Hodgson R. Protein misfolding in neurodegenerative diseases: Implications and strategies. Transl. Neurodegener. 2017;6(1):6. doi: 10.1186/s40035-017-0077-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Larsen S.B., Hanss Z., Krüger R. The genetic architecture of mitochondrial dysfunction in Parkinson’s disease. Cell Tissue Res. 2018;373(1):21–37. doi: 10.1007/s00441-017-2768-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kaufman D.M., Wu X., Scott B.A., Itani O.A., Van Gilst M.R., Bruce J.E., Michael Crowder C. Ageing and hypoxia cause protein aggregation in mitochondria. Cell Death Differ. 2017;24(10):1730–1738. doi: 10.1038/cdd.2017.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Gitschlag B.L., Kirby C.S., Samuels D.C., Gangula R.D., Mallal S.A., Patel M.R. Homeostatic responses regulate selfish mitochondrial genome dynamics in C. elegans. Cell Metab. 2016;24(1):91–103. doi: 10.1016/j.cmet.2016.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lin Y.F., Schulz A.M., Pellegrino M.W., Lu Y., Shaham S., Haynes C.M. Maintenance and propagation of a deleterious mitochondrial genome by the mitochondrial unfolded protein response. Nature. 2016;533(7603):416–419. doi: 10.1038/nature17989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Calabrese V., Mancuso C., Calvani M., Rizzarelli E., Butterfield D.A., Giuffrida Stella A.M. Nitric oxide in the central nervous system: Neuroprotection versus neurotoxicity. Nat. Rev. Neurosci. 2007;8(10):766–775. doi: 10.1038/nrn2214. [DOI] [PubMed] [Google Scholar]
  • 44.Calabrese V., Cornelius C., Mancuso C., Pennisi G., Calafato S., Bellia F., Bates T.E., Giuffrida Stella A.M., Schapira T., Dinkova Kostova A.T., Rizzarelli E. Cellular stress response: A novel target for chemoprevention and nutritional neuroprotection in aging, neurodegenerative disorders and longevity. Neurochem. Res. 2008;33(12):2444–2471. doi: 10.1007/s11064-008-9775-9. [DOI] [PubMed] [Google Scholar]
  • 45.Cornelius C., Perrotta R., Graziano A., Calabrese E., Calabrese V. Stress responses, vitagenes and hormesis as critical determinants in aging and longevity: Mitochondria as a “chi”. Immun. Ageing. 2013;10:15. doi: 10.1186/1742-4933-10-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Calabrese V., Cornelius C., Dinkova-Kostova A.T., Calabrese E.J., Mattson M.P. Cellular stress responses, the hormesis paradigm, and vitagenes: Novel targets for therapeutic intervention in neurodegenerative disorders. Antioxid. Redox Signal. 2010;13(11):1763–1811. doi: 10.1089/ars.2009.3074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Trovato Salinaro A., Pennisi M., Di Paola R., Scuto M., Crupi R., Cambria M.T., Ontario M.L., Tomasello M., Uva M., Maiolino L., Calabrese E.J., Cuzzocrea S., Calabrese V. Neuroinflammation and neurohormesis in the pathogenesis of Alzheimer’s disease and Alzheimer-linked pathologies: Modulation by nutritional mushrooms. Immun. Ageing. 2018;15(1):8. doi: 10.1186/s12979-017-0108-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Iuso A., Scacco S., Piccoli C., Bellomo F., Petruzzella V., Trentadue R., Minuto M., Ripoli M., Capitanio N., Zeviani M., Papa S. Dysfunctions of cellular oxidative metabolism in patients with mutations in the NDUFS1 and NDUFS4 genes of complex I. J. Biol. Chem. 2006;281(15):10374–10380. doi: 10.1074/jbc.M513387200. [DOI] [PubMed] [Google Scholar]
  • 49.Distelmaier F., Visch H.J., Smeitink J.A.M., Mayatepek E., Koopman W.J.H., Willems P.H.G.M. The antioxidant Trolox restores mitochondrial membrane potential and Ca2+-stimulated ATP production in human complex I deficiency. J. Mol. Med. (Berl.) 2009;87(5):515–522. doi: 10.1007/s00109-009-0452-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Morán M., Rivera H., Sánchez-Aragó M., Blázquez A., Merinero B., Ugalde C., Arenas J., Cuezva J.M., Martín M.A. Mitochondrial bioenergetics and dynamics interplay in complex I-deficient fibroblasts. Biochim. Biophys. Acta Mol. Basis Dis. 2010;1802(5):443–453. doi: 10.1016/j.bbadis.2010.02.001. [DOI] [PubMed] [Google Scholar]
  • 51.Luft R. The development of mitochondrial medicine. Proc. Natl. Acad. Sci. USA. 1994;91(19):8731–8738. doi: 10.1073/pnas.91.19.8731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Smeitink J., Ruitenbeek W., Lith T., Sengers R., Trijbels F., Wevers R., Sperl W., de Graaf R. Maturation of mitochondrial and other isoenzymes of creatine kinase in skeletal muscle of preterm born infants. Ann. Clin. Biochem. 1992;29(3):302–306. doi: 10.1177/000456329202900309. [DOI] [PubMed] [Google Scholar]
  • 53.Lin M.T., Beal M.F. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. 2006;443(7113):787–795. doi: 10.1038/nature05292. [DOI] [PubMed] [Google Scholar]
  • 54.Zeviani M., Carelli V. Mitochondrial disorders. Curr. Opin. Neurol. 2007;20(5):564–571. doi: 10.1097/WCO.0b013e3282ef58cd. [DOI] [PubMed] [Google Scholar]
  • 55.Aksenov M.Y., Tucker H.M., Nair P., Aksenova M.V., Butterfield D.A., Estus S., Markesbery W.R. The expression of several mitochondrial and nuclear genes encoding the subunits of electron transport chain enzyme complexes, cytochrome c oxidase, and NADH dehydrogenase, in different brain regions in Alzheimer’s disease. Neurochem. Res. 1999;24(6):767–774. doi: 10.1023/A:1020783614031. [DOI] [PubMed] [Google Scholar]
  • 56.Fukuyama R., Hatanpää K., Rapoport S.I., Chandrasekaran K. Gene expression of ND4, a subunit of complex I of oxidative phosphorylation in mitochondria, is decreased in temporal cortex of brains of Alzheimer’s disease patients. Brain Res. 1996;713(1-2):290–293. doi: 10.1016/0006-8993(95)01517-5. [DOI] [PubMed] [Google Scholar]
  • 57.Chandrasekaran K., Hatanpää K., Brady D.R., Rapoport S.I. Evidence for physiological down-regulation of brain oxidative phosphorylation in Alzheimer’s disease. Exp. Neurol. 1996;142(1):80–88. doi: 10.1006/exnr.1996.0180. [DOI] [PubMed] [Google Scholar]
  • 58.Parker W.D., Jr, Ba J.P., Filley C.M., Kleinschmidt-DeMasters B.K. Electron transport chain defects in Alzheimer’s disease brain. Neurology. 1994;44(6):1090–1096. doi: 10.1212/WNL.44.6.1090. [DOI] [PubMed] [Google Scholar]
  • 59.Manczak M., Park B.S., Jung Y., Reddy P.H. Differential expression of oxidative phosphorylation genes in patients with Alzheimer’s disease: Implications for early mitochondrial dysfunction and oxidative damage. Neuromol. Med. 2004;5(2):147–162. doi: 10.1385/NMM:5:2:147. [DOI] [PubMed] [Google Scholar]
  • 60.Vila M., Przedborski S. Targeting programmed cell death in neurodegenerative diseases. Nat. Rev. Neurosci. 2003;4(5):365–375. doi: 10.1038/nrn1100. [DOI] [PubMed] [Google Scholar]
  • 61.Dauer W., Przedborski S. Parkinson’s disease. Neuron. 2003;39(6):889–909. doi: 10.1016/S0896-6273(03)00568-3. [DOI] [PubMed] [Google Scholar]
  • 62.Betarbet R., Sherer T.B., MacKenzie G., Garcia-Osuna M., Panov A.V., Greenamyre J.T. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat. Neurosci. 2000;3(12):1301–1306. doi: 10.1038/81834. [DOI] [PubMed] [Google Scholar]
  • 63.Qi X., Lewin A.S., Hauswirth W.W., Guy J. Suppression of complex I gene expression induces optic neuropathy. Ann. Neurol. 2003;53(2):198–205. doi: 10.1002/ana.10426. [DOI] [PubMed] [Google Scholar]
  • 64.Fato R., Bergamini C., Leoni S., Strocchi P., Lenaz G. Generation of reactive oxygen species by mitochondrial complex I: Implications in neurodegeneration. Neurochem. Res. 2008;33(12):2487–2501. doi: 10.1007/s11064-008-9747-0. [DOI] [PubMed] [Google Scholar]
  • 65.Miyoshi H. Structure–activity relationships of some complex I inhibitors. Biochim. Biophys. Acta Bioenerg. 1998;1364(2):236–244. doi: 10.1016/S0005-2728(98)00030-9. [DOI] [PubMed] [Google Scholar]
  • 66.Degli Esposti M. Inhibitors of NADH–ubiquinone reductase: An overview. Biochim. Biophys. Acta Bioenerg. 1998;1364(2):222–235. doi: 10.1016/S0005-2728(98)00029-2. [DOI] [PubMed] [Google Scholar]
  • 67.Vila M., Jackson-Lewis V., Vukosavic S., Djaldetti R., Liberatore G., Offen D., Korsmeyer S.J., Przedborski S. Bax ablation prevents dopaminergic neurodegeneration in the 1-methyl- 4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. Proc. Natl. Acad. Sci. USA. 2001;98(5):2837–2842. doi: 10.1073/pnas.051633998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Chin M.H., Qian W.J., Wang H., Petyuk V.A., Bloom J.S., Sforza D.M., Laćan G., Liu D., Khan A.H., Cantor R.M., Bigelow D.J., Melega W.P., Camp D.G., II, Smith R.D., Smith D.J. Mitochondrial dysfunction, oxidative stress, and apoptosis revealed by proteomic and transcriptomic analyses of the striata in two mouse models of Parkinson’s disease. J. Proteome Res. 2008;7(2):666–677. doi: 10.1021/pr070546l. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Starkov A.A. The role of mitochondria in reactive oxygen species metabolism and signaling. Ann. N. Y. Acad. Sci. 2008;1147(1):37–52. doi: 10.1196/annals.1427.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Murphy M.P. How mitochondria produce reactive oxygen species. Biochem. J. 2009;417(1):1–13. doi: 10.1042/BJ20081386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Turrens J.F. Mitochondrial formation of reactive oxygen species. J. Physiol. 2003;552(2):335–344. doi: 10.1113/jphysiol.2003.049478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Verkaart S. Superoxide production is inversely related to complex I activity in inherited complex I deficiency. Biochim. Biophys. Acta Mol. Basis Dis. 1772;2007:373–381. doi: 10.1016/j.bbadis.2006.12.009. [DOI] [PubMed] [Google Scholar]
  • 73.Verkaart S., Koopman W.J.H., Cheek J., van Emst-de Vries S.E., van den Heuvel L.W.P.J., Smeitink J.A.M., Willems P.H.G.M. Mitochondrial and cytosolic thiol redox state are not detectably altered in isolated human NADH:ubiquinone oxidoreductase deficiency. Biochim. Biophys. Acta Mol. Basis Dis. 2007;1772(9):1041–1051. doi: 10.1016/j.bbadis.2007.05.004. [DOI] [PubMed] [Google Scholar]
  • 74.Hinson J.T., Fantin V.R., Schönberger J., Breivik N., Siem G., McDonough B., Sharma P., Keogh I., Godinho R., Santos F., Esparza A., Nicolau Y., Selvaag E., Cohen B.H., Hoppel C.L., Tranebjærg L., Eavey R.D., Seidman J.G., Seidman C.E. Missense mutations in the BCS1L gene as a cause of the Björnstad syndrome. N. Engl. J. Med. 2007;356(8):809–819. doi: 10.1056/NEJMoa055262. [DOI] [PubMed] [Google Scholar]
  • 75.Diaz F., Enríquez J.A., Moraes C.T. Cells lacking Rieske iron-sulfur protein have a reactive oxygen species-associated decrease in respiratory complexes I and IV. Mol. Cell. Biol. 2012;32(2):415–429. doi: 10.1128/MCB.06051-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Krause K.H., Bedard K. NOX enzymes in immuno-inflammatory pathologies. Semin. Immunopathol. 2008;30(3):193–194. doi: 10.1007/s00281-008-0127-2. [DOI] [PubMed] [Google Scholar]
  • 77.Andreyev A.Y., Kushnareva Y.E., Starkov A.A. Mitochondrial metabolism of reactive oxygen species. Biochemistry (Mosc.) 2005;70(2):200–214. doi: 10.1007/s10541-005-0102-7. [DOI] [PubMed] [Google Scholar]
  • 78.Dröge W. Free radicals in the physiological control of cell function. Physiol. Rev. 2002;82(1):47–95. doi: 10.1152/physrev.00018.2001. [DOI] [PubMed] [Google Scholar]
  • 79.Qin B., Cartier L., Dubois-Dauphin M., Li B., Serrander L., Krause K.H. A key role for the microglial NADPH oxidase in APP-dependent killing of neurons. Neurobiol. Aging. 2006;27(11):1577–1587. doi: 10.1016/j.neurobiolaging.2005.09.036. [DOI] [PubMed] [Google Scholar]
  • 80.Zhang Y., Dawson V.L., Dawson T.M. Oxidative stress and genetics in the pathogenesis of Parkinson’s disease. Neurobiol. Dis. 2000;7(4):240–250. doi: 10.1006/nbdi.2000.0319. [DOI] [PubMed] [Google Scholar]
  • 81.Andreyev A.Y., Kushnareva Y.E., Murphy A.N., Starkov A.A., Mitochondrial R.O.S. Metabolism: 10 years later. Biochem. Biokhimiia. 2015;80(5):517–531. doi: 10.1134/S0006297915050028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Liu Y., Fiskum G., Schubert D. Generation of reactive oxygen species by the mitochondrial electron transport chain. J. Neurochem. 2002;80(5):780–787. doi: 10.1046/j.0022-3042.2002.00744.x. [DOI] [PubMed] [Google Scholar]
  • 83.Mailer K. Superoxide radical as electron donor for oxidative phosphorylation of ADP. Biochem. Biophys. Res. Commun. 1990;170(1):59–64. doi: 10.1016/0006-291X(90)91240-S. [DOI] [PubMed] [Google Scholar]
  • 84.Smeitink J., van den Heuvel L., DiMauro S. The genetics and pathology of oxidative phosphorylation. Nat. Rev. Genet. 2001;2(5):342–352. doi: 10.1038/35072063. [DOI] [PubMed] [Google Scholar]
  • 85.Maker H.S., Weiss C., Silides D.J., Cohen G. Coupling of dopamine oxidation (monoamine oxidase activity) to glutathione oxidation via the generation of hydrogen peroxide in rat brain homogenates. J. Neurochem. 1981;36(2):589–593. doi: 10.1111/j.1471-4159.1981.tb01631.x. [DOI] [PubMed] [Google Scholar]
  • 86.Zoccarato F., Toscano P., Alexandre A. Dopamine-derived dopaminochrome promotes H2O2 release at mitochondrial complex I: Stimulation by rotenone, control by Ca2+, and relevance to Parkinson disease. J. Biol. Chem. 2005;280(16):15587–15594. doi: 10.1074/jbc.M500657200. [DOI] [PubMed] [Google Scholar]
  • 87.Guo J., Lemire B.D. The ubiquinone-binding site of the Saccharomyces cerevisiae succinate-ubiquinone oxidoreductase is a source of superoxide. J. Biol. Chem. 2003;278(48):47629–47635. doi: 10.1074/jbc.M306312200. [DOI] [PubMed] [Google Scholar]
  • 88.Brouillet E., Condé F., Beal M.F., Hantraye P. Replicating Huntington’s disease phenotype in experimental animals. Prog. Neurobiol. 1999;59(5):427–468. doi: 10.1016/S0301-0082(99)00005-2. [DOI] [PubMed] [Google Scholar]
  • 89.Liot G., Bossy B., Lubitz S., Kushnareva Y., Sejbuk N., Bossy-Wetzel E. Complex II inhibition by 3-NP causes mitochondrial fragmentation and neuronal cell death via an NMDA- and ROS-dependent pathway. Cell Death Differ. 2009;16(6):899–909. doi: 10.1038/cdd.2009.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Jenner P. Oxidative stress and Parkinson’s disease. Handb. Clin. Neurol. 2007;83:507–520. doi: 10.1016/S0072-9752(07)83024-7. [DOI] [PubMed] [Google Scholar]
  • 91.Chen V.T., Huang C.L., Lee Y.C., Liao W.C., Huang N.K. The roles of the thioredoxin system and peroxiredoxins in 1-methyl-4-phenyl-pyridinium ion-induced cytotoxicity in rat pheochromocytoma cells. Toxicol. In Vitro. 2010;24(6):1577–1583. doi: 10.1016/j.tiv.2010.06.010. [DOI] [PubMed] [Google Scholar]
  • 92.Reddy P.H., Beal M.F. Are mitochondria critical in the pathogenesis of Alzheimer’s disease? Brain Res. Brain Res. Rev. 2005;49(3):618–632. doi: 10.1016/j.brainresrev.2005.03.004. [DOI] [PubMed] [Google Scholar]
  • 93.Allanbutterfield D., Castegna A., Lauderback C., Drake J. Evidence that amyloid beta-peptide-induced lipid peroxidation and its sequelae in Alzheimer’s disease brain contribute to neuronal death1. Neurobiol. Aging. 2002;23(5):655–664. doi: 10.1016/S0197-4580(01)00340-2. [DOI] [PubMed] [Google Scholar]
  • 94.Esposito L., Raber J., Kekonius L., Yan F., Yu G.Q., Bien-Ly N., Puoliväli J., Scearce-Levie K., Masliah E., Mucke L. Reduction in mitochondrial superoxide dismutase modulates Alzheimer’s disease-like pathology and accelerates the onset of behavioral changes in human amyloid precursor protein transgenic mice. J. Neurosci. 2006;26(19):5167–5179. doi: 10.1523/JNEUROSCI.0482-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kim S.H., Fountoulakis M., Cairns N., Lubec G. Protein levels of human peroxiredoxin subtypes in brains of patients with Alzheimer’s disease and Down Syndrome. J. Neural Transm. Suppl. 2001;(61):223–235. doi: 10.1007/978-3-7091-6262-0_18. [DOI] [PubMed] [Google Scholar]
  • 96.Krapfenbauer K., Engidawork E., Cairns N., Fountoulakis M., Lubec G. Aberrant expression of peroxiredoxin subtypes in neurodegenerative disorders. Brain Res. 2003;967(1-2):152–160. doi: 10.1016/S0006-8993(02)04243-9. [DOI] [PubMed] [Google Scholar]
  • 97.Kirby J., Halligan E., Baptista M.J., Allen S., Heath P.R., Holden H., Barber S.C., Loynes C.A., Wood-Allum C.A., Lunec J., Shaw P.J. Mutant SOD1 alters the motor neuronal transcriptome: Implications for familial ALS. Brain. 2005;128(7):1686–1706. doi: 10.1093/brain/awh503. [DOI] [PubMed] [Google Scholar]
  • 98.Bosch M., Marí M., Herms A., Fernández A., Fajardo A., Kassan A., Giralt A., Colell A., Balgoma D., Barbero E., González-Moreno E., Matias N., Tebar F., Balsinde J., Camps M., Enrich C., Gross S.P., García-Ruiz C., Pérez-Navarro E., Fernández-Checa J.C., Pol A. Caveolin-1 deficiency causes cholesterol-dependent mitochondrial dysfunction and apoptotic susceptibility. Curr. Biol. 2011;21(8):681–686. doi: 10.1016/j.cub.2011.03.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Anderson S., Bankier A.T., Barrell B.G., de Bruijn M.H.L., Coulson A.R., Drouin J., Eperon I.C., Nierlich D.P., Roe B.A., Sanger F., Schreier P.H., Smith A.J.H., Staden R., Young I.G. Sequence and organization of the human mitochondrial genome. Nature. 1981;290(5806):457–465. doi: 10.1038/290457a0. [DOI] [PubMed] [Google Scholar]
  • 100.Shadel G.S., Horvath T.L. Mitochondrial ROS signaling in organismal homeostasis. Cell. 2015;163(3):560–569. doi: 10.1016/j.cell.2015.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Beal M.F. Mitochondria take center stage in aging and neurodegeneration. Ann. Neurol. 2005;58(4):495–505. doi: 10.1002/ana.20624. [DOI] [PubMed] [Google Scholar]
  • 102.Stuart J.A., Hashiguchi K., Wilson D.M., III, Copeland W.C., Souza-Pinto N.C., Bohr V.A. DNA base excision repair activities and pathway function in mitochondrial and cellular lysates from cells lacking mitochondrial DNA. Nucleic Acids Res. 2004;32(7):2181–2192. doi: 10.1093/nar/gkh533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Bohr V.A. Repair of oxidative DNA damage in nuclear and mitochondrial DNA, and some changes with aging in mammalian cells1,2 1Guest Editor: Miral Dizdaroglu 2This article is part of a series of reviews on “Oxidative DNA Damage and Repair.” The full list of papers may be found on the homepage of the journal. Free Radic. Biol. Med. 2002;32(9):804-812. doi: 10.1016/S0891-5849(02)00787-6. [DOI] [PubMed] [Google Scholar]
  • 104.Coskun P.E., Beal M.F., Wallace D.C. Alzheimer’s brains harbor somatic mtDNA control-region mutations that suppress mitochondrial transcription and replication. Proc. Natl. Acad. Sci. USA. 2004;101(29):10726–10731. doi: 10.1073/pnas.0403649101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Hutchin T.P., Heath P.R., Pearson R.C.A., Sinclair A.J. Mitochondrial DNA mutations in Alzheimer’s disease. Biochem. Biophys. Res. Commun. 1997;241(2):221–225. doi: 10.1006/bbrc.1997.7793. [DOI] [PubMed] [Google Scholar]
  • 106.Wiedemann F.R., Manfredi G., Mawrin C., Beal M.F., Schon E.A. Mitochondrial DNA and respiratory chain function in spinal cords of ALS patients. J. Neurochem. 2002;80(4):616–625. doi: 10.1046/j.0022-3042.2001.00731.x. [DOI] [PubMed] [Google Scholar]
  • 107.Hamblet N.S., Ragland B., Ali M., Conyers B., Castora F.J. Mutations in mitochondrial-encoded cytochromec oxidase subunits I, II, and III genes detected in Alzheimer’s disease using single-strand conformation polymorphism. Electrophoresis. 2006;27(2):398–408. doi: 10.1002/elps.200500420. [DOI] [PubMed] [Google Scholar]
  • 108.Cardoso S.M., Santana I., Swerdlow R.H., Oliveira C.R. Mitochondria dysfunction of Alzheimer’s disease cybrids enhances Aβ toxicity. J. Neurochem. 2004;89(6):1417–1426. doi: 10.1111/j.1471-4159.2004.02438.x. [DOI] [PubMed] [Google Scholar]
  • 109.Richter G., Sonnenschein A., Grünewald T., Reichmann H., Janetzky B. Novel mitochondrial DNA mutations in Parkinson’s disease. J. Neural Transm. (Vienna) 2002;109(5-6):721–729. doi: 10.1007/s007020200060. [DOI] [PubMed] [Google Scholar]
  • 110.Swerdlow R.H., Parks J.K., Davis J.N., II, Cassarino D.S., Trimmer P.A., Currie L.J., Dougherty J., Bridges W.S., Bennett J.P., Jr, Wooten G.F., Parker W.D. Matrilineal inheritance of complex I dysfunction in a multigenerational Parkinson’s disease family. Ann. Neurol. 1998;44(6):873–881. doi: 10.1002/ana.410440605. [DOI] [PubMed] [Google Scholar]
  • 111.Krishnan K.J., Ratnaike T.E., De Gruyter H.L.M., Jaros E., Turnbull D.M. Mitochondrial DNA deletions cause the biochemical defect observed in Alzheimer’s disease. Neurobiol. Aging. 2012;33(9):2210–2214. doi: 10.1016/j.neurobiolaging.2011.08.009. [DOI] [PubMed] [Google Scholar]
  • 112.Murakami T., Nagai M., Miyazaki K., Morimoto N., Ohta Y., Kurata T., Takehisa Y., Kamiya T., Abe K. Early decrease of mitochondrial DNA repair enzymes in spinal motor neurons of presymptomatic transgenic mice carrying a mutant SOD1 gene. Brain Res. 2007;1150:182–189. doi: 10.1016/j.brainres.2007.02.057. [DOI] [PubMed] [Google Scholar]
  • 113.Israelson A., Arbel N., Da Cruz S., Ilieva H., Yamanaka K., Shoshan-Barmatz V., Cleveland D.W. Misfolded mutant SOD1 directly inhibits VDAC1 conductance in a mouse model of inherited ALS. Neuron. 2010;67(4):575–587. doi: 10.1016/j.neuron.2010.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Warita H., Hayashi T., Murakami T., Manabe Y., Abe K. Oxidative damage to mitochondrial DNA in spinal motoneurons of transgenic ALS mice. Brain Res. Mol. Brain Res. 2001;89(1-2):147–152. doi: 10.1016/S0169-328X(01)00029-8. [DOI] [PubMed] [Google Scholar]
  • 115.Dhaliwal G.K., Grewal R.P. Mitochondrial DNA deletion mutation levels are elevated in ALS brains. Neuroreport. 2000;11(11):2507–2509. doi: 10.1097/00001756-200008030-00032. [DOI] [PubMed] [Google Scholar]
  • 116.Tsai F.C., Seki A., Yang H.W., Hayer A., Carrasco S., Malmersjö S., Meyer T. A polarized Ca2+, diacylglycerol and STIM1 signalling system regulates directed cell migration. Nat. Cell Biol. 2014;16(2):133–144. doi: 10.1038/ncb2906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Yang S., Huang X.Y. Ca2+ influx through L-type Ca2+ channels controls the trailing tail contraction in growth factor-induced fibroblast cell migration. J. Biol. Chem. 2005;280(29):27130–27137. doi: 10.1074/jbc.M501625200. [DOI] [PubMed] [Google Scholar]
  • 118.Hartmann J., Verkhratsky A. Relations between intracellular Ca2+ stores and store-operated Ca2+ entry in primary cultured human glioblastoma cells. J. Physiol. 1998;513(Pt 2):411–424. doi: 10.1111/j.1469-7793.1998.411bb.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Roos D., Seeger R., Puntel R., Vargas Barbosa N. Role of calcium and mitochondria in MeHg-mediated cytotoxicity. J. Biomed. Biotechnol. 2012;2012:1–15. doi: 10.1155/2012/248764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Imbert N., Cognard C., Duport G., Guillou C., Raymond G. Abnormal calcium homeostasis in Duchenne muscular dystrophy myotubes contracting in vitro. Cell Calcium. 1995;18(3):177–186. doi: 10.1016/0143-4160(95)90062-4. [DOI] [PubMed] [Google Scholar]
  • 121.Xiong J., Camello P.J., Verkhratsky A., Toescu E.C. Mitochondrial polarisation status and Ca2+ signalling in rat cerebellar granule neurones aged in vitro. Neurobiol. Aging. 2004;25(3):349–359. doi: 10.1016/S0197-4580(03)00123-4. [DOI] [PubMed] [Google Scholar]
  • 122.Tang S., Wang X., Shen Q., Yang X., Yu C., Cai C., Cai G., Meng X., Zou F. Mitochondrial Ca2+ uniporter is critical for store-operated Ca2+ entry-dependent breast cancer cell migration. Biochem. Biophys. Res. Commun. 2015;458(1):186–193. doi: 10.1016/j.bbrc.2015.01.092. [DOI] [PubMed] [Google Scholar]
  • 123.Panov A.V., Lund S., Greenamyre J.T. Ca2+-induced permeability transition in human lymphoblastoid cell mitochondria from normal and Huntington?s disease individuals. Mol. Cell. Biochem. 2005;269(1):143–152. doi: 10.1007/s11010-005-3454-9. [DOI] [PubMed] [Google Scholar]
  • 124.Quintanilla R.A., Johnson G.V.W. Role of mitochondrial dysfunction in the pathogenesis of Huntington’s disease. Brain Res. Bull. 2009;80(4-5):242–247. doi: 10.1016/j.brainresbull.2009.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Jaiswal M.K., Zech W.D., Goos M., Leutbecher C., Ferri A., Zippelius A., Carrì M.T., Nau R., Keller B.U. Impairment of mitochondrial calcium handling in a mtSOD1 cell culture model of motoneuron disease. BMC Neurosci. 2009;10(1):64. doi: 10.1186/1471-2202-10-64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Sheehan J.P., Swerdlow R.H., Miller S.W., Davis R.E., Parks J.K., Parker W.D., Tuttle J.B. Calcium homeostasis and reactive oxygen species production in cells transformed by mitochondria from individuals with sporadic Alzheimer’s disease. J. Neurosci. 1997;17(12):4612–4622. doi: 10.1523/JNEUROSCI.17-12-04612.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Du H., Guo L., Zhang W., Rydzewska M., Yan S. Cyclophilin D deficiency improves mitochondrial function and learning/memory in aging Alzheimer disease mouse model. Neurobiol. Aging. 2011;32(3):398–406. doi: 10.1016/j.neurobiolaging.2009.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Alevriadou B.R., Patel A., Noble M., Ghosh S., Gohil V.M., Stathopulos P.B., Madesh M. Molecular nature and physiological role of the mitochondrial calcium uniporter channel. Am. J. Physiol. Cell Physiol. 2021;320(4):C465–C482. doi: 10.1152/ajpcell.00502.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Petersén Å., Castilho R.F., Hansson O., Wieloch T., Brundin P. Oxidative stress, mitochondrial permeability transition and activation of caspases in calcium ionophore A23187-induced death of cultured striatal neurons. Brain Res. 2000;857(1-2):20–29. doi: 10.1016/S0006-8993(99)02320-3. [DOI] [PubMed] [Google Scholar]
  • 130.Halestrap A.P. Calcium, mitochondria and reperfusion injury: A pore way to die. Biochem. Soc. Trans. 2006;34(2):232–237. doi: 10.1042/BST0340232. [DOI] [PubMed] [Google Scholar]
  • 131.Halestrap A.P., Griffiths E.J., Connern C.P. Mitochondrial calcium handling and oxidative stress. Biochem. Soc. Trans. 1993;21(2):353–358. doi: 10.1042/bst0210353. [DOI] [PubMed] [Google Scholar]
  • 132.Kantrow S.P., Tatro L.G., Piantadosi C.A. Oxidative stress and adenine nucleotide control of mitochondrial permeability transition. Free Radic. Biol. Med. 2000;28(2):251–260. doi: 10.1016/S0891-5849(99)00238-5. [DOI] [PubMed] [Google Scholar]
  • 133.Leung A.W.C., Halestrap A.P. Recent progress in elucidating the molecular mechanism of the mitochondrial permeability transition pore. Biochim. Biophys. Acta Bioenerg. 2008;1777(7-8):946–952. doi: 10.1016/j.bbabio.2008.03.009. [DOI] [PubMed] [Google Scholar]
  • 134.Du H., Yan S.S. Mitochondrial permeability transition pore in Alzheimer’s disease: Cyclophilin D and amyloid beta. Biochim. Biophys. Acta Mol. Basis Dis. 2010;1802(1):198–204. doi: 10.1016/j.bbadis.2009.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Brustovetsky N., Brustovetsky T., Purl K.J., Capano M., Crompton M., Dubinsky J.M. Increased susceptibility of striatal mitochondria to calcium-induced permeability transition. J. Neurosci. 2003;23(12):4858–4867. doi: 10.1523/JNEUROSCI.23-12-04858.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Xu W., Marseglia A., Ferrari C., Wang H.X. Alzheimer’s disease: A clinical perspective. Neurodegener. Dis. 2013 doi: 10.5772/54539. [DOI] [Google Scholar]
  • 137.Pathak D., Berthet A., Nakamura K. Energy failure. Ann. Neurol. 2013;74(4):506–516. doi: 10.1002/ana.24014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Sun N., Youle R.J., Finkel T. The mitochondrial basis of aging. Mol. Cell. 2016;61(5):654–666. doi: 10.1016/j.molcel.2016.01.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Zhao X.Y., Lu M.H., Yuan D.J., Xu D.E., Yao P.P., Ji W.L., Chen H., Liu W.L., Yan C.X., Xia Y.Y., Li S., Tao J., Ma Q.H. Mitochondrial dysfunction in neural injury. Front. Neurosci. 2019;13:30. doi: 10.3389/fnins.2019.00030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Fricker M., Tolkovsky A.M., Borutaite V., Coleman M., Brown G.C. Neuronal cell death. Physiol. Rev. 2018;98(2):813–880. doi: 10.1152/physrev.00011.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Johri A., Beal M.F. Mitochondrial dysfunction in neurodegenerative diseases. J. Pharmacol. Exp. Ther. 2012;342(3):619–630. doi: 10.1124/jpet.112.192138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Hoekstra J.G., Montine K.S., Zhang J., Montine T.J. Mitochondrial therapeutics in Alzheimer’s disease and Parkinson’s disease. Alzheimers Res. Ther. 2011;3(3):21. doi: 10.1186/alzrt83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Monzio Compagnoni G., Di Fonzo A., Corti S., Comi G.P., Bresolin N., Masliah E. The role of mitochondria in neurodegenerative diseases: The lesson from Alzheimer’s disease and Parkinson’s disease. Mol. Neurobiol. 2020;57(7):2959–2980. doi: 10.1007/s12035-020-01926-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Hroudová J., Singh N., Fišar Z. Mitochondrial dysfunctions in neurodegenerative diseases: Relevance to Alzheimer’s disease. BioMed Res. Int. 2014;2014:1–9. doi: 10.1155/2014/175062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Sims N.R., Muyderman H. Mitochondria, oxidative metabolism and cell death in stroke. Biochim. Biophys. Acta Mol. Basis Dis. 2010;1802(1):80–91. doi: 10.1016/j.bbadis.2009.09.003. [DOI] [PubMed] [Google Scholar]
  • 146.Kish S.J., Bergeron C., Rajput A., Dozic S., Mastrogiacomo F., Chang L.J., Wilson J.M., DiStefano L.M., Nobrega J.N. Brain cytochrome oxidase in Alzheimer’s disease. J. Neurochem. 1992;59(2):776–779. doi: 10.1111/j.1471-4159.1992.tb09439.x. [DOI] [PubMed] [Google Scholar]
  • 147.Maurer I., Zierz S., Möller H.J. A selective defect of cytochrome c oxidase is present in brain of Alzheimer disease patients. Neurobiol. Aging. 2000;21(3):455–462. doi: 10.1016/S0197-4580(00)00112-3. [DOI] [PubMed] [Google Scholar]
  • 148.Siasos G., Tsigkou V., Kosmopoulos M., Theodosiadis D., Simantiris S., Tagkou N.M., Tsimpiktsioglou A., Stampouloglou P.K., Oikonomou E., Mourouzis K., Philippou A., Vavuranakis M., Stefanadis C., Tousoulis D., Papavassiliou A.G. Mitochondria and cardiovascular diseases—from pathophysiology to treatment. Ann. Transl. Med. 2018;6(12):256. doi: 10.21037/atm.2018.06.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Moreira O.C., Estébanez B., Martínez-Florez S., Paz J.A., Cuevas M.J., González-Gallego J. Mitochondrial function and mitophagy in the elderly: Effects of exercise. Oxid. Med. Cell. Longev. 2017;2017:1–13. doi: 10.1155/2017/2012798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Chen J.Q., Cammarata P.R., Baines C.P., Yager J.D. Regulation of mitochondrial respiratory chain biogenesis by estrogens/estrogen receptors and physiological, pathological and pharmacological implications. Biochim. Biophys. Acta Mol. Cell Res. 2009;1793(10):1540–1570. doi: 10.1016/j.bbamcr.2009.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Prossnitz E.R., Barton M. The G-protein-coupled estrogen receptor GPER in health and disease. Nat. Rev. Endocrinol. 2011;7(12):715–726. doi: 10.1038/nrendo.2011.122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Brann D.W., Dhandapani K., Wakade C., Mahesh V.B., Khan M.M. Neurotrophic and neuroprotective actions of estrogen: Basic mechanisms and clinical implications. Steroids. 2007;72(5):381–405. doi: 10.1016/j.steroids.2007.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Lejri I., Grimm A., Eckert A. Mitochondria, estrogen and female brain aging. Front. Aging Neurosci. 2018;10:124. doi: 10.3389/fnagi.2018.00124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Fetisova E., Chernyak B., Korshunova G., Muntyan M., Skulachev V. Mitochondria-targeted Antioxidants as a Prospective Therapeutic Strategy for Multiple Sclerosis. Curr. Med. Chem. 2017;24(19):2086–2114. doi: 10.2174/0929867324666170316114452. [DOI] [PubMed] [Google Scholar]
  • 155.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(4):891–909. doi: 10.1042/BST20170501. [DOI] [PubMed] [Google Scholar]
  • 156.Fão L., Rego A.C. Mitochondrial and redox-based therapeutic strategies in Huntington’s disease. Antioxid. Redox Signal. 2021;34(8):650–673. doi: 10.1089/ars.2019.8004. [DOI] [PubMed] [Google Scholar]
  • 157.Van Giau V., An S.S.A., Hulme J.P. Mitochondrial therapeutic interventions in Alzheimer’s disease. J. Neurol. Sci. 2018;395:62–70. doi: 10.1016/j.jns.2018.09.033. [DOI] [PubMed] [Google Scholar]
  • 158.Zinovkin R.A., Zamyatnin A.A. Mitochondria-targeted drugs. Curr. Mol. Pharmacol. 2019;12(3):202–214. doi: 10.2174/1874467212666181127151059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Burns R.J., Smith R.A.J., Murphy M.P. Synthesis and characterization of thiobutyltriphenylphosphonium bromide, a novel thiol reagent targeted to the mitochondrial matrix. Arch. Biochem. Biophys. 1995;322(1):60–68. doi: 10.1006/abbi.1995.1436. [DOI] [PubMed] [Google Scholar]
  • 160.Adam-Vizi V., Chinopoulos C. Bioenergetics and the formation of mitochondrial reactive oxygen species. Trends Pharmacol. Sci. 2006;27(12):639–645. doi: 10.1016/j.tips.2006.10.005. [DOI] [PubMed] [Google Scholar]
  • 161.Zorova L.D., Popkov V.A., Plotnikov E.Y., Silachev D.N., Pevzner I.B., Jankauskas S.S., Babenko V.A., Zorov S.D., Balakireva A.V., Juhaszova M., Sollott S.J., Zorov D.B. Mitochondrial membrane potential. Anal. Biochem. 2018;552:50–59. doi: 10.1016/j.ab.2017.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Mileykovskaya E., Dowhan W. Cardiolipin-dependent formation of mitochondrial respiratory supercomplexes. Chem. Phys. Lipids. 2014;179:42–48. doi: 10.1016/j.chemphyslip.2013.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Kang Y., Fielden L.F., Stojanovski D. Mitochondrial protein transport in health and disease. Semin. Cell Dev. Biol. 2018;76:142–153. doi: 10.1016/j.semcdb.2017.07.028. [DOI] [PubMed] [Google Scholar]
  • 164.Korshunov S.S., Skulachev V.P., Starkov A.A. High protonic potential actuates a mechanism of production of reactive oxygen species in mitochondria. FEBS Lett. 1997;416(1):15–18. doi: 10.1016/S0014-5793(97)01159-9. [DOI] [PubMed] [Google Scholar]
  • 165.Antonenko Y.N., Avetisyan A.V., Bakeeva L.E., Chernyak B.V., Chertkov V.A., Domnina L.V., Ivanova O.Y., Izyumov D.S., Khailova L.S., Klishin S.S., Korshunova G.A., Lyamzaev K.G., Muntyan M.S., Nepryakhina O.K., Pashkovskaya A.A., Pletjushkina O.Y., Pustovidko A.V., Roginsky V.A., Rokitskaya T.I., Ruuge E.K., Saprunova V.B., Severina I.I., Simonyan R.A., Skulachev I.V., Skulachev M.V., Sumbatyan N.V., Sviryaeva I.V., Tashlitsky V.N., Vassiliev J.M., Vyssokikh M.Y., Yaguzhinsky L.S., Zamyatnin A.A., Jr, Skulachev V.P. Mitochondria-targeted plastoquinone derivatives as tools to interrupt execution of the aging program. 1. Cationic plastoquinone derivatives: Synthesis and in vitro studies. Biochemistry (Mosc.) 2008;73(12):1273–1287. doi: 10.1134/S0006297908120018. [DOI] [PubMed] [Google Scholar]
  • 166.Fink B.D., Herlein J.A., Yorek M.A., Fenner A.M., Kerns R.J., Sivitz W.I. Bioenergetic effects of mitochondrial-targeted coenzyme Q analogs in endothelial cells. J. Pharmacol. Exp. Ther. 2012;342(3):709–719. doi: 10.1124/jpet.112.195586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Nickel A., Kohlhaas M., Maack C. Mitochondrial reactive oxygen species production and elimination. J. Mol. Cell. Cardiol. 2014;73:26–33. doi: 10.1016/j.yjmcc.2014.03.011. [DOI] [PubMed] [Google Scholar]
  • 168.Chiurchiù V., Orlacchio A., Maccarrone M. Is modulation of oxidative stress an answer? The state of the art of redox therapeutic actions in neurodegenerative diseases. Oxid. Med. Cell. Longev. 2016;2016:1–11. doi: 10.1155/2016/7909380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Bravo L. Polyphenols: Chemistry, dietary sources, metabolism, and nutritional significance. Nutr. Rev. 1998;56(11):317–333. doi: 10.1111/j.1753-4887.1998.tb01670.x. [DOI] [PubMed] [Google Scholar]
  • 170.Rudra A., Arvind I., Mehra R. Polyphenols: Types, sources and therapeutic applications. Int. J. Home Sci. 2021;7(3):69–75. doi: 10.22271/23957476.2021.v7.i3a.1182. [DOI] [Google Scholar]
  • 171.Leri M., Scuto M., Ontario M.L., Calabrese V., Calabrese E.J., Bucciantini M., Stefani M. Healthy effects of plant polyphenols: Molecular mechanisms. Int. J. Mol. Sci. 2020;21(4):1250. doi: 10.3390/ijms21041250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Losada-Barreiro S., Bravo-Díaz C. Free radicals and polyphenols: The redox chemistry of neurodegenerative diseases. Eur. J. Med. Chem. 2017;133:379–402. doi: 10.1016/j.ejmech.2017.03.061. [DOI] [PubMed] [Google Scholar]
  • 173.Miquel S., Champ C., Day J., Aarts E., Bahr B.A., Bakker M., Bánáti D., Calabrese V., Cederholm T., Cryan J., Dye L., Farrimond J.A., Korosi A., Layé S., Maudsley S., Milenkovic D., Mohajeri M.H., Sijben J., Solomon A., Spencer J.P.E., Thuret S., Vanden B.W., Vauzour D., Vellas B., Wesnes K., Willatts P., Wittenberg R., Geurts L. Poor cognitive ageing: Vulnerabilities, mechanisms and the impact of nutritional interventions. Ageing Res. Rev. 2018;42:40–55. doi: 10.1016/j.arr.2017.12.004. [DOI] [PubMed] [Google Scholar]
  • 174.Franco R., Navarro G., Martínez-Pinilla E. Plant-derived compounds, vitagens, vitagenes and mitochondrial function. PharmaNutrition. 2022;19:100287. doi: 10.1016/j.phanu.2021.100287. [DOI] [Google Scholar]
  • 175.Trovato Salinaro A., Cornelius C., Koverech G., Koverech A., Scuto M., Lodato F., Fronte V., Muccilli V., Reibaldi M., Longo A., Uva M.G., Calabrese V. Cellular stress response, redox status, and vitagenes in glaucoma: A systemic oxidant disorder linked to Alzheimer’s disease. Front. Pharmacol. 2014;5:129. doi: 10.3389/fphar.2014.00129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Wakabayashi N., Itoh K., Wakabayashi J., Motohashi H., Noda S., Takahashi S., Imakado S., Kotsuji T., Otsuka F., Roop D.R., Harada T., Engel J.D., Yamamoto M. Keap1-null mutation leads to postnatal lethality due to constitutive Nrf2 activation. Nat. Genet. 2003;35(3):238–245. doi: 10.1038/ng1248. [DOI] [PubMed] [Google Scholar]
  • 177.Calabrese E.J., Kozumbo W.J. The hormetic dose-response mechanism: Nrf2 activation. Pharmacol. Res. 2021;167:105526. doi: 10.1016/j.phrs.2021.105526. [DOI] [PubMed] [Google Scholar]
  • 178.Calabrese E.J. Hormesis: Principles and applications. Homeopathy. 2015;104(2):69–82. doi: 10.1016/j.homp.2015.02.007. [DOI] [PubMed] [Google Scholar]
  • 179.Mattson M.P. Hormesis and disease resistance: Activation of cellular stress response pathways. Hum. Exp. Toxicol. 2008;27(2):155–162. doi: 10.1177/0960327107083417. [DOI] [PubMed] [Google Scholar]
  • 180.Cornelius C., Trovato S.A., Scuto M., Fronte V., Cambria M.T., Pennisi M., Bella R., Milone P., Graziano A., Crupi R., Cuzzocrea S., Pennisi G., Calabrese V. Cellular stress response, sirtuins and UCP proteins in Alzheimer disease: Role of vitagenes. Immun. Ageing. 2013;10(1):41. doi: 10.1186/1742-4933-10-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Mancuso C., Santangelo R., Calabrese V. The heme oxygenase/biliverdin reductase system: A potential drug target in Alzheimer’s disease. J. Biol. Regul. Homeost. Agents. 2013;27(2) Suppl.:75–87. [PubMed] [Google Scholar]
  • 182.Pilipenko V., Narbute K., Amara I., Trovato A., Scuto M., Pupure J., Jansone B., Poikans J., Bisenieks E., Klusa V., Calabrese V. GABA‐containing compound gammapyrone protects against brain impairments in Alzheimer’s disease model male rats and prevents mitochondrial dysfunction in cell culture. J. Neurosci. Res. 2019;97(6):708–726. doi: 10.1002/jnr.24396. [DOI] [PubMed] [Google Scholar]
  • 183.Crompton M., Ellinger H., Costi A. Inhibition by cyclosporin A of a Ca2+-dependent pore in heart mitochondria activated by inorganic phosphate and oxidative stress. Biochem. J. 1988;255(1):357–360. [PMC free article] [PubMed] [Google Scholar]
  • 184.Briston T., Selwood D.L., Szabadkai G., Duchen M.R. Mitochondrial permeability transition: A molecular lesion with multiple drug targets. Trends Pharmacol. Sci. 2019;40(1):50–70. doi: 10.1016/j.tips.2018.11.004. [DOI] [PubMed] [Google Scholar]
  • 185.Amanakis G., Murphy E., Cyclophilin D., Cyclophilin D. An integrator of mitochondrial function. Front. Physiol. 2020;11:595. doi: 10.3389/fphys.2020.00595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Baines C.P., Kaiser R.A., Purcell N.H., Blair N.S., Osinska H., Hambleton M.A., Brunskill E.W., Sayen M.R., Gottlieb R.A., Dorn G.W., II, Robbins J., Molkentin J.D. Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature. 2005;434(7033):658–662. doi: 10.1038/nature03434. [DOI] [PubMed] [Google Scholar]
  • 187.Baines C.P., Gutiérrez-Aguilar M. The still uncertain identity of the channel-forming unit(s) of the mitochondrial permeability transition pore. Cell Calcium. 2018;73:121–130. doi: 10.1016/j.ceca.2018.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Kalani K., Yan S.F., Yan S.S. Mitochondrial permeability transition pore: A potential drug target for neurodegeneration. Drug Discov. Today. 2018;23(12):1983–1989. doi: 10.1016/j.drudis.2018.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Du H., Guo L., Fang F., Chen D., Sosunov A. A.; M McKhann, G.; Yan, Y.; Wang, C.; Zhang, H.; Molkentin, J.D.; Gunn-Moore, F.J.; Vonsattel, J.P.; Arancio, O.; Chen, J.X.; Yan, S.D. Cyclophilin D deficiency attenuates mitochondrial and neuronal perturbation and ameliorates learning and memory in Alzheimer’s disease. Nat. Med. 2008;14(10):1097–1105. doi: 10.1038/nm.1868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Thomas B., Banerjee R., Starkova N.N., Zhang S.F., Calingasan N.Y., Yang L., Wille E., Lorenzo B.J., Ho D.J., Beal M.F., Starkov A. Mitochondrial permeability transition pore component cyclophilin D distinguishes nigrostriatal dopaminergic death paradigms in the MPTP mouse model of Parkinson’s disease. Antioxid. Redox Signal. 2012;16(9):855–868. doi: 10.1089/ars.2010.3849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Brouhard B.H., Graham R.M. Cyclosporine: Mechanisms of action and toxicity. Cleve. Clin. J. Med. 1994;61(4):308–313. doi: 10.3949/ccjm.61.4.308. [DOI] [PubMed] [Google Scholar]
  • 192.Valasani K.R., Chaney M.O., Day V.W. ShiDu Yan, S. S. Acetylcholinesterase inhibitors: Structure based design, synthesis, pharmacophore modeling, and virtual screening. J. Chem. Inf. Model. 2013;53(8):2033–2046. doi: 10.1021/ci400196z. [DOI] [PubMed] [Google Scholar]
  • 193.Ahmed-Belkacem A., Colliandre L., Ahnou N., Nevers Q., Gelin M., Bessin Y., Brillet R., Cala O., Douguet D., Bourguet W., Krimm I., Pawlotsky J.M., Guichou J.F. Fragment-based discovery of a new family of non-peptidic small-molecule cyclophilin inhibitors with potent antiviral activities. Nat. Commun. 2016;7(1):12777. doi: 10.1038/ncomms12777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Guo H., Wang F., Yu K., Chen J., Bai D., Chen K., Shen X., Jiang H. Novel cyclophilin D inhibitors derived from quinoxaline exhibit highly inhibitory activity against rat mitochondrial swelling and Ca2+ uptake/release. Acta Pharmacol. Sin. 2005;26(10):1201–1211. doi: 10.1111/j.1745-7254.2005.00189.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Hudry E., Vandenberghe L.H. Therapeutic AAV gene transfer to the nervous system: A clinical reality. Neuron. 2019;101(5):839–862. doi: 10.1016/j.neuron.2019.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Weinberg M.S., Samulski R.J., McCown T.J. Adeno-associated virus (AAV) gene therapy for neurological disease. Neuropharmacology. 2013;69:82–88. doi: 10.1016/j.neuropharm.2012.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Chen W., Hu Y., Ju D. Gene therapy for neurodegenerative disorders: Advances, insights and prospects. Acta Pharm. Sin. B. 2020;10(8):1347–1359. doi: 10.1016/j.apsb.2020.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Rafii M.S., Tuszynski M.H., Thomas R.G., Barba D., Brewer J.B., Rissman R.A., Siffert J., Aisen P.S. Adeno-associated viral vector (Serotype 2)–nerve growth factor for patients with Alzheimer disease. JAMA Neurol. 2018;75(7):834–841. doi: 10.1001/jamaneurol.2018.0233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Nilsson P., Iwata N., Muramatsu S., Tjernberg L.O., Winblad B., Saido T.C. Gene therapy in Alzheimer’s disease - potential for disease modification. J. Cell. Mol. Med. 2010;14(4):741–757. doi: 10.1111/j.1582-4934.2010.01038.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Choong C.J., Mochizuki H. Gene therapy targeting mitochondrial pathway in Parkinson’s disease. J. Neural Transm. 2017;124(2):193–207. doi: 10.1007/s00702-016-1616-4. [DOI] [PubMed] [Google Scholar]
  • 201.Zhang L., Reyes A., Wang X. The role of mitochondria-targeted antioxidant MitoQ in neurodegenerative disease. Mol. Cell. Ther. 2018:1–8. [Google Scholar]
  • 202.Ross M.F., Kelso G.F., Blaikie F.H., James A.M., Cochemé H.M., Filipovska A., Da Ros T., Hurd T.R., Smith R.A.J., Murphy M.P. Lipophilic triphenylphosphonium cations as tools in mitochondrial bioenergetics and free radical biology. Biochemistry (Mosc.) 2005;70(2):222–230. doi: 10.1007/s10541-005-0104-5. [DOI] [PubMed] [Google Scholar]
  • 203.Kelso G.F., Porteous C.M., Coulter C.V., Hughes G., Porteous W.K., Ledgerwood E.C., Smith R.A.J., Murphy M.P. Selective targeting of a redox-active ubiquinone to mitochondria within cells: Antioxidant and antiapoptotic properties. J. Biol. Chem. 2001;276(7):4588–4596. doi: 10.1074/jbc.M009093200. [DOI] [PubMed] [Google Scholar]
  • 204.Oyewole A.O., Birch-Machin M.A. Mitochondria‐targeted antioxidants. FASEB J. 2015;29(12):4766–4771. doi: 10.1096/fj.15-275404. [DOI] [PubMed] [Google Scholar]
  • 205.Shinn L.J., Lagalwar S. Treating neurodegenerative disease with antioxidants: Efficacy of the bioactive phenol resveratrol and mitochondrial-targeted MitoQ and SkQ. Antioxidants. 2021;10(4) doi: 10.3390/antiox10040573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Solesio M.E., Prime T.A., Logan A., Murphy M.P., del Mar Arroyo-Jimenez M., Jordán J., Galindo M.F. The mitochondria-targeted anti-oxidant MitoQ reduces aspects of mitochondrial fission in the 6-OHDA cell model of Parkinson’s disease. Biochim. Biophys. Acta Mol. Basis Dis. 2013;1832(1):174–182. doi: 10.1016/j.bbadis.2012.07.009. [DOI] [PubMed] [Google Scholar]
  • 207.Ghosh A., Chandran K., Kalivendi S.V., Joseph J., Antholine W.E., Hillard C.J., Kanthasamy A., Kanthasamy A., Kalyanaraman B. Neuroprotection by a mitochondria-targeted drug in a Parkinson’s disease model. Free Radic. Biol. Med. 2010;49(11):1674–1684. doi: 10.1016/j.freeradbiomed.2010.08.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.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(44):15703–15715. doi: 10.1523/JNEUROSCI.0552-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Gane E.J., Weilert F., Orr D.W., Keogh G.F., Gibson M., Lockhart M.M., Frampton C.M., Taylor K.M., Smith R.A., Murphy M.P. The mitochondria-targeted anti-oxidant mitoquinone decreases liver damage in a phase II study of hepatitis C patients. Liver Int. 2010;30(7):1019–1026. doi: 10.1111/j.1478-3231.2010.02250.x. [DOI] [PubMed] [Google Scholar]
  • 210.Ulatowski L.M., Manor D. Vitamin E and neurodegeneration. Neurobiol. Dis. 2015;84:78–83. doi: 10.1016/j.nbd.2015.04.002. [DOI] [PubMed] [Google Scholar]
  • 211.Sokol R.J. Vitamin E deficiency and neurologic disease. Annu. Rev. Nutr. 1988;8(1):351–373. doi: 10.1146/annurev.nu.08.070188.002031. [DOI] [PubMed] [Google Scholar]
  • 212.Bourre J.M., Clement M. Kinetics of rat peripheral nerve, forebrain and cerebellum α-tocopherol depletion: Comparison with different organs. J. Nutr. 1991;121(8):1204–1207. doi: 10.1093/jn/121.8.1204. [DOI] [PubMed] [Google Scholar]
  • 213.Gohil K., Oommen S., Quach H.T., Vasu V.T., Aung H.H., Schock B., Cross C.E., Vatassery G.T. Mice lacking α-tocopherol transfer protein gene have severe α-tocopherol deficiency in multiple regions of the central nervous system. Brain Res. 2008;1201:167–176. doi: 10.1016/j.brainres.2008.01.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Oppedisano F., Maiuolo J., Gliozzi M., Musolino V., Carresi C., Nucera S., Scicchitano M., Scarano F., Bosco F., Macrì R., Ruga S., Zito M.C., Palma E., Muscoli C., Mollace V. The potential for natural antioxidant supplementation in the early stages of neurodegenerative disorders. Int. J. Mol. Sci. 2020;21(7):2618. doi: 10.3390/ijms21072618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Khanna S., Parinandi N.L., Kotha S.R., Roy S., Rink C., Bibus D., Sen C.K. Nanomolar vitamin E α-tocotrienol inhibits glutamate-induced activation of phospholipase A 2 and causes neuroprotection. J. Neurochem. 2010;112(5):1249–1260. doi: 10.1111/j.1471-4159.2009.06550.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Schirinzi T., Martella G., Imbriani P., Di Lazzaro G., Franco D., Colona V.L., Alwardat M., Sinibaldi S.P., Mercuri N.B., Pierantozzi M., Pisani A. Dietary vitamin E as a protective factor for Parkinson’s disease: Clinical and experimental evidence. Front. Neurol. 2019;10:148. doi: 10.3389/fneur.2019.00148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Etminan M., Gill S.S., Samii A. Intake of vitamin E, vitamin C, and carotenoids and the risk of Parkinson’s disease: A meta-analysis. Lancet Neurol. 2005;4(6):362–365. doi: 10.1016/S1474-4422(05)70097-1. [DOI] [PubMed] [Google Scholar]
  • 218.Oliver D.M.A., Reddy P.H. Small molecules as therapeutic drugs for Alzheimer’s disease. Mol. Cell. Neurosci. 2019;96:47–62. doi: 10.1016/j.mcn.2019.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Jin H., Kanthasamy A., Ghosh A., Anantharam V., Kalyanaraman B., Kanthasamy A.G. Mitochondria-targeted antioxidants for treatment of Parkinson’s disease: Preclinical and clinical outcomes. Biochim. Biophys. Acta Mol. Basis Dis. 2014;1842(8):1282–1294. doi: 10.1016/j.bbadis.2013.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Poeggeler B., Durand G., Polidori A., Pappolla M.A., Vega-Naredo I., Coto-Montes A., Böker J., Hardeland R., Pucci B. Mitochondrial medicine: Neuroprotection and life extension by the new amphiphilic nitrone LPBNAH1 acting as a highly potent antioxidant agent. J. Neurochem. 2005;95(4):962–973. doi: 10.1111/j.1471-4159.2005.03425.x. [DOI] [PubMed] [Google Scholar]
  • 221.Bachurin S., Bukatina E., Lermontova N., Tkachenko S., Afanasiev A., Grigoriev V., Grigorieva I., Ivanov Y.U., Sablin S., Zefirov N. Antihistamine agent Dimebon as a novel neuroprotector and a cognition enhancer. Ann. N. Y. Acad. Sci. 2001;939(1):425–435. doi: 10.1111/j.1749-6632.2001.tb03654.x. [DOI] [PubMed] [Google Scholar]
  • 222.Doody R.S., Gavrilova S.I., Sano M., Thomas R.G., Aisen P.S., Bachurin S.O., Seely L., Hung D. Effect of dimebon on cognition, activities of daily living, behaviour, and global function in patients with mild-to-moderate Alzheimer’s disease: A randomised, double-blind, placebo-controlled study. Lancet. 2008;372(9634):207–215. doi: 10.1016/S0140-6736(08)61074-0. [DOI] [PubMed] [Google Scholar]
  • 223.Grigor’ev V.V., Dranyi O.A., Bachurin S.O. Comparative study of action mechanisms of dimebon and memantine on AMPA- and NMDA-subtypes glutamate receptors in rat cerebral neurons. Bull. Exp. Biol. Med. 2003;136(5):474–477. doi: 10.1023/B:BEBM.0000017097.75818.14. [DOI] [PubMed] [Google Scholar]
  • 224.Tang T.S., Slow E., Lupu V., Stavrovskaya I.G., Sugimori M., Llinás R., Kristal B.S., Hayden M.R., Bezprozvanny I. Disturbed Ca2+ signaling and apoptosis of medium spiny neurons in Huntington’s disease. Proc. Natl. Acad. Sci. USA. 2005;102(7):2602–2607. doi: 10.1073/pnas.0409402102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Wu J., Li Q., Bezprozvanny I. Evaluation of Dimebon in cellular model of Huntington’s disease. Mol. Neurodegener. 2008;3(1):15. doi: 10.1186/1750-1326-3-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Lermontova N.N., Redkozubov A.E., Shevtsova E.F., Serkova T.P., Kireeva E.G., Bachurin S.O. Dimebon and tacrine inhibit neurotoxic action of beta-amyloid in culture and block L-type Ca2+ channels. Bull. Exp. Biol. Med. 2001;132(5):1079–1083. doi: 10.1023/A:1017972709652. [DOI] [PubMed] [Google Scholar]
  • 227.Bachurin S.O., Shevtsova E.P., Kireeva E.G., Oxenkrug G.F., Sablin S.O. Mitochondria as a target for neurotoxins and neuroprotective agents. Ann. N. Y. Acad. Sci. 2003;993(1):334–344. doi: 10.1111/j.1749-6632.2003.tb07541.x. [DOI] [PubMed] [Google Scholar]
  • 228.Nguyen L., Lucke-Wold B.P., Mookerjee S.A., Cavendish J.Z., Robson M.J., Scandinaro A.L., Matsumoto R.R. Role of sigma-1 receptors in neurodegenerative diseases. J. Pharmacol. Sci. 2015;127(1):17–29. doi: 10.1016/j.jphs.2014.12.005. [DOI] [PubMed] [Google Scholar]
  • 229.Prolla T.A., Mattson M.P. Molecular mechanisms of brain aging and neurodegenerative disorders: Lessons from dietary restriction. Trends Neurosci. 2001;24(11), (Suppl.):S21-S31. doi: 10.1016/S0166-2236(00)01957-3. [DOI] [PubMed] [Google Scholar]
  • 230.Colangelo A.M., Alberghina L., Papa M. Astrogliosis as a therapeutic target for neurodegenerative diseases. Neurosci. Lett. 2014;565:59–64. doi: 10.1016/j.neulet.2014.01.014. [DOI] [PubMed] [Google Scholar]
  • 231.Kim J., Min K.J., Seol W., Jou I., Joe E. Astrocytes in injury states rapidly produce anti-inflammatory factors and attenuate microglial inflammatory responses. J. Neurochem. 2010;115(5):1161–1171. doi: 10.1111/j.1471-4159.2010.07004.x. [DOI] [PubMed] [Google Scholar]
  • 232.Li J., Liu D., Sun L., Lu Y., Zhang Z. Advanced glycation end products and neurodegenerative diseases: Mechanisms and perspective. J. Neurol. Sci. 2012;317(1-2):1–5. doi: 10.1016/j.jns.2012.02.018. [DOI] [PubMed] [Google Scholar]
  • 233.Dringen R., Gutterer J.M., Hirrlinger J. Glutathione metabolism in brain. Eur. J. Biochem. 2000;267(16):4912–4916. doi: 10.1046/j.1432-1327.2000.01597.x. [DOI] [PubMed] [Google Scholar]
  • 234.Fernandez-Fernandez S., Almeida A., Bolaños J.P. Antioxidant and bioenergetic coupling between neurons and astrocytes. Biochem. J. 2012;443(1):3–11. doi: 10.1042/BJ20111943. [DOI] [PubMed] [Google Scholar]
  • 235.Shih A.Y., Johnson D.A., Wong G., Kraft A.D., Jiang L., Erb H., Johnson J.A., Murphy T.H. Coordinate regulation of glutathione biosynthesis and release by Nrf2-expressing glia potently protects neurons from oxidative stress. J. Neurosci. 2003;23(8):3394–3406. doi: 10.1523/JNEUROSCI.23-08-03394.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Williamson T.P., Johnson D.A., Johnson J.A. Activation of the Nrf2-ARE pathway by siRNA knockdown of Keap1 reduces oxidative stress and provides partial protection from MPTP-mediated neurotoxicity. Neurotoxicology. 2012;33(3):272–279. doi: 10.1016/j.neuro.2012.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Reed J., Jurgensmeier J., Matsuyama S. Bcl-2 family proteins and mitochondria. Biochim. Biophys. Acta Bioenerg. 1998;1366(1-2):127–137. doi: 10.1016/S0005-2728(98)00108-X. [DOI] [PubMed] [Google Scholar]
  • 238.Sugasawa T., Tome Y., Takeuchi Y., Yoshida Y., Yahagi N., Sharma R., Aita Y., Ueda H., Maruyama R., Takeuchi K., Morita S., Kawamai Y., Takekoshi K. Influence of intermittent cold stimulations on CREB and its targeting genes in muscle: Investigations into molecular mechanisms of local cryotherapy. Int. J. Mol. Sci. 2020;21(13):4588. doi: 10.3390/ijms21134588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Ribas V., García-Ruiz C., Fernández-Checa J.C. Glutathione and mitochondria. Front. Pharmacol. 2014;5:151. doi: 10.3389/fphar.2014.00151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Craig E.A. Hsp70 at the membrane: Driving protein translocation. BMC Biol. 2018;16(1):11. doi: 10.1186/s12915-017-0474-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Tang B.L. Sirt1 and the mitochondria. Mol. Cells. 2016;39(2):87–95. doi: 10.14348/molcells.2016.2318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Lombard D.B., Tishkoff D.X., Bao J. Mitochondrial sirtuins in the regulation of mitochondrial activity and metabolic adaptation. Handb. Exp. Pharmacol. 2011;206:163–188. doi: 10.1007/978-3-642-21631-2_8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Miriyala S., Holley A.K., St Clair D.K. Mitochondrial superoxide dismutase-signals of distinction. Anticancer. Agents Med. Chem. 2011;11(2):181–190. doi: 10.2174/187152011795255920. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Current Neuropharmacology are provided here courtesy of Bentham Science Publishers

RESOURCES