Abstract
A large body of evidence from post-mortem brain tissue and genetic analysis in man and biochemical and pathological studies in animal models (transgenic and toxin) of neurodegeneration suggest that mitochondrial dysfunction is a common pathological mechanism. Mitochondrial dysfunction due to oxidative stress, mitochondrial DNA deletions, pathological mutations, altered mitochondrial morphology and interaction of pathogenic proteins with mitochondria leads to neuronal demise. Therefore, therapeutic approaches targeting mitochondrial dysfunction and oxidative damage hold great promise in neurodegenerative diseases. This review discusses the potential therapeutic efficacy of creatine, coenzyme Q10, idebenone, synthetic triterpenoids, and mitochondrial targeted antioxidants (MitoQ) and peptides (SS-31) in in vitro studies and in animal models of Parkinson's disease (PD), Huntington's disease (HD), Amyotrophic Lateral Sclerosis (ALS) and Alzheimer's disease (AD). We have also reviewed the current status of clinical trials of creatine, coenzyme Q10, idebenone and MitoQ in neurodegenerative disorders. Further, we discuss newly identified therapeutic targets including PGC-1α and Sirtuins, which provide promise for future therapeutic developments in neurodegenerative disorders.
Keywords: Coenzyme Q10, Creatine, Triterpenoids, PGC-1α, Parkinson's disease, Huntington's disease, Alzheimer's disease
Mitochondrial dysfunction in Parkinson's disease (PD)
Much evidence suggests a role for mitochondrial dysfunction in the pathophysiology of PD, a common age-associated neurodegenerative disease characterized by progressive degeneration of the nigrostriatal dopamine (DA) pathway and presence of the intraneuronal protein inclusions (Lewy bodies)1-3. The most compelling evidence of mitochondrial dysfunction in PD comes from the accidental discovery of the synthetic meperidine analogue 1-methyl-4-phenyl-1,2,3,6-tetrahydrodropyridine (MPTP), which causes parkinsonism by selective inhibition of mitochondrial complex-I of the electron transport chain4, 5. Similar to MPTP, infusion of other selective complex-I inhibitors such as rotenone, pyridaben, fenazaquin, tebunfenpyrad, trichloroethylene and fenpyroximate results in loss of the nigral dopaminergic neurons and parkinsonian phenotypes in in vivo models, implicating mitochondrial dysfunction in PD pathogenesis6-8. An impairment of mitochondrial complex-I activity has been observed in the substantia nigra (SN), platelets, and skeletal muscle of PD patients 1, 9-13, in some but not in all studies14, 15. Similarly, reduced complex-I activity and an increased susceptibility to MPP+ were also observed in cybrids containing mitochondrial DNA from PD patients, suggesting mitochondrial DNA encoded defects in PD 16, 17, although in one study no significant reduction in complex-I activity was found18. Besides the mitochondrial complex-I defect, there is genetic evidence that suggests that mutations in mitochondrial DNA (mtDNA) also play a role in the pathogenesis of PD. We observed G11778A mtDNA point mutation in a subunit of mitochondrial complex-I in a family with parkinsonism and multisystem degeneration19. We also identified high levels of somatic mtDNA point mutations in elderly PD patients20. A mutation in mitochondrial DNA polymerase gamma (POLG1) has been observed in five Finnish families with idiopathic sporadic PD21. To date several mtDNA mutations including large scale rearrangements, point mutations, and micro deletions have been identified in the SN and other brain regions of PD patients20, 22-25. Recently, another compelling piece of evidence of mitochondrial dysfunction in PD has come from conditional knockout “MitoPark” mice, who have a disrupted mitochondrial transcription factor A (Tfam) gene in DA neurons. These mice show reduced mtDNA expression, reduced respiratory chain function in DA neurons, and a progressive PD phenotype, consistent with involvement of respiratory chain dysfunction in PD pathogenesis26. Further, there is evidence of reduced mitochondrial mass and size in the mouse SN DA neurons as compared to non-DA neurons, suggesting selective vulnerability of DA neurons may be due to mitochondrial dysfunction in PD27. In addition to mtDNA mutations, pathogenic mutations in several genes including α–synuclein, parkin, UCHL-1, DJ-1, PINK-1, LRRK-2, NURR-1, tau, and HtrA2 also directly or indirectly implicate a role of mitochondrial dysfunction in familial PD pathogenesis3, 28, 29. α–Synuclein is a major component of Lewy bodies, and gene mutations are associated with autosomal dominant familial PD28, 29. Over expression of α–synuclein in cell culture and in transgenic mice impairs mitochondrial function and increases vulnerability to MPTP30, 31. In contrast, α–synuclein-null mice are resistant to respiratory chain inhibitors such as MPTP, 3-nitropropionic acid (3-NP) and malonate, thus implicating mitochondria in α–synuclein mediated toxicity32, 33.
Mitochondrial dysfunction in Huntington's disease (HD)
HD is an autosomal-dominant disorder characterized by lesions in the striatum of the brain that cause progressive behavioral and cognitive impairments and involuntary choreiform movements. HD is caused by the abnormal expansion of a CAG repeat in exon 1 of the HD gene, resulting in elongated polyglutamine stretches in the protein product known as mutant huntingtin34. Various lines of evidence suggest involvement of mitochondrial dysfunction in HD. Evidence for bioenergetic defects in HD pathogenesis comes from the finding of pronounced weight loss in HD patients, despite sustained caloric intake. Glucose metabolism is reduced in the basal ganglia and cerebral cortex of symptomatic HD patients and even of presymptomatic gene carriers, as assessed by PET imaging35-37. Further, NMR spectroscopy revealed a decrease in N-acetylaspartate (NAA) and an increase in lactate in the basal ganglia of HD patients, and was found to directly correlate with the subject's CAG repeat number38, 39. Biochemical studies revealed reduced activity of several key components of oxidative phosphorylation and the TCA cycle, including complexes II-III and aconitase, in advanced stage HD patients, while complex-I activity was unaltered40-43. Similarly, a recent study showed increased glucose utilization relative to oxygen utilization in the striatum of early HD patients44.
Mitochondrial dysfunction in HD is not restricted only to the brain, but is also evident in the peripheral tissues including muscle and platelets39, 45-47, and in cultured cells48. In muscle of symptomatic and presymptomatic HD patients, there is a reduced ATP / phosphocreatine ratio, a decreased ratio of phosphocreatine to inorganic phosphate, and impaired complex-I activity, suggesting abnormal skeletal muscle mitochondrial energy metabolism39, 45, 46. We observed significantly reduced cellular respiration and COX activity in myoblasts from HD patients, and these defects were exacerbated in chronic energy deprivation conditions (Chaturvedi et. al., unpublished findings). In addition, lymphoblasts from HD patients and brain mitochondria from YAC transgenic mice expressing Htt with 72 repeats show mitochondrial dysfunction including a decreased membrane potential, impaired calcium ion homeostasis49, and marked morphological abnormalities50. Similarly, mouse immortalized striatal cells expressing endogenous mutant huntingtin (STHdhQ111) and the human lymphoblastoid cell lines bearing HD-causing alleles showed decreased ATP levels and ADP uptake51. Further, in striatal cells from mutant Htt knock-in mouse embryos, mitochondrial respiration and ATP production are significantly impaired48. Over expression of complex-II subunits in striatal neurons expressing Htt171-82Q restored complex-II activity and blocked mitochondrial dysfunction and cell death52.
In addition, a number of studies provide genetic evidence that mtDNA mutations may also be involved in HD pathogenesis. Recently, it was shown that HD patients have higher frequencies of mtDNA deletions in lymphocytes and leucocytes in comparison to the controls, and the number of mutations is directly related to the size of the CAG repeats expansion53, 54, This may be a consequence of oxidative damage to mtDNA. Moreover, the fact that mitochondrial toxins such as 3-NP and malonate, which selectively inhibit complex-II and induce a pathological phenotype resembling HD in human, rodents, and primates, further enforce the significance of mitochondrial dysfunction in HD pathogenesis 34, 40, 55, 56.
Mutant Htt plays an important role in mitochondrial dysfunction in HD. Several mechanisms may be involved. First, it may directly bind to the mitochondria. There is evidence from an HD mouse model, and from cultured HD striatal cells (STHdhQ111), that mutant Htt associates directly with the outer mitochondrial membrane49, 57. In addition, by electron microscopy, N-terminal mutant Htt was found to localize on neuronal mitochondrial membranes49. Second, mutant Htt protein directly increases susceptibility of mitochondria to the calcium-induced permeability transition and cytochrome c release49, 58. Similarly, mutant Htt expressing STHdhQ111 cells show a reduced mitochondrial Ca2+ uptake capacity, Ca2+-induced decrease in mitochondrial respiration and increased mitochondrial membrane permeability in comparison with wild type cells59. Mitochondrial defects seen in these studies are a direct effect of the mutant Htt, as incubation of mitochondria from normal lymphoblasts with mutant Htt (fusion protein composed of a peptide containing a pathogenic polyglutamine tract) reproduced mitochondrial abnormalities seen in the HD patients and knock-in HD mice49. Third, besides a direct effect on the mitochondria, mutant Htt also impairs in vitro and in vivo trafficking of mitochondria in neurons, leading to loss of mitochondrial motility and eventually mitochondrial dysfunction60, 61. These findings clearly indicate that mutant Htt induces mitochondrial Ca2+ handling defects, respiratory deficits, and impaired mitochondrial movement which could be contributing mechanisms to mitochondrial dysfunction in HD.
Mutant Htt also impairs mitochondrial function by altering transcription. Htt directly binds to and down regulates the activity of several transcription factors including CREB-binding protein, TAFII130 and SP162-68. Binding of Htt on these transcription factors leads to alteration of expression of several genes involved in mitochondrial respiration and normal mitochondrial function. Htt binding on p53 causes upregulation of the downstream target genes BAX and PUMA, which leads to increased mitochondrial membrane depolarization69. Recently, Peroxisome proliferator-activated receptor-γ-coactivator (PGC-1α), which is a coactivator of several transcription factors and is a key regulator of mitochondrial biogenesis was found to be impaired in HD patients and transgenic HD models70-72. Mutant Htt protein directly impairs the ability of PGC-1α to switch on downstream target genes such as the gene encoding UCP-1. Moreover, mutant Htt also binds with the CREB/TAF4 complex on the PGC-1α promoter and impairs the PGC-1α activity by blocking the transcription of its target genes71. Impairment of the PGC-1α function and down regulation of its mitochondrial target genes leads to abnormalities in mitochondrial function and energy metabolism, contributing to neurodegeneration in susceptible neurons70-72. Similarly, impaired PGC-1α transcription and activity impact the enzyme system that combats reactive oxygen species (ROS). This leads to down regulation of ROS defense genes encoding SOD1, SOD2, and glutathione peroxidase, resulting in increased oxidative damage and neuronal death73.
Mitochondrial dysfunction in Alzheimer's disease (AD)
AD is a late-onset, progressive, age-dependent neurodegenerative disorder, characterized by the progressive cognitive decline, and the presence of amyloid-β senile plaques and tau neurofibrillary tangles. Mitochondrial dysfunction is widely implicated in the pathogenesis of AD. Evidence for mitochondrial dysfunction in the AD pathogenesis comes from impaired activities of the three key TCA enzyme complexes, pyruvate dehydrogenase, isocitrate dehydrogenase, and α-ketoglutarate dehydrogenase, observed in postmortem AD brain and fibroblasts from AD patients74,75. Similarly, reduced respiratory chain complex I, III, and IV activity was found in platelets and lymphocytes from AD patients and postmortem brain tissue76-79. In addition to a direct mitochondrial respiratory chain defect, more recently, increased autophagic degradation of mitochondria has also been observed in AD80.
There is a growing body of evidence that mtDNA mutations also play a pivotal role in mitochondrial dysfunction in AD pathogenesis. In a recent study, 20 point mutations were detected in the mitochondrial-encoded cytochrome c oxidase subunits I, II, and III genes in AD patients81. Other studies showed two missense mutations in the mitochondrial DNA of cytochrome c oxidase and high aggregate burden of somatic mtDNA point mutations in the patients with AD82, 83, however direct sequencing of the complete mtDNA coding region has not identified disease specific mutations84. Moreover, there is also evidence that some of the proteins implicated in AD pathogenesis directly target mitochondria and cause mitochondrial dysfunction. These include, the amyloid precursor protein (APP) which binds directly to the mitochondria in cortical neuronal cells from AD transgenic mouse85. Similarly, amyloid-βfrom mutant APP transgenic mice also binds to mitochondria and causes mitochondrial dysfunction86.
Mitochondrial dysfunction in Amyotrophic lateral sclerosis (ALS)
ALS is a neurodegenerative disease, characterized by a selective loss of motor neurons in the spinal cord brainstem, and motor cortex, which leads to skeletal muscle atrophy, paralysis and death3. The etiology of ALS is unknown. Some 90% of cases are sporadic with unknown cause (SALS) and 10% are familial (FALS). Among familial cases, 20% are linked to mutations in Cu/Zn- superoxide dismutase (SOD). Several different pathogenic mechanisms have been identified in the CNS and peripheral tissues during the disease course in ALS, but mitochondrial and bioenergetic defects are implicated widely in ALS pathogenesis2. There is evidence of abnormal structure and number of mitochondria and compromised function in ALS motor neurons and skeletal muscle. That is corroborated by findings of altered respiratory chain enzyme activities and CNS energy hypometabolism in ALS spinal cord and motor cortex87-91. Morphological changes in mitochondria are the earliest detectable pathologic events followed by decreased mitochondrial respiration in the SOD1 over expressing G93A transgenic mouse 89, 92. Decreased ATP levels and impaired respiratory chain enzyme activities were reported in motor neuron cell lines expressing mutant SOD1, suggesting involvement of mutant SOD1 in mitochondrial defects93, 94. We showed significantly decreased mitochondrial Ca2+ loading capacity, oxygen consumption, respiratory chain complex activities and ATP synthesis in the brain and spinal cord mitochondria from mutant SOD1 transgenic mice confirming that mutant SOD1 is associated with impaired mitochondrial dysfunction95, 96. In addition, mitochondrial dysfunction could be due to the direct interaction of mutant SOD1 with mitochondria, as mutant SOD1 localizes to the mitochondrial intermembrane space and matrix and affects mitochondrial protein import97. It also selectively associates with the outer mitochondrial membrane in spinal cord motor neurons97-99. Moreover, recently it was shown that mutant SOD1 impairs fast axonal mitochondrial transport in the anterograde direction in motor neurons derived from SOD1 G93A transgenic mice100.
Mitochondrial approaches for neuroprotection
There is ample evidence showing involvement of mitochondrial dysfunction in the pathogenesis of neurodegenerative disorders, therefore, one would predict that agents that alleviate mitochondrial dysfunction could be beneficial and exert neuroprotective effects. Several bioenergetic agents that improve mitochondrial function including creatine, coenzyme Q10 (CoQ10), nicotinamide, riboflavin and lipoic acid are being tested for their neuroprotective efficacy in neurodegenerative disorders. Among them, creatine and CoQ10 are in clinical trials for PD, HD and AD.
Creatine
Creatine is a nitrogenous guanidine compound that occurs naturally in vertebrates and helps to supply energy to muscle and nerve cells. It is taken up into the brain and skeletal muscle by a sodium-dependent creatine transporter, and becomes physiologically active when it is transformed enzymatically into phosphocreatine (PCr). The creatine/phosphocreatine system, regulated by the mitochondrial creatine kinase (CK), plays an important role in maintaining energy balance in the brain. The presence of this energy buffer system keeps the ATP/ADP ratio high at sub-cellular sites where ATP is needed, which ensures that the free energy of ATP remains high. That also minimizes the loss of adenosine nucleotides, which causes cellular dysfunction.
Creatine supplementation was found to be neuroprotective in a series of in vitro and in vivo studies101. Recently, creatine was shown to enhance the survival of glutamate-treated neuronal/glial cells by modulation of Ras/NF-kappaB signaling102. Creatine treatment also promotes differentiation of cultured GABA-ir neurons and provides significant neuroprotection against glucose, serum deprivation and 3-NP induced toxicity103, 104. Chronic administration of creatine was shown to significantly increase survival, tyrosine hydroxylase immunoreactive (TH-IR) fibers density, and soma size of dopaminergic neurons in mesencephalic culture, by protecting against neurotoxic insults induced by serum and glucose deprivation, MPP+, and 6-hydroxydopamine (6-OHDA)105, 106. In aged mice, creatine improves health and survival107. We examined the effects of creatine supplementation on MPTP-induced parkinsonism108. We found that creatine produced protection against dopamine loss, as well as an attenuation of neuron loss in the SN of mice treated with MPTP. Further, oral creatine supplementation produced dose-dependent significant protection against MPTP-induced dopamine depletion. Moreover, creatine protected against MPTP-induced loss of TH-IR neurons in the SN. We showed that prophylactic administration of oral creatine reduces the size of ischemic brain infarctions in mice109. In another study we showed that oral administration of 1% creatine in the diet significantly attenuated striatal excitotoxic lesions produced by NMDA110. Creatine and nicotinamide in combination exerted significant additive neuroprotective effects against malonate-induced striatal lesions110. We also found additive neuroprotective effects of creatine and a cyclooxygenase-2 inhibitor in the MPTP mouse model of PD and the G93A transgenic mouse model of ALS111, 112. In rats, creatine prevented excitotoxic death of intrinsic neurons caused by intrastriatal infusion of the neurotoxin ibotenic acid113. Oral administration of creatine produces a dose-dependent improvement in motor performance and extends survival in G93A transgenic ALS mice, where it protects against loss of both motor neurons and SN neurons114. However, creatine does not exert a beneficial effect on skeletal muscle function in G93A ALS mice115. Creatine significantly attenuates elevated glutamate concentrations in cerebral cortex of G93A ALS mice116. The combination of creatine and minocycline also provides additive neuroprotective effects in the G93A mouse model of ALS117. We found that creatine significantly improves survival, improves motor performance, slows the development of brain atrophy, increases brain ATP levels, delays atrophy of striatal neurons and the formation of Htt-positive aggregates in the R6/2 and N-171-82Q transgenic mouse models of HD118-120. Creatine administration protects against glutamate and β-amyloid toxicity in rat hippocampal neurons121. Creatine is also beneficial in the animal models of traumatic brain injury, spinal cord injury and cerebral ischemia122-126. In addition, preincubation of anoxic rat hippocampal slices with creatine attenuated the decrease in PCr and ATP content127.
The success of creatine in experimental studies led to clinical trials in neurodegenerative diseases including PD, HD and ALS. We found beneficial effects using combination therapy of creatine monohydrate, CoQ10, and lipoic acid in a randomized, double-blind, placebo-controlled, crossover study in patients with mitochondrial cytopathies128. In a small pilot trial, creatine improved patient mood but had no effect on overall Unified Parkinson's Disease Rating Scale (UPDRS)129. In order to see the protective effect of creatine (10g/day) and minocycline on PD progression, a randomized, double-blind, Phase II futility clinical trial was carried out by NINDS NET-PD investigators in early PD130. Creatine was shown to reduce the UPDRS scores, and could not be rejected as futile based on the 30% DATATOP futility threshold. Moreover in this study, tolerability was 91% for creatine and 77% for minocycline. As compared to DATATOP controls, disease progression on the UPDRS was slowed by almost 50% at 1 year in the creatine treated patients. In one clinical trial creatine supplementation at 20 g/day with resistance training in PD patients resulted in increased muscle endurance and upper body strength131. Recently, the NIH announced a phase III clinical trial of creatine for PD, with a goal of recruiting 1720 participants randomized to 10g of creatine or placebo132, 133. To determine the safety and tolerability of creatine in HD we carried out a randomized, double-blind, placebo-controlled study in 64 subjects134. We found that oral administration of creatine at a dose of 8 g/day for 16 weeks is well tolerated, safe and bioavailable to the brain, and it decreased serum 8-hydroxy-2-deoxyguanosine levels, a marker for oxidative stress. Taken together, these findings clearly indicate that creatine is a promising agent for use as a neuroprotectant in a variety of neurodegenerative disorders.
CoQ10
CoQ10 is an essential biological cofactor of the electron transport chain, where it accepts electrons from complexes I and II. CoQ10, also known as ubiquinone, serves as an important antioxidant in mitochondrial and lipid membranes135. It is a particularly important antioxidant in the inner mitochondrial membrane, where it can directly scavenge free radicals through interactions with α-tocopherol135. Besides its free radical scavenging activity, it also prevents apoptotic cell death by blocking Bax binding to mitochondria, and by inhibiting activation of the mitochondrial permeability transition (MPT)136, 137. CoQ10 is a co-factor of mitochondrial uncoupling proteins (UCP), and may also exert neuroprotective effects by activation of these proteins, leading to a reduction in mitochondrial-free radical generation101, 138. CoQ10 increases mitochondrial UCP expression in the SN of primates, and this is associated with marked neuroprotection against the MPTP toxicity101, 139. Further, increased expression of the mitochondrial UCP also protects against neuronal damage in experimental models of stroke and epilepsy101.
CoQ10 exerts neuroprotective effects in several in vivo and in vitro models of neurodegenerative disorders. It protects against paraquat and rotenone induced mitochondrial dysfunction and neuronal cell death in human neuroblastoma (SHSY-5Y) cells and primary rat mesencephalic neurons respectively140, 141. It provides neuroprotection in iron-induced apoptosis in dopaminergic neurons, and exerts anti-amyloidogenic effects by destabilizing preformed beta-amyloid fibrils in vitro142, 143. Further, CoQ10 protects SHSY5Y neuronal cells from β-amyloid toxicity through inhibition of the MPT pore144. Similarly, pretreatment of neuronal cells with CoQ10 maintains mitochondrial membrane potential during oxidative stress and reduces the amount of mitochondrial ROS generation145.
The protective role of CoQ10 was also studied in several toxin models. We demonstrated CoQ10 provided significant protection against MPTP induced dopamine depletion and loss of TH-IR neurons in aged mice146. More recently, we found that dietary administration of either of two formulations of CoQ10 resulted in significant protection in a chronic MPTP model induced by the administration of MPTP by Alzet pump for 1 month147. We found neuroprotective effects against DA depletion, loss of TH-IR neurons in the SN and the development of α–synuclein aggregates. The finding that CoQ10 is effective in a chronic model of MPTP toxicity is of particular interest, as this may be more relevant to PD, which is a progressive neurodegenerative disorder. Oral administration of CoQ10 exerts significant neuroprotective effects against striatal lesions in rats produced by aminoxyacetic acid, and the mitochondrial toxins malonate and 3-NP148-150. Oral administration of CoQ10 produced dose-dependent neuroprotective effects against malonate induced striatal lesions, depletion of ATP and increases in lactate concentration148. Similarly, oral administration of CoQ10 for 1 week, prior to administration of 3-NP resulted in a significant 90% neuroprotection against 3-NP induced striatal lesions150. We also demonstrated that CoQ10 produces neuroprotective effects in transgenic mouse models of ALS and HD150, 151. Recently, we found that CoQ10 treatment decreases brain oxidative stress, Aβ42 levels, β-amyloid plaque area and number, and improves cognitive performance in Tg19959 mice, a transgenic mouse model of AD (Kipiani et al., unpublished findings). A dose ranging and efficacy study of highdose coenzyme Q10 formulations in the R6/2 HD transgenic mice demonstrated that high-dose CoQ10 significantly extends survival, improves motor performance, grip strength and reduces brain atrophy in R6/2 HD mice, in a dose-dependent manner152.
As in cancer chemotherapy, combinations of two or more compounds may be a better approach for neuroprotection. The combination therapy of CoQ10 and minocycline in R6/2 transgenic mouse model of HD resulted in a significantly increased amelioration of behavioral and neuropathological deficits, extended survival, improved rotarod performance and attenuated striatal neuron atrophy, as compared to either minocycline or CoQ10 alone153. Similarly, we and others, by using CoQ10 and remacemide (NMDA antagonist) combination therapy observed significantly improved motor performance and increased survival in the R6/2 and the N-171-82Q transgenic mouse models of HD151, 154. Recently, we found that a combination of creatine and CoQ10 exerts synergistic neuroprotective effects in the MPTP model of PD (Yang et al., unpublished findings).
On the basis of results from animal studies, we and others initiated several clinical trials of CoQ10 in neurodegenerative disorders. We initially tested the effect of oral administration of 360mg/day CoQ10 on elevated cortical lactate concentrations in 18 patients with HD. We observed that CoQ10 treatment produced a 37% reduction in occipital cortex lactate concentrations, which was reversed following withdrawal of therapy39. Since CoQ10 and remacemide in combination was protective in a HD transgenic mouse models151, 154, the Huntington's Study Group performed the CARE-HD trial using CoQ10 and remacemide in 340 patients155. Patients were treated with CoQ10 (600mg /day) and remacemide (600mg /day), or a combination of the two for 30 months. Administration of CoQ10 resulted in a 14% decrease in disease progression. Remacemide demonstrated no efficacy. This trial was powered to be able to determine a 40% attenuation of total functional capacity decline with 80% power.
The ratio of reduced to oxidized CoQ10 is significantly reduced in the platelets and cerebrospinal fluid (CSF) of PD patients156, 157 and in plasma of ALS patients158. We found a significant reduction in CoQ10 levels in mitochondria isolated from platelets of PD patients, that directly correlated with a decrease in complex-I activity159. In this study, oral administration of CoQ10 in PD patients was well tolerated and resulted in significant, dose-dependent increases in plasma CoQ10 levels. The reduced level of CoQ10 in the platelets and CSF suggested that CoQ10 supplementation might be efficacious in the treatment of PD. Therefore, we carried out a multicenter, randomized, parallel-group, placebo-controlled, double-blind and dosage-ranging phase II clinical trial of CoQ10 in early PD patients (Parkinson Study Group)160. Patients were treated with placebo or 300, 600, or 1,200 mg/day of CoQ10 for 16 months to see the safety, tolerability and neuroprotective efficacy of CoQ10. The primary outcome measure was the change in the UPDRS between baseline and final visits. CoQ10 was safe and well tolerated at dosages of up to 1200 mg/day and a statistically significant dose-dependent reduction in UPDRS score was evident in subjects assigned to CoQ10 compared to those assigned to placebo. Subsequently, in other open label dose-escalation clinical trials, CoQ10 dosages as high as 3,600 mg/day were safe and well tolerated in PD, HD and ALS. However, plasma CoQ10 levels reached a plateau at the 2400 mg/day dosage and did not increase further at the 3000 mg/day dosage in PD, HD or ALS patients161, 162. A recent study showed that CoQ10 does not exert symptomatic effects163. Therefore, any benefits in a phase III study of 1200 mg and 2400 mg daily (QE3 study) are likely to be neuroprotective rather than symptomatic. An NINDS sponsored clinical trial of 600 patients randomized to placebo, 1200 mg CoQ10 or 2400 mg CoQ10 has recently started. Similarly a phase III trial of 2400 mg of CoQ10 daily has recently started in HD. This dose was chosen based on the studies of plasma CoQ10 concentrations discussed above. A phase II trial of CoQ10 in presymptomatic gene positive HD patients (PREQUEL) will begin soon.
Idebenone, a synthetic analogue of CoQ10 has been studied in several neurodegenerative disorders for neuroprotection. Idebenone protects neuronal cells against β-amyloid induced toxicity164. Similarly, when used in combination with α-tocopherol, it alleviates β-amyloid induced learning and memory deficits in rats165. A multicentre, placebo controlled, double-blind trial of idebenone 45 mg twice/day orally, in patients of AD, showed statistically significant improvement of the memory, attention and behavior166. In another randomized, double-blind, placebo-controlled multicentre study 300 patients with Alzheimer type dementia were treated for 2 years, with 30 or 90 mg /day idebenone or placebo167. The outcome of study was a statistically significant improvement in the Alzheimer's Disease Assessment Scale (ADAS) score after 6 months, in a dose dependent manner. Subsequently, in two other clinical trials, idebenone was safe and tolerable up to 360 mg/day and slowed progression of cognitive deficits in patients with AD168, 169. These results are controversial since another large trial showed no benefits170. There was no significant improvement in a small trial in HD171. Initial results of idebenone in Friedrich's Ataxia are very promising172. These findings strongly support the notion that creatine, CoQ10 and idebenone are promising neuroprotective agents in debilitating neurodegenerative disorders.
Mitochondrial targeted antioxidants/peptides
Mitochondrial-targeted antioxidants including mitochondrial CoQ10 (MitoQ) and Szeto-Schiller peptide (SS-31) play an important role in modulating ROS induced mitochondrial permeability transition and cell death, and were found to be protective in several in vitro and in vivo models of ischemia, reperfusion injury and neurodegeneration173. MitoQ is a derivative of CoQ10 conjugated to triphenylalkylphosphonium ions, which promotes uptake into mitochondria, where it can combat ROS originating in mitochondria174. MitoQ reduces ROS formation and preserves mitochondrial function after glutathione depletion, even in the cells lacking mitochondrial DNA175. Murphy and colleagues have developed MitoQ, which is currently in a phase II clinical trial for PD in New Zealand (http://www.parkinsons.org.nz/news/protectstudy.asp)174.
Similarly, a novel peptide antioxidant (SS-31) targeted to the inner mitochondrial membrane prevents apoptosis, necrosis, oxidative stress and inhibition of the mitochondrial electron transport chain176. SS-31 protects neuronal cells against tert-butyl-hydroperoxide (tBHP) induced mitochondrial depolarization and apoptotic cell death by reducing intracellular ROS, decreasing markers of apoptotic cell death and caspase activity177. It decreases mitochondrial ROS production and inhibits the MPT and mitochondrial swelling. SS-31 also prevents Ca2+ induced cytochrome c release and inhibits 3-NP-induced activation of the MPT pore and mitochondrial depolarization in isolated mitochondria178. It is also effective against myocardial ischemia-reperfusion injury in both ex vivo and in vivo models176, 179. Recently, we investigated the therapeutic effects of SS-31 in neuronal cells, stably transfected with either wildtype or mutant Cu/Zn superoxide dismutase (SOD1), and in the G93A mouse model of ALS180. We found SS-31 mediated significant neuroprotection against cell death induced by hydrogen peroxide in SOD1 neuronal cells. Further, daily intraperitoneal injections of SS-31 (5 mg/kg), starting at 30 days of age, led to a significant improvement in survival and motor performance in the G93A transgenic mouse model of ALS. SS-31 treated G93A ALS mice showed decreased cell loss and a decrease in immunostaining for markers of oxidative stress in the lumbar spinal cord, as compared with the vehicle-treated mice. Both SS-31 and SS-20 also produce complete protection against MPTP neurotoxicity (Yang et al., unpublished findings). These studies suggest that mitochondrial drug-targeting strategies are effective in several in vitro and in vivo models of neurodegenerative disorders. In the future selective manipulation of mitochondrial function may allow effective treatment of debilitating neurodegenerative disorders.
Triterpenoids
Synthetic triterpenoids (TP), analogues of oleanolic acid, are powerful inhibitors of oxidative stress and cellular inflammatory processes, and thus may be neuroprotective in neurodegeneration. Synthetic TP compounds are potent inducers of the antioxidant response element (ARE)–Nrf2–Keap1 signaling pathway and are useful in a variety of the cancers including leukemia, multiple myeloma, osteosarcoma, breast and lung cancer181. Following activation by TP, Nrf2 dissociates from Keap1, translocates to the nucleus, and binds to the ARE promoter sequences, leading to coordinated induction of a battery of cytoprotective genes including antioxidant and anti-inflammatory genes182.
Nrf-2 over expression is protective in a rat model of cerebral ischemia183. Neuronal cultures derived from the Nrf2 knockout mice show increased susceptibility to oxidative stress, as well as damage produced by mitochondrial electron transport chain complex inhibitors such as MPP+ and rotenone184. In the Nrf2 knockout (Nrf2−/−) mice, injection of the mitochondrial complex-II inhibitor 3-NP causes increased motor deficits and striatal lesions as compared to the Nrf2+/+ mice, and these toxic effects of 3-NP were protected by adenoviral mediated over expression of Nrf2185. Additionally, the Nrf2 activator tertbutylhydroquinone, has attenuated 3-NP toxicity in Nrf2+/+ mice, but not in Nrf2−/− mice185. Further, Nrf2-/- mice also show profound susceptibility to striatal lesions produced by the mitochondrial complex-II inhibitor malonate186. Conversely, the implantation of intrastriatal grafts of astrocytes over expressing Nrf2 produces neuroprotection against the neurotoxic effects of malonate. Recently, we tested neuroprotective effects of the synthetic triterpenoid CDDO-methyl amide (2-cyano-N-methyl-3,12-dioxooleana-1,9(11)-dien-28 amide; CDDO-MA), a potent activator of the Nrf2/ARE signaling, in the 3-NP rat model and MPTP mouse model (Yang et al., unpublished findings). We found that CDDO-MA exerts significant neuroprotective effects against MPTP and 3-NP induced toxicity. It reduces t-butylhydroperoxide induced ROS generation, attenuates MPTP-induced nigrostriatal dopaminergic neurodegeneration and reduces 3-NP induced striatal lesions. In another study we found that CDDO-MA also increases the expression of genes involved in mitochondrial biogenesis including PGC1-α, in the spinal cord of G93A ALS transgenic mice, (Kiaei et al., unpublished findings). These studies suggest a potential neuroprotective effect of triterpenoids in cell and animal models of neurodegenerative disorders, and these compounds hold great promise as therapeutic agents for these disorders.
Nicotinamide, Lipoic acid, Carnitine, β-hydroxybutyrate and estradiol
Nicotinamide is a precursor of nicotinamide adenine dinucleotide (NADH), which is a substrate for complex-I of the electron transport chain as well as other biochemical pathways. Nicotinamide prevents MPTP induced neurodegeneration in mice139. Lipoic acid is a disulfide compound that is found naturally in mitochondria as a coenzyme for pyruvate dehydrogenase and α-ketoglutarate dehydrogenase, and also has antioxidant effects128, 187, 188. We found that α-lipoic acid exerts significant neuroprotective effects in transgenic mouse models of HD and ALS128, 187, 188. Carnitine and β-hydroxybutyrate has shown to protect against MPTP toxicity189-191. Similarly, estradiol has shown neuroprotective efficacy in animal models of stroke, Alzheimer's disease (AD), and Parkinson's disease (PD)192.
PGC-1α and Sirtuins (Sir2)
Recently, PGC-1α, a transcriptional regulator, which is involved in mitochondrial biogenesis and respiration, has emerged as a new therapeutic target for neurodegenerative disorders72. PGC-1α regulates diverse metabolic functions in different tissues by activating a cascade of thousands of genes including the nuclear respiratory factors-1 and 2 (NRF-1 and NRF-2), mitochondrial transcription factor (Tfam) and estrogen related receptor-α (ERR-α)72. Recently, a role of impaired function of PGC-1α in HD pathogenesis was suggested and decreased PGC-1α expression was observed in HD transgenic mice and HD patients70-72. Similarly, we observed a reduction of PGC-1α and expression of its target genes in the muscle of HD transgenic mice and HD patients (Chaturvedi et al., unpublished findings). Further in another study it was demonstrated that PGC-1α reduces oxidative stress by inducing the expression of ROS scavenging enzymes73. Dopaminergic neurons in PGC-1α null mice are much more susceptible to MPTP toxicity73. Conversely, PGC-1α over expression protects neural cells from oxidative stress induced by mitochondrial toxins71. These studies provide compelling evidence for a role of PGC-1α in the pathogenesis of neurodegenerative diseases, and thus it is a promising therapeutic target for neurodegenerative disorders2, 72.
Sirtuins (silent information regulators) are member of the NAD+ dependent histone deacetylase family of proteins in yeast, and its homologs in mice and humans, that participate in a variety of cellular processes including mitochondrial functions, cellular metabolism, energy metabolism, gluconeogenesis, cell survival and aging193. Sirtuin activation improves mitochondrial function, extends lifespan, and promotes longevity in various species including yeast193. Activation of sirtuins may, therefore, potentially delay the onset of age-related neurodegenerative disorders. The mammalian Sirtuin gene family has seven homologues (SIRT 1-7). SIRT-1 activation by resveratrol increases survival of motor neurons from transgenic ALS mice and reduces learning impairment and neurodegeneration in AD mouse models by decreasing the acetylation of SIRT1 substrates PGC-1α and p53194. In addition, direct injection of SIRT1 lentivirus in the hippocampus of AD transgenic mice shows significant neuroprotection194.
Recently, resveratrol, which is very potent activator of SIRT1, was shown to improve the survival of mice placed on a high caloric diet195. There was increased insulin sensitivity, reduced insulin-like growth factor levels, increased AMP kinase activity, increased PGC-1α activity, increased mitochondrial number and improved motor function. Similarly, resveratrol in another study improved motor function in mice which was accompanied by a decrease in PGC1α acetylation leading to increased activity and mitochondrial biogenesis196. This was attributed to increased SIRT1 activity. Increased intracellular NAD+ concentration activates SIRT1 in brain following caloric restriction and this results in a reduction in amyloid pathology in a mouse model of Alzheimer disease197. Increased SIRT1 protected against hippocampal degeneration in another mouse model of Alzheimer disease194. Resveratrol also protects against 3-NP induced motor and behavioral deficits198. In a recent study it was found that over expression of SIRT1 deacetylase and SIRT1 activation by resveratrol significantly protects against microglia-dependent amyloid-β toxicity by inhibiting NF-kB signaling199. Altogether, theses studies provide strong evidence for involvement of Sirtuins in neuroprotection and targeting of Sirtuins could be an attractive therapeutic approach in neurodegenerative disorders.
Conclusion and future perspective
Analysis of postmortem brain tissue from patients as well as experimental studies in animal models indicate that mitochondrial dysfunction and oxidative damage play a central role in the pathogenesis of a number of common neurodegenerative diseases including PD, AD, HD and ALS. Therefore, therapeutic agents having the ability to target mitochondria and reduce mitochondrial dysfunction hold great promise for therapeutic interventions. Several agents, including creatine, CoQ10, MitoQ and idebenone are presently in clinical trials and show promising neuroprotective effects in these disorders. In the future, larger clinical trials with larger numbers of patients will provide more definitive information on the therapeutic efficacy of these compounds. Phase III clinical trials for PD and HD have commenced this year. Mitochondrial targeted antioxidants (MitoQ) and peptides (SS-31) are also of great interest, as they combat against ROS at their site of origin in mitochondria. In addition, combination therapy with two or more compounds may be more efficacious than a single compound for neuroprotection. Identification of new therapeutic targets including PGC-1α and Sirtuins may provide new avenues for therapy of neurodegenerative diseases.
Acknowledgements
This work is supported by NINDS, NIA and the Department of Defense.
Footnotes
Conflict of interest: None declared.
References
- 1.Beal MF. Mitochondria take center stage in aging and neurodegeneration. Ann Neurol. 2005;58:495–505. doi: 10.1002/ana.20624. [DOI] [PubMed] [Google Scholar]
- 2.Lin MT, Beal MF. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. 2006;443:787–95. doi: 10.1038/nature05292. [DOI] [PubMed] [Google Scholar]
- 3.Kwong JQ, Beal MF, Manfredi G. The role of mitochondria in inherited neurodegenerative diseases. J Neurochem. 2006;97:1659–75. doi: 10.1111/j.1471-4159.2006.03990.x. [DOI] [PubMed] [Google Scholar]
- 4.Langston JW, et al. Chronic Parkinsonism in humans due to a product of meperidineanalog synthesis. Science. 1983;219:979–80. doi: 10.1126/science.6823561. [DOI] [PubMed] [Google Scholar]
- 5.Burns RS, et al. The clinical syndrome of striatal dopamine deficiency. Parkinsonism induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) N Engl J Med. 1985;312:1418–21. doi: 10.1056/NEJM198505303122203. [DOI] [PubMed] [Google Scholar]
- 6.Betarbet R, et al. Chronic systemic pesticide exposure reproduces features of Parkinson's disease. Nat Neurosci. 2000;3:1301–6. doi: 10.1038/81834. [DOI] [PubMed] [Google Scholar]
- 7.Sherer TB, et al. Mechanism of toxicity of pesticides acting at complex I: relevance to environmental etiologies of Parkinson's disease. J Neurochem. 2007;100:1469–79. doi: 10.1111/j.1471-4159.2006.04333.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Gash DM, et al. Trichloroethylene: Parkinsonism and complex 1 mitochondrial neurotoxicity. Ann Neurol. 2007. [DOI] [PubMed]
- 9.Haas RH, et al. Low platelet mitochondrial complex I and complex II/III activity in early untreated Parkinson's disease. Ann Neurol. 2007;37:714–22. doi: 10.1002/ana.410370604. [DOI] [PubMed] [Google Scholar]
- 10.Schapira AH. Mitochondrial dysfunction in Parkinson's disease. Cell Death Differ. 2007;14:1261–6. doi: 10.1038/sj.cdd.4402160. [DOI] [PubMed] [Google Scholar]
- 11.Schapira AH. Mitochondria in the aetiology and pathogenesis of Parkinson's disease. Lancet Neurol. 2008;7:97–109. doi: 10.1016/S1474-4422(07)70327-7. [DOI] [PubMed] [Google Scholar]
- 12.Winkler-Stuck K, et al. Re-evaluation of the dysfunction of mitochondrial respiratory chain in skeletal muscle of patients with Parkinson's disease. J Neural Transm. 2005;112:499–518. doi: 10.1007/s00702-004-0195-y. [DOI] [PubMed] [Google Scholar]
- 13.Benecke R, Strumper P, Weiss H. Electron transfer complexes I and IV of platelets are abnormal in Parkinson's disease but normal in Parkinson-plus syndromes. Brain. 1993;116(Pt 6):1451–63. doi: 10.1093/brain/116.6.1451. [DOI] [PubMed] [Google Scholar]
- 14.Mann VM, et al. Brain, skeletal muscle and platelet homogenate mitochondrial function in Parkinson's disease. Brain. 1992;115(Pt 2):333–42. doi: 10.1093/brain/115.2.333. [DOI] [PubMed] [Google Scholar]
- 15.Blandini F, Nappi G, Greenamyre JT. Quantitative study of mitochondrial complex I in platelets of parkinsonian patients. Mov Disord. 1998;13:11–5. doi: 10.1002/mds.870130106. [DOI] [PubMed] [Google Scholar]
- 16.Gu M, et al. Mitochondrial DNA transmission of the mitochondrial defect in Parkinson's disease. Ann Neurol. 1998;44:177–86. doi: 10.1002/ana.410440207. [DOI] [PubMed] [Google Scholar]
- 17.Swerdlow RH, et al. Origin and functional consequences of the complex I defect in Parkinson's disease. Ann Neurol. 1996;40:663–71. doi: 10.1002/ana.410400417. [DOI] [PubMed] [Google Scholar]
- 18.Aomi Y, et al. Cytoplasmic transfer of platelet mtDNA from elderly patients with Parkinson's disease to mtDNA-less HeLa cells restores complete mitochondrial respiratory function. Biochem Biophys Res Commun. 2001;280:265–73. doi: 10.1006/bbrc.2000.4113. [DOI] [PubMed] [Google Scholar]
- 19.Simon DK, et al. Familial multisystem degeneration with parkinsonism associated with the 11778 mitochondrial DNA mutation. Neurology. 1999;53:1787–93. doi: 10.1212/wnl.53.8.1787. [DOI] [PubMed] [Google Scholar]
- 20.Simon DK, et al. Somatic mitochondrial DNA mutations in cortex and substantia nigra in aging and Parkinson's disease. Neurobiol Aging. 2004;25:71–81. doi: 10.1016/s0197-4580(03)00037-x. [DOI] [PubMed] [Google Scholar]
- 21.Luoma PT, et al. Mitochondrial DNA polymerase gamma variants in idiopathic sporadic Parkinson disease. Neurology. 2007;69:1152–9. doi: 10.1212/01.wnl.0000276955.23735.eb. [DOI] [PubMed] [Google Scholar]
- 22.Bender A, et al. High levels of mitochondrial DNA deletions in substantia nigra neurons in aging and Parkinson disease. Nat Genet. 2006;38:515–7. doi: 10.1038/ng1769. [DOI] [PubMed] [Google Scholar]
- 23.Siciliano G, et al. Mitochondrial DNA rearrangements in young onset parkinsonism: two case reports. J Neurol Neurosurg Psychiatry. 2001;71:685–7. doi: 10.1136/jnnp.71.5.685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Thyagarajan D, et al. A novel mitochondrial 12SrRNA point mutation in parkinsonism, deafness, and neuropathy. Ann Neurol. 2000;48:730–6. [PubMed] [Google Scholar]
- 25.Kraytsberg Y, et al. Mitochondrial DNA deletions are abundant and cause functional impairment in aged human substantia nigra neurons. Nat Genet. 2006;38:518–20. doi: 10.1038/ng1778. [DOI] [PubMed] [Google Scholar]
- 26.Ekstrand MI, et al. Progressive parkinsonism in mice with respiratory-chain-deficient dopamine neurons. Proc Natl Acad Sci U S A. 2007;104:1325–30. doi: 10.1073/pnas.0605208103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Liang CL, et al. Mitochondria mass is low in mouse substantia nigra dopamine neurons: implications for Parkinson's disease. Exp Neurol. 2007;203:370–80. doi: 10.1016/j.expneurol.2006.08.015. [DOI] [PubMed] [Google Scholar]
- 28.Thomas B, Beal MF. Parkinson's disease. Hum Mol Genet. 2007;16:183–94. doi: 10.1093/hmg/ddm159. Spec No. 2. [DOI] [PubMed] [Google Scholar]
- 29.Dauer W, Przedborski S. Parkinson's disease: mechanisms and models. Neuron. 2003;39:889–909. doi: 10.1016/s0896-6273(03)00568-3. [DOI] [PubMed] [Google Scholar]
- 30.Hsu LJ, et al. alpha-synuclein promotes mitochondrial deficit and oxidative stress. Am J Pathol. 2000;157:401–10. doi: 10.1016/s0002-9440(10)64553-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Song DD, et al. Enhanced substantia nigra mitochondrial pathology in human alphasynuclein transgenic mice after treatment with MPTP. Exp Neurol. 2004;186:158–72. doi: 10.1016/S0014-4886(03)00342-X. [DOI] [PubMed] [Google Scholar]
- 32.Dauer W, et al. Resistance of alpha -synuclein null mice to the parkinsonian neurotoxin MPTP. Proc Natl Acad Sci U S A. 2002;99:14524–9. doi: 10.1073/pnas.172514599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Klivenyi P, et al. Mice lacking alpha-synuclein are resistant to mitochondrial toxins. Neurobiol Dis. 2006;21:541–8. doi: 10.1016/j.nbd.2005.08.018. [DOI] [PubMed] [Google Scholar]
- 34.Beal MF, Ferrante J. Experimental therapeutics in transgenic mouse models of Huntington's disease. Nat Rev Neurosci. 2004;5:373–84. doi: 10.1038/nrn1386. [DOI] [PubMed] [Google Scholar]
- 35.Grafton ST, et al. Serial changes of cerebral glucose metabolism and caudate size in persons at risk for Huntington's disease. Arch Neurol. 1992;49:1161–7. doi: 10.1001/archneur.1992.00530350075022. [DOI] [PubMed] [Google Scholar]
- 36.Kuwert T, et al. Striatal glucose consumption in chorea-free subjects at risk of Huntington's disease. J Neurol. 1993;241:31–6. doi: 10.1007/BF00870669. [DOI] [PubMed] [Google Scholar]
- 37.Feigin A, et al. Metabolic network abnormalities in early Huntington's disease: an [(18)F]FDG PET study. J Nucl Med. 2001;42:1591–5. [PubMed] [Google Scholar]
- 38.Jenkins BG, et al. 1H NMR spectroscopy studies of Huntington's disease: correlations with CAG repeat numbers. Neurology. 1998;50:1357–65. doi: 10.1212/wnl.50.5.1357. [DOI] [PubMed] [Google Scholar]
- 39.Koroshetz WJ, et al. Energy metabolism defects in Huntington's disease and effects of coenzyme Q10. Ann Neurol. 1997;41:160–5. doi: 10.1002/ana.410410206. [DOI] [PubMed] [Google Scholar]
- 40.Browne SE, Beal MF. The energetics of Huntington's disease. Neurochem Res. 2004;29:531–46. doi: 10.1023/b:nere.0000014824.04728.dd. [DOI] [PubMed] [Google Scholar]
- 41.Gu M, et al. Mitochondrial defect in Huntington's disease caudate nucleus. Ann Neurol. 1996;39:385–9. doi: 10.1002/ana.410390317. [DOI] [PubMed] [Google Scholar]
- 42.Browne SE, et al. Oxidative damage and metabolic dysfunction in Huntington's disease: selective vulnerability of the basal ganglia. Ann Neurol. 1997;41:646–53. doi: 10.1002/ana.410410514. [DOI] [PubMed] [Google Scholar]
- 43.Tabrizi SJ, et al. Biochemical abnormalities and excitotoxicity in Huntington's disease brain. Ann Neurol. 1997;45:25–32. doi: 10.1002/1531-8249(199901)45:1<25::aid-art6>3.0.co;2-e. [DOI] [PubMed] [Google Scholar]
- 44.Powers WJ, et al. Selective defect of in vivo glycolysis in early Huntington's disease striatum. Proc Natl Acad Sci U S A. 2007;104:2945–9. doi: 10.1073/pnas.0609833104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Lodi R, et al. Abnormal in vivo skeletal muscle energy metabolism in Huntington's disease and dentatorubropallidoluysian atrophy. Ann Neurol. 2000;48:72–6. [PubMed] [Google Scholar]
- 46.Arenas J, et al. Complex I defect in muscle from patients with Huntington's disease. Ann Neurol. 1998;43:397–400. doi: 10.1002/ana.410430321. [DOI] [PubMed] [Google Scholar]
- 47.Parker WD, Jr., et al. Evidence for a defect in NADH: ubiquinone oxidoreductase (complex I) in Huntington's disease. Neurology. 1990;40:1231–4. doi: 10.1212/wnl.40.8.1231. [DOI] [PubMed] [Google Scholar]
- 48.Milakovic T, Johnson GV. Mitochondrial respiration and ATP production are significantly impaired in striatal cells expressing mutant huntingtin. J Biol Chem. 2005;280:30773–82. doi: 10.1074/jbc.M504749200. [DOI] [PubMed] [Google Scholar]
- 49.Panov AV, et al. Early mitochondrial calcium defects in Huntington's disease are a direct effect of polyglutamines. Nat Neurosci. 2002;5:731–6. doi: 10.1038/nn884. [DOI] [PubMed] [Google Scholar]
- 50.Squitieri F, et al. Severe ultrastructural mitochondrial changes in lymphoblasts homozygous for Huntington disease mutation. Mech Ageing Dev. 2006;127:217–20. doi: 10.1016/j.mad.2005.09.010. [DOI] [PubMed] [Google Scholar]
- 51.Seong IS, et al. HD CAG repeat implicates a dominant property of huntingtin in mitochondrial energy metabolism. Hum Mol Genet. 2005;14:2871–80. doi: 10.1093/hmg/ddi319. [DOI] [PubMed] [Google Scholar]
- 52.Benchoua A, et al. Involvement of mitochondrial complex II defects in neuronal death produced by N-terminus fragment of mutated huntingtin. Mol Biol Cell. 2006;17:1652–63. doi: 10.1091/mbc.E05-07-0607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Banoei MM, et al. Huntington's Disease and Mitochondrial DNA Deletions: Event or Regular Mechanism for Mutant Huntingtin Protein and CAG Repeats Expansion?! Cell Mol Neurobiol. 2007;27:867–75. doi: 10.1007/s10571-007-9206-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Chen CM, et al. Increased oxidative damage and mitochondrial abnormalities in the peripheral blood of Huntington's disease patients. Biochem Biophys Res Commun. 2007;359:335–40. doi: 10.1016/j.bbrc.2007.05.093. [DOI] [PubMed] [Google Scholar]
- 55.Beal MF, et al. Age-dependent striatal excitotoxic lesions produced by the endogenous mitochondrial inhibitor malonate. J Neurochem. 2007;61:1147–50. doi: 10.1111/j.1471-4159.1993.tb03633.x. [DOI] [PubMed] [Google Scholar]
- 56.Ludolph AC, et al. 3-Nitropropionic acid-exogenous animal neurotoxin and possible human striatal toxin. Can J Neurol Sci. 1991;18:492–8. doi: 10.1017/s0317167100032212. [DOI] [PubMed] [Google Scholar]
- 57.Choo YS, et al. Mutant huntingtin directly increases susceptibility of mitochondria to the calcium-induced permeability transition and cytochrome c release. Hum Mol Genet. 2004;13:1407–20. doi: 10.1093/hmg/ddh162. [DOI] [PubMed] [Google Scholar]
- 58.Panov AV, Lund S, Greenamyre JT. Ca2+-induced permeability transition in human lymphoblastoid cell mitochondria from normal and Huntington's disease individuals. Mol Cell Biochem. 2005;269:143–52. doi: 10.1007/s11010-005-3454-9. [DOI] [PubMed] [Google Scholar]
- 59.Milakovic T, Quintanilla RA, Johnson GV. Mutant huntingtin expression induces mitochondrial calcium handling defects in clonal striatal cells: functional consequences. J Biol Chem. 2006;281:34785–95. doi: 10.1074/jbc.M603845200. [DOI] [PubMed] [Google Scholar]
- 60.Trushina E, et al. Mutant huntingtin impairs axonal trafficking in mammalian neurons in vivo and in vitro. Mol Cell Biol. 2004;24:8195–209. doi: 10.1128/MCB.24.18.8195-8209.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Orr AL, et al. N-terminal mutant huntingtin associates with mitochondria and impairs mitochondrial trafficking. J Neurosci. 2008;28:2783–92. doi: 10.1523/JNEUROSCI.0106-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Sugars KL, Rubinsztein DC. Transcriptional abnormalities in Huntington disease. Trends Genet. 2003;19:233–8. doi: 10.1016/S0168-9525(03)00074-X. [DOI] [PubMed] [Google Scholar]
- 63.Sugars KL, et al. Decreased cAMP response element-mediated transcription: an early event in exon 1 and full-length cell models of Huntington's disease that contributes to polyglutamine pathogenesis. J Biol Chem. 2004;279:4988–99. doi: 10.1074/jbc.M310226200. [DOI] [PubMed] [Google Scholar]
- 64.Nucifora FC, Jr., et al. Interference by huntingtin and atrophin-1 with cbp-mediated transcription leading to cellular toxicity. Science. 2001;291:2423–8. doi: 10.1126/science.1056784. [DOI] [PubMed] [Google Scholar]
- 65.Chen-Plotkin AS, et al. Decreased association of the transcription factor Sp1 with genes downregulated in Huntington's disease. Neurobiol Dis. 2006;22:233–41. doi: 10.1016/j.nbd.2005.11.001. [DOI] [PubMed] [Google Scholar]
- 66.Zhai W, et al. In vitro analysis of huntingtin-mediated transcriptional repression reveals multiple transcription factor targets. Cell. 2005;123:1241–53. doi: 10.1016/j.cell.2005.10.030. [DOI] [PubMed] [Google Scholar]
- 67.Dunah AW, et al. Sp1 and TAFII130 transcriptional activity disrupted in early Huntington's disease. Science. 2002;296:2238–43. doi: 10.1126/science.1072613. [DOI] [PubMed] [Google Scholar]
- 68.Steffan JS, et al. The Huntington's disease protein interacts with p53 and CREB-binding protein and represses transcription. Proc Natl Acad Sci U S A. 2000;97:6763–8. doi: 10.1073/pnas.100110097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Bae BI, et al. p53 mediates cellular dysfunction and behavioral abnormalities in Huntington's disease. Neuron. 2005;47:29–41. doi: 10.1016/j.neuron.2005.06.005. [DOI] [PubMed] [Google Scholar]
- 70.Weydt P, et al. Thermoregulatory and metabolic defects in Huntington's disease transgenic mice implicate PGC-1alpha in Huntington's disease neurodegeneration. Cell Metab. 2006;4:349–62. doi: 10.1016/j.cmet.2006.10.004. [DOI] [PubMed] [Google Scholar]
- 71.Cui L, et al. Transcriptional repression of PGC-1alpha by mutant huntingtin leads to mitochondrial dysfunction and neurodegeneration. Cell. 2006;127:59–69. doi: 10.1016/j.cell.2006.09.015. [DOI] [PubMed] [Google Scholar]
- 72.McGill JK, Beal MF. PGC-1alpha, a new therapeutic target in Huntington's disease? Cell. 2006;127:465–8. doi: 10.1016/j.cell.2006.10.023. [DOI] [PubMed] [Google Scholar]
- 73.St-Pierre J, et al. Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators. Cell. 2006;127:397–408. doi: 10.1016/j.cell.2006.09.024. [DOI] [PubMed] [Google Scholar]
- 74.Huang HM, et al. Inhibition of the alpha-ketoglutarate dehydrogenase complex alters mitochondrial function and cellular calcium regulation. Biochim Biophys Acta. 2003;1637:119–26. doi: 10.1016/s0925-4439(02)00222-3. [DOI] [PubMed] [Google Scholar]
- 75.Bubber P, et al. Mitochondrial abnormalities in Alzheimer brain: mechanistic implications. Ann Neurol. 2005;57:695–703. doi: 10.1002/ana.20474. [DOI] [PubMed] [Google Scholar]
- 76.Valla J, et al. Impaired platelet mitochondrial activity in Alzheimer's disease and mild cognitive impairment. Mitochondrion. 2006;6:323–30. doi: 10.1016/j.mito.2006.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Parker WD, Jr., et al. Reduced platelet cytochrome c oxidase activity in Alzheimer's disease. Neurology. 1994;44:1086–90. doi: 10.1212/wnl.44.6.1086. [DOI] [PubMed] [Google Scholar]
- 78.Kish SJ, et al. Brain cytochrome oxidase in Alzheimer's disease. J Neurochem. 1992;59:776–9. doi: 10.1111/j.1471-4159.1992.tb09439.x. [DOI] [PubMed] [Google Scholar]
- 79.Bosetti F, et al. Cytochrome c oxidase and mitochondrial F1F0-ATPase (ATP synthase) activities in platelets and brain from patients with Alzheimer's disease. Neurobiol Aging. 2002;23:371–6. doi: 10.1016/s0197-4580(01)00314-1. [DOI] [PubMed] [Google Scholar]
- 80.Moreira PI, et al. Increased autophagic degradation of mitochondria in Alzheimer disease. Autophagy. 2007;3:614–5. doi: 10.4161/auto.4872. [DOI] [PubMed] [Google Scholar]
- 81.Hamblet NS, et al. Mutations in mitochondrial-encoded cytochrome c oxidase subunits I, II, and III genes detected in Alzheimer's disease using single-strand conformation polymorphism. Electrophoresis. 2006;27:398–408. doi: 10.1002/elps.200500420. [DOI] [PubMed] [Google Scholar]
- 82.Qiu X, Chen Y, Zhou M. Two point mutations in mitochondrial DNA of cytochrome coxidase coexist with normal mtDNA in a patient with Alzheimer's disease. Brain Res. 2001;893:261–3. doi: 10.1016/s0006-8993(00)03190-5. [DOI] [PubMed] [Google Scholar]
- 83.Lin MT, et al. High aggregate burden of somatic mtDNA point mutations in aging and Alzheimer's disease brain. Hum Mol Genet. 2002;11:133–45. doi: 10.1093/hmg/11.2.133. [DOI] [PubMed] [Google Scholar]
- 84.Elson JL, et al. Does the mitochondrial genome play a role in the etiology of Alzheimer's disease? Hum Genet. 2006;119:241–54. doi: 10.1007/s00439-005-0123-8. [DOI] [PubMed] [Google Scholar]
- 85.Anandatheerthavarada HK, et al. Mitochondrial targeting and a novel transmembrane arrest of Alzheimer's amyloid precursor protein impairs mitochondrial function in neuronal cells. J Cell Biol. 2003;161:41–54. doi: 10.1083/jcb.200207030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Caspersen C, et al. Mitochondrial Abeta: a potential focal point for neuronal metabolic dysfunction in Alzheimer's disease. Faseb J. 2005;19:2040–1. doi: 10.1096/fj.05-3735fje. [DOI] [PubMed] [Google Scholar]
- 87.Sasaki S, et al. Ultrastructural study of aggregates in the spinal cord of transgenic mice with a G93A mutant SOD1 gene. Acta Neuropathol. 2005;109:247–55. doi: 10.1007/s00401-004-0939-7. [DOI] [PubMed] [Google Scholar]
- 88.Sasaki S, Iwata M. Ultrastructural study of synapses in the anterior horn neurons of patients with amyotrophic lateral sclerosis. Neurosci Lett. 1996;204:53–6. doi: 10.1016/0304-3940(96)12314-4. [DOI] [PubMed] [Google Scholar]
- 89.Browne SE, et al. Metabolic dysfunction in familial, but not sporadic, amyotrophic lateral sclerosis. J Neurochem. 1998;71:281–7. doi: 10.1046/j.1471-4159.1998.71010281.x. [DOI] [PubMed] [Google Scholar]
- 90.Borthwick GM, et al. Mitochondrial enzyme activity in amyotrophic lateral sclerosis: implications for the role of mitochondria in neuronal cell death. Ann Neurol. 1999;46:787–90. doi: 10.1002/1531-8249(199911)46:5<787::aid-ana17>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
- 91.Hatazawa J, et al. Cortical motor-sensory hypometabolism in amyotrophic lateral sclerosis: a PET study. J Comput Assist Tomogr. 1988;12:630–6. doi: 10.1097/00004728-198807000-00019. [DOI] [PubMed] [Google Scholar]
- 92.Sasaki S, et al. Ultrastructural study of mitochondria in the spinal cord of transgenic mice with a G93A mutant SOD1 gene. Acta Neuropathol. 2004;107:461–74. doi: 10.1007/s00401-004-0837-z. [DOI] [PubMed] [Google Scholar]
- 93.Beretta S, et al. Mitochondrial dysfunction due to mutant copper/zinc superoxide dismutase associated with amyotrophic lateral sclerosis is reversed by N-acetylcysteine. Neurobiol Dis. 2003;13:213–21. doi: 10.1016/s0969-9961(03)00043-3. [DOI] [PubMed] [Google Scholar]
- 94.Kirby J, et al. Differential gene expression in a cell culture model of SOD1-related familial motor neurone disease. Hum Mol Genet. 2002;11:2061–75. doi: 10.1093/hmg/11.17.2061. [DOI] [PubMed] [Google Scholar]
- 95.Damiano M, et al. Neural mitochondrial Ca2+ capacity impairment precedes the onset of motor symptoms in G93A Cu/Zn-superoxide dismutase mutant mice. J Neurochem. 2006;96:1349–61. doi: 10.1111/j.1471-4159.2006.03619.x. [DOI] [PubMed] [Google Scholar]
- 96.Mattiazzi M, et al. Mutated human SOD1 causes dysfunction of oxidative phosphorylation in mitochondria of transgenic mice. J Biol Chem. 2002;277:29626–33. doi: 10.1074/jbc.M203065200. [DOI] [PubMed] [Google Scholar]
- 97.Liu J, et al. Toxicity of familial ALS-linked SOD1 mutants from selective recruitment to spinal mitochondria. Neuron. 2004;43:5–17. doi: 10.1016/j.neuron.2004.06.016. [DOI] [PubMed] [Google Scholar]
- 98.Vande Velde C, et al. Selective association of misfolded ALS-linked mutant SOD1 with the cytoplasmic face of mitochondria. Proc Natl Acad Sci U S A. 2008;105:4022–7. doi: 10.1073/pnas.0712209105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Pasinelli P, et al. Amyotrophic lateral sclerosis-associated SOD1 mutant proteins bind and aggregate with Bcl-2 in spinal cord mitochondria. Neuron. 2008;43:19–30. doi: 10.1016/j.neuron.2004.06.021. [DOI] [PubMed] [Google Scholar]
- 100.De Vos KJ, et al. Familial amyotrophic lateral sclerosis-linked SOD1 mutants perturb fast axonal transport to reduce axonal mitochondria content. Hum Mol Genet. 2007;16:2720–8. doi: 10.1093/hmg/ddm226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Beal MF. Bioenergetic approaches for neuroprotection in Parkinson's disease. Ann Neurol. 2003;53(Suppl 3):S39–47. doi: 10.1002/ana.10479. discussion S47-8. [DOI] [PubMed] [Google Scholar]
- 102.Juravleva E, et al. Creatine enhances survival of glutamate-treated neuronal/glial cells, modulates Ras/NF-kappaB signaling, and increases the generation of reactive oxygen species. J Neurosci Res. 2005;79:224–30. doi: 10.1002/jnr.20291. [DOI] [PubMed] [Google Scholar]
- 103.Ducray AD, et al. Creatine treatment promotes differentiation of GABA-ergic neuronal precursors in cultured fetal rat spinal cord. J Neurosci Res. 2007;85:1863–75. doi: 10.1002/jnr.21337. [DOI] [PubMed] [Google Scholar]
- 104.Andres RH, et al. Effects of creatine treatment on survival and differentiation of GABA-ergic neurons in cultured striatal tissue. J Neurochem. 2005;95:33–45. doi: 10.1111/j.1471-4159.2005.03337.x. [DOI] [PubMed] [Google Scholar]
- 105.Andres RH, et al. Effects of creatine treatment on the survival of dopaminergic neurons in cultured fetal ventral mesencephalic tissue. Neuroscience. 2005;133:701–13. doi: 10.1016/j.neuroscience.2005.03.004. [DOI] [PubMed] [Google Scholar]
- 106.Andres RH, et al. Creatine supplementation improves dopaminergic cell survival and protects against MPP+ toxicity in an organotypic tissue culture system. Cell Transplant. 2005;14:537–50. doi: 10.3727/000000005783982756. [DOI] [PubMed] [Google Scholar]
- 107.Bender A, et al. Creatine improves health and survival of mice. Neurobiol Aging. 2007. [DOI] [PubMed]
- 108.Matthews RT, et al. Creatine and cyclocreatine attenuate MPTP neurotoxicity. Exp Neurol. 1999;157:142–9. doi: 10.1006/exnr.1999.7049. [DOI] [PubMed] [Google Scholar]
- 109.Zhu S, et al. Prophylactic creatine administration mediates neuroprotection in cerebral ischemia in mice. J Neurosci. 2004;24:5909–12. doi: 10.1523/JNEUROSCI.1278-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Malcon C, Kaddurah-Daouk R, Beal MF. Neuroprotective effects of creatine administration against NMDA and malonate toxicity. Brain Res. 2000;860:195–8. doi: 10.1016/s0006-8993(00)02038-2. [DOI] [PubMed] [Google Scholar]
- 111.Klivenyi P, et al. Additive neuroprotective effects of creatine and a cyclooxygenase 2 inhibitor against dopamine depletion in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. J Mol Neurosci. 2003;21:191–8. doi: 10.1385/jmn:21:3:191. [DOI] [PubMed] [Google Scholar]
- 112.Klivenyi P, et al. Additive neuroprotective effects of creatine and cyclooxygenase 2 inhibitors in a transgenic mouse model of amyotrophic lateral sclerosis. J Neurochem. 2004;88:576–82. doi: 10.1046/j.1471-4159.2003.02160.x. [DOI] [PubMed] [Google Scholar]
- 113.Pena-Altamira E, et al. Neurochemical correlates of differential neuroprotection by longterm dietary creatine supplementation. Brain Res. 2005;1058:183–8. doi: 10.1016/j.brainres.2005.07.011. [DOI] [PubMed] [Google Scholar]
- 114.Klivenyi P, et al. Neuroprotective effects of creatine in a transgenic animal model of amyotrophic lateral sclerosis. Nat Med. 1999;5:347–50. doi: 10.1038/6568. [DOI] [PubMed] [Google Scholar]
- 115.Derave W, et al. Skeletal muscle properties in a transgenic mouse model for amyotrophic lateral sclerosis effects of creatine treatment. Neurobiol Dis. 2003;13:264–72. doi: 10.1016/s0969-9961(03)00041-x. [DOI] [PubMed] [Google Scholar]
- 116.Andreassen OA, et al. Increases in cortical glutamate concentrations in transgenic amyotrophic lateral sclerosis mice are attenuated by creatine supplementation. J Neurochem. 2001;77:383–90. doi: 10.1046/j.1471-4159.2001.00188.x. [DOI] [PubMed] [Google Scholar]
- 117.Zhang W, Narayanan M, Friedlander RM. Additive neuroprotective effects of minocycline with creatine in a mouse model of ALS. Ann Neurol. 2003;53:267–70. doi: 10.1002/ana.10476. [DOI] [PubMed] [Google Scholar]
- 118.Andreassen OA, et al. Creatine increase survival and delays motor symptoms in a transgenic animal model of Huntington's disease. Neurobiol Dis. 2001;8:479–91. doi: 10.1006/nbdi.2001.0406. [DOI] [PubMed] [Google Scholar]
- 119.Dedeoglu A, et al. Creatine therapy provides neuroprotection after onset of clinical symptoms in Huntington's disease transgenic mice. J Neurochem. 2003;85:1359–67. doi: 10.1046/j.1471-4159.2003.01706.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Ferrante RJ, et al. Neuroprotective effects of creatine in a transgenic mouse model of Huntington's disease. J Neurosci. 2000;20:4389–97. doi: 10.1523/JNEUROSCI.20-12-04389.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Brewer GJ, Wallimann TW. Protective effect of the energy precursor creatine against toxicity of glutamate and beta-amyloid in rat hippocampal neurons. J Neurochem. 2000;74:1968–78. doi: 10.1046/j.1471-4159.2000.0741968.x. [DOI] [PubMed] [Google Scholar]
- 122.Sullivan PG, et al. Dietary supplement creatine protects against traumatic brain injury. Ann Neurol. 2000;48:723–9. [PubMed] [Google Scholar]
- 123.Hausmann ON, et al. Protective effects of oral creatine supplementation on spinal cord injury in rats. Spinal Cord. 2002;40:449–56. doi: 10.1038/sj.sc.3101330. [DOI] [PubMed] [Google Scholar]
- 124.Rabchevsky AG, et al. Creatine diet supplement for spinal cord injury: influences on functional recovery and tissue sparing in rats. J Neurotrauma. 2003;20:659–69. doi: 10.1089/089771503322144572. [DOI] [PubMed] [Google Scholar]
- 125.Adcock KH, et al. Neuroprotection of creatine supplementation in neonatal rats with transient cerebral hypoxia-ischemia. Dev Neurosci. 2003;24:382–8. doi: 10.1159/000069043. [DOI] [PubMed] [Google Scholar]
- 126.Prass K, et al. Improved reperfusion and neuroprotection by creatine in a mouse model of stroke. J Cereb Blood Flow Metab. 2007;27:452–9. doi: 10.1038/sj.jcbfm.9600351. [DOI] [PubMed] [Google Scholar]
- 127.Wilken B, et al. Anoxic ATP depletion in neonatal mice brainstem is prevented by creatine supplementation. Arch Dis Child Fetal Neonatal Ed. 2000;82:F224–7. doi: 10.1136/fn.82.3.F224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Rodriguez MC, et al. Beneficial effects of creatine, CoQ10, and lipoic acid in mitochondrial disorders. Muscle Nerve. 2000;35:235–42. doi: 10.1002/mus.20688. [DOI] [PubMed] [Google Scholar]
- 129.Bender A, et al. Creatine supplementation in Parkinson disease: a placebo-controlled randomized pilot trial. Neurology. 2006;67:1262–4. doi: 10.1212/01.wnl.0000238518.34389.12. [DOI] [PubMed] [Google Scholar]
- 130.A randomized, double-blind, futility clinical trial of creatine and minocycline in early Parkinson disease. Neurology. 2006;66:664–71. doi: 10.1212/01.wnl.0000201252.57661.e1. [DOI] [PubMed] [Google Scholar]
- 131.Hass CJ, Collins MA, Juncos JL. Resistance training with creatine monohydrate improves upper-body strength in patients with Parkinson disease: a randomized trial. Neurorehabil Neural Repair. 2007;21:107–15. doi: 10.1177/1545968306293449. [DOI] [PubMed] [Google Scholar]
- 132.Bloom MZ. NIH announces phase III clinical trial of creatine for Parkinson's disease. Consult Pharm. 2007;22:378. [PubMed] [Google Scholar]
- 133.Couzin J. Clinical research. Testing a novel strategy against Parkinson's disease. Science. 2007;315:1778. doi: 10.1126/science.315.5820.1778. [DOI] [PubMed] [Google Scholar]
- 134.Hersch SM, et al. Creatine in Huntington disease is safe, tolerable, bioavailable in brain and reduces serum 8OH2'dG. Neurology. 2006;66:250–2. doi: 10.1212/01.wnl.0000194318.74946.b6. [DOI] [PubMed] [Google Scholar]
- 135.Littarru GP, Tiano L. Bioenergetic and antioxidant properties of coenzyme Q10: recent developments. Mol Biotechnol. 2007;37:31–7. doi: 10.1007/s12033-007-0052-y. [DOI] [PubMed] [Google Scholar]
- 136.Naderi J, et al. Water-soluble formulation of Coenzyme Q10 inhibits Bax-induced destabilization of mitochondria in mammalian cells. Apoptosis. 2006;11:1359–69. doi: 10.1007/s10495-006-8417-4. [DOI] [PubMed] [Google Scholar]
- 137.Papucci L, et al. Coenzyme q10 prevents apoptosis by inhibiting mitochondrial depolarization independently of its free radical scavenging property. J Biol Chem. 2003;278:28220–8. doi: 10.1074/jbc.M302297200. [DOI] [PubMed] [Google Scholar]
- 138.Beal MF. Therapeutic effects of coenzyme Q10 in neurodegenerative diseases. Methods Enzymol. 2004;382:473–87. doi: 10.1016/S0076-6879(04)82026-3. [DOI] [PubMed] [Google Scholar]
- 139.Schulz JB, et al. Coenzyme Q10 and nicotinamide and a free radical spin trap protect against MPTP neurotoxicity. Exp Neurol. 1995;132:279–83. doi: 10.1016/0014-4886(95)90033-0. [DOI] [PubMed] [Google Scholar]
- 140.McCarthy S, et al. Paraquat induces oxidative stress and neuronal cell death; neuroprotection by water-soluble Coenzyme Q10. Toxicol Appl Pharmacol. 2004;201:21–31. doi: 10.1016/j.taap.2004.04.019. [DOI] [PubMed] [Google Scholar]
- 141.Moon Y, et al. Mitochondrial membrane depolarization and the selective death of dopaminergic neurons by rotenone: protective effect of coenzyme Q10. J Neurochem. 2005;93:1199–208. doi: 10.1111/j.1471-4159.2005.03112.x. [DOI] [PubMed] [Google Scholar]
- 142.Kooncumchoo P, et al. Coenzyme Q(10) provides neuroprotection in iron-induced apoptosis in dopaminergic neurons. J Mol Neurosci. 2006;28:125–41. doi: 10.1385/JMN:28:2:125. [DOI] [PubMed] [Google Scholar]
- 143.Ono K, et al. Preformed beta-amyloid fibrils are destabilized by coenzyme Q10 in vitro. Biochem Biophys Res Commun. 2005;330:111–6. doi: 10.1016/j.bbrc.2005.02.132. [DOI] [PubMed] [Google Scholar]
- 144.Li G, et al. Coenzyme Q10 protects SHSY5Y neuronal cells from beta amyloid toxicity and oxygen-glucose deprivation by inhibiting the opening of the mitochondrial permeability transition pore. Biofactors. 2005;25:97–107. doi: 10.1002/biof.5520250111. [DOI] [PubMed] [Google Scholar]
- 145.Somayajulu M, et al. Role of mitochondria in neuronal cell death induced by oxidative stress; neuroprotection by Coenzyme Q10. Neurobiol Dis. 2005;18:618–27. doi: 10.1016/j.nbd.2004.10.021. [DOI] [PubMed] [Google Scholar]
- 146.Beal MF, et al. Coenzyme Q10 attenuates the 1-methyl-4-phenyl-1,2,3,tetrahydropyridine (MPTP) induced loss of striatal dopamine and dopaminergic axons in aged mice. Brain Res. 1998;783:109–14. doi: 10.1016/s0006-8993(97)01192-x. [DOI] [PubMed] [Google Scholar]
- 147.Cleren C, et al. Therapeutic effects of coenzyme Q(10) (Q(10)) and reduced CoQ(10) in the MPTP model of Parkinsonism. J Neurochem. 2007. [DOI] [PubMed]
- 148.Beal MF, et al. Coenzyme Q10 and nicotinamide block striatal lesions produced by the mitochondrial toxin malonate. Ann Neurol. 1994;36:882–8. doi: 10.1002/ana.410360613. [DOI] [PubMed] [Google Scholar]
- 149.Brouillet E, et al. Aminooxyacetic acid striatal lesions attenuated by 1,3-butanediol and coenzyme Q10. Neurosci Lett. 1994;177:58–62. doi: 10.1016/0304-3940(94)90044-2. [DOI] [PubMed] [Google Scholar]
- 150.Matthews RT, et al. Coenzyme Q10 administration increases brain mitochondrial concentrations and exerts neuroprotective effects. Proc Natl Acad Sci U S A. 1998;95:8892–7. doi: 10.1073/pnas.95.15.8892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Ferrante RJ, et al. Therapeutic effects of coenzyme Q10 and remacemide in transgenic mouse models of Huntington's disease. J Neurosci. 2002;22:1592–9. doi: 10.1523/JNEUROSCI.22-05-01592.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Smith KM, et al. Dose ranging and efficacy study of high-dose coenzyme Q10 formulations in Huntington's disease mice. Biochim Biophys Acta. 2006;1762:616–26. doi: 10.1016/j.bbadis.2006.03.004. [DOI] [PubMed] [Google Scholar]
- 153.Stack EC, et al. Combination therapy using minocycline and coenzyme Q10 in R6/2 transgenic Huntington's disease mice. Biochim Biophys Acta. 2006;1762:373–80. doi: 10.1016/j.bbadis.2005.11.002. [DOI] [PubMed] [Google Scholar]
- 154.Schilling G, et al. Coenzyme Q10 and remacemide hydrochloride ameliorate motor deficits in a Huntington's disease transgenic mouse model. Neurosci Lett. 2001;315:149–53. doi: 10.1016/s0304-3940(01)02326-6. [DOI] [PubMed] [Google Scholar]
- 155.A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington's disease. Neurology. 2001;57:397–404. doi: 10.1212/wnl.57.3.397. [DOI] [PubMed] [Google Scholar]
- 156.Gotz ME, et al. Altered redox state of platelet coenzyme Q10 in Parkinson's disease. J Neural Transm. 2000;107:41–8. doi: 10.1007/s007020050003. [DOI] [PubMed] [Google Scholar]
- 157.Isobe C, et al. Increase of oxidized/total coenzyme Q-10 ratio in cerebrospinal fluid in patients with Parkinson's disease. J Clin Neurosci. 2007;14:340–3. doi: 10.1016/j.jocn.2005.12.023. [DOI] [PubMed] [Google Scholar]
- 158.Sohmiya M, et al. An increase of oxidized coenzyme Q-10 occurs in the plasma of sporadic ALS patients. J Neurol Sci. 2005;228:49–53. doi: 10.1016/j.jns.2004.09.030. [DOI] [PubMed] [Google Scholar]
- 159.Shults CW, et al. Coenzyme Q10 levels correlate with the activities of complexes I and II/III in mitochondria from parkinsonian and nonparkinsonian subjects. Ann Neurol. 1997;42:261–4. doi: 10.1002/ana.410420221. [DOI] [PubMed] [Google Scholar]
- 160.Shults CW, et al. Effects of coenzyme Q10 in early Parkinson disease: evidence of slowing of the functional decline. Arch Neurol. 2002;59:1541–50. doi: 10.1001/archneur.59.10.1541. [DOI] [PubMed] [Google Scholar]
- 161.Shults CW, et al. Pilot trial of high dosages of coenzyme Q10 in patients with Parkinson's disease. Exp Neurol. 2004;188:491–4. doi: 10.1016/j.expneurol.2004.05.003. [DOI] [PubMed] [Google Scholar]
- 162.Ferrante KL, et al. Tolerance of high-dose (3,000 mg/day) coenzyme Q10 in ALS. Neurology. 2005;65:1834–6. doi: 10.1212/01.wnl.0000187070.35365.d7. [DOI] [PubMed] [Google Scholar]
- 163.Storch A, et al. Randomized, double-blind, placebo-controlled trial on symptomatic effects of coenzyme Q(10) in Parkinson disease. Arch Neurol. 2007;64:938–44. doi: 10.1001/archneur.64.7.nct60005. [DOI] [PubMed] [Google Scholar]
- 164.Pereira C, Santos MS, Oliveira C. Involvement of oxidative stress on the impairment of energy metabolism induced by A beta peptides on PC12 cells: protection by antioxidants. Neurobiol Dis. 1999;6:209–19. doi: 10.1006/nbdi.1999.0241. [DOI] [PubMed] [Google Scholar]
- 165.Yamada K, et al. Protective effects of idebenone and alpha-tocopherol on beta-amyloid-(1-42)-induced learning and memory deficits in rats: implication of oxidative stress in beta-amyloidinduced neurotoxicity in vivo. Eur J Neurosci. 1999;11:83–90. doi: 10.1046/j.1460-9568.1999.00408.x. [DOI] [PubMed] [Google Scholar]
- 166.Senin U, et al. Idebenone in senile dementia of Alzheimer type: a multicentre study. Arch Gerontol Geriatr. 1992;15:249–60. doi: 10.1016/0167-4943(92)90060-h. [DOI] [PubMed] [Google Scholar]
- 167.Weyer G, et al. A controlled study of 2 doses of idebenone in the treatment of Alzheimer's disease. Neuropsychobiology. 1997;36:73–82. doi: 10.1159/000119366. [DOI] [PubMed] [Google Scholar]
- 168.Gutzmann H, Hadler D. Sustained efficacy and safety of idebenone in the treatment of Alzheimer's disease: update on a 2-year double-blind multicentre study. J Neural Transm Suppl. 1998;54:301–10. doi: 10.1007/978-3-7091-7508-8_30. [DOI] [PubMed] [Google Scholar]
- 169.Gutzmann H, et al. Safety and efficacy of idebenone versus tacrine in patients with Alzheimer's disease: results of a randomized, double-blind, parallel-group multicenter study. Pharmacopsychiatry. 2002;35:12–8. doi: 10.1055/s-2002-19833. [DOI] [PubMed] [Google Scholar]
- 170.Thal LJ, et al. Idebenone treatment fails to slow cognitive decline in Alzheimer's disease. Neurology. 2003;61:1498–502. doi: 10.1212/01.wnl.0000096376.03678.c1. [DOI] [PubMed] [Google Scholar]
- 171.Ranen NG, et al. A controlled trial of idebenone in Huntington's disease. Mov Disord. 1996;11:549–54. doi: 10.1002/mds.870110510. [DOI] [PubMed] [Google Scholar]
- 172.Di Prospero NA, et al. Neurological effects of high-dose idebenone in patients with Friedreich's ataxia: a randomized, placebo-controlled trial. Lancet Neurol. 2007;6:878–86. doi: 10.1016/S1474-4422(07)70220-X. [DOI] [PubMed] [Google Scholar]
- 173.Armstrong JS. Mitochondria-Directed Therapeutics. Antioxid Redox Signal. 2007. [DOI] [PubMed]
- 174.Tauskela JS. MitoQ--a mitochondria-targeted antioxidant. IDrugs. 2007;10:399–412. [PubMed] [Google Scholar]
- 175.Lu C, et al. Is Antioxidant Potential of the Mitochondrial Targeted Ubiquinone Derivative MitoQ Conserved in Cells Lacking mtDNA? Antioxid Redox Signal. 2007. [DOI] [PubMed]
- 176.Szeto HH. Mitochondria-Targeted Cytoprotective Peptides for Ischemia-Reperfusion Injury. Antioxid Redox Signal. 2007. [DOI] [PubMed]
- 177.Zhao K, et al. Mitochondria-targeted peptide prevents mitochondrial depolarization and apoptosis induced by tert-butyl hydroperoxide in neuronal cell lines. Biochem Pharmacol. 2005;70:1796–806. doi: 10.1016/j.bcp.2005.08.022. [DOI] [PubMed] [Google Scholar]
- 178.Zhao K, et al. Cell-permeable peptide antioxidants targeted to inner mitochondrial membrane inhibit mitochondrial swelling, oxidative cell death, and reperfusion injury. J Biol Chem. 2004;279:34682–90. doi: 10.1074/jbc.M402999200. [DOI] [PubMed] [Google Scholar]
- 179.Cho J, et al. Potent mitochondria-targeted peptides reduce myocardial infarction in rats. Coron Artery Dis. 2007;18:215–20. doi: 10.1097/01.mca.0000236285.71683.b6. [DOI] [PubMed] [Google Scholar]
- 180.Petri S, et al. Cell-permeable peptide antioxidants as a novel therapeutic approach in a mouse model of amyotrophic lateral sclerosis. J Neurochem. 2006;98:1141–8. doi: 10.1111/j.1471-4159.2006.04018.x. [DOI] [PubMed] [Google Scholar]
- 181.Liby KT, Yore MM, Sporn MB. Triterpenoids and rexinoids as multifunctional agents for the prevention and treatment of cancer. Nat Rev Cancer. 2007;7:357–69. doi: 10.1038/nrc2129. [DOI] [PubMed] [Google Scholar]
- 182.Lee JM, et al. Nrf2, a multi-organ protector? Faseb J. 2005;19:1061–6. doi: 10.1096/fj.04-2591hyp. [DOI] [PubMed] [Google Scholar]
- 183.Shih AY, Li P, Murphy TH. A small-molecule-inducible Nrf2-mediated antioxidant response provides effective prophylaxis against cerebral ischemia in vivo. J Neurosci. 2005;25:10321–35. doi: 10.1523/JNEUROSCI.4014-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Lee JM, et al. NF-E2-related factor-2 mediates neuroprotection against mitochondrial complex I inhibitors and increased concentrations of intracellular calcium in primary cortical neurons. J Biol Chem. 2003;278:37948–56. doi: 10.1074/jbc.M305204200. [DOI] [PubMed] [Google Scholar]
- 185.Shih AY, et al. Induction of the Nrf2-driven antioxidant response confers neuroprotection during mitochondrial stress in vivo. J Biol Chem. 2005;280:22925–36. doi: 10.1074/jbc.M414635200. [DOI] [PubMed] [Google Scholar]
- 186.Calkins MJ, et al. Protection from mitochondrial complex II inhibition in vitro and in vivo by Nrf2-mediated transcription. Proc Natl Acad Sci U S A. 2005;102:244–9. doi: 10.1073/pnas.0408487101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Andreassen OA, et al. Lipoic acid improves survival in transgenic mouse models of Huntington's disease. Neuroreport. 2001;12:3371–3. doi: 10.1097/00001756-200110290-00044. [DOI] [PubMed] [Google Scholar]
- 188.Andreassen OA, et al. Effects of an inhibitor of poly(ADP-ribose) polymerase, desmethylselegiline, trientine, and lipoic acid in transgenic ALS mice. Exp Neurol. 2001;168:419–24. doi: 10.1006/exnr.2001.7633. [DOI] [PubMed] [Google Scholar]
- 189.Bodis-Wollner I, et al. Acetyl-levo-carnitine protects against MPTP-induced parkinsonism in primates. J Neural Transm Park Dis Dement Sect. 1991;3:63–72. doi: 10.1007/BF02251137. [DOI] [PubMed] [Google Scholar]
- 190.Wang C, et al. L-carnitine protects neurons from 1-methyl-4-phenylpyridinium-induced neuronal apoptosis in rat forebrain culture. Neuroscience. 2007;144:46–55. doi: 10.1016/j.neuroscience.2006.08.083. [DOI] [PubMed] [Google Scholar]
- 191.Tieu K, et al. D-beta-hydroxybutyrate rescues mitochondrial respiration and mitigates features of Parkinson disease. J Clin Invest. 2003;112:892–901. doi: 10.1172/JCI18797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Dykens JA, Moos WH, Howell N. Development of 17alpha-estradiol as a neuroprotective therapeutic agent: rationale and results from a phase I clinical study. Ann N Y Acad Sci. 2005;1052:116–35. doi: 10.1196/annals.1347.008. [DOI] [PubMed] [Google Scholar]
- 193.Dali-Youcef N, et al. Sirtuins: the ‘magnificent seven’, function, metabolism and longevity. Ann Med. 2007;39:335–45. doi: 10.1080/07853890701408194. [DOI] [PubMed] [Google Scholar]
- 194.Kim D, et al. SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer's disease and amyotrophic lateral sclerosis. Embo J. 2007;26:3169–79. doi: 10.1038/sj.emboj.7601758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Baur JA, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006;444:337–42. doi: 10.1038/nature05354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Lagouge M, et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell. 2006;127:1109–22. doi: 10.1016/j.cell.2006.11.013. [DOI] [PubMed] [Google Scholar]
- 197.Qin W, et al. Neuronal SIRT1 activation as a novel mechanism underlying the prevention of Alzheimer disease amyloid neuropathology by calorie restriction. J Biol Chem. 2006;281:21745–54. doi: 10.1074/jbc.M602909200. [DOI] [PubMed] [Google Scholar]
- 198.Kumar P, et al. Effect of resveratrol on 3-nitropropionic acid-induced biochemical and behavioural changes: possible neuroprotective mechanisms. Behav Pharmacol. 2006;17:485–92. doi: 10.1097/00008877-200609000-00014. [DOI] [PubMed] [Google Scholar]
- 199.Chen J, et al. SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-kappaB signaling. J Biol Chem. 2005;280:40364–74. doi: 10.1074/jbc.M509329200. [DOI] [PubMed] [Google Scholar]