Glutathione Depletion and MicroRNA Dysregulation in MSA: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , ,

Multiple system atrophy (MSA) is a rare neurodegenerative disease characterized by parkinsonism, cerebellar impairment, and autonomic failure. Although the causes of MSA onset and progression remain uncertain, its pathogenesis may involve oxidative stress via the generation of excess reactive oxygen species and/or destruction of the antioxidant system. One of the most powerful antioxidants is glutathione, which plays essential roles as an antioxidant enzyme cofactor, cysteine-storage molecule, major redox buffer, and neuromodulator, in addition to being a key antioxidant in the central nervous system. Glutathione levels are known to be reduced in neurodegenerative diseases. In addition, genes regulating redox states have been shown to be post-transcriptionally modified by microRNA (miRNA), one of the most important types of non-coding RNA. miRNAs have been reported to be dysregulated in several diseases, including MSA.

  • multiple system atrophy
  • neurodegenerative disease
  • glutathione
  • microRNA
  • oxidative stress
  • α-synuclein

1. Introduction

Multiple system atrophy (MSA) is a rare, adult-onset, fatal neurodegenerative disease (ND) characterized by progressive loss of neuronal and oligodendroglial cells in various sites in the brain [1]. Disease onset is usually around 50 years old or later, with the age distribution of onset peaking in the late 50s. Currently, MSA is diagnosed when parkinsonism or cerebellar ataxia presents with prominent autonomic failure, and it is often accompanied by rapid eye movement (REM) sleep behavior disorder [2]. MSA belongs to the group of α-synucleinopathies, which are morphologically characterized by abnormal accumulation of fibrillary α-synuclein (α-syn) in the neurons and oligodendrocytes [3]. Oligodendroglial inclusions are specifically detected in the brains of MSA patients; these inclusions are the main hallmark of the disease and are considered to play a critical role in the primary events leading to MSA [4]. Although the origin of α-syn inclusions in the oligodendrocytes has been controversial, the leading hypotheses are that either internalization of neuron-secreted α-syn or de novo α-syn abnormality in oligodendrocytes is a key event in the pathogenic cascade leading to the propagation and spread of α-synucleinopathies of MSA [5]. Symptomatic therapies are available for MSA-associated parkinsonism and autonomic failures, but the response to these treatments is often poor [6]. Moreover, there is currently no effective medicine to ameliorate or even slow the progress of MSA. The rapid and devastating disease progression of MSA symptoms results in a relatively short survival period after disease onset [7]. Indeed, the estimated survival is approximately 9 years from symptom onset [8].
Because of the low incidence and uncertain diagnosis, there have been far fewer studies on MSA than on major NDs such as Alzheimer’s disease (AD), Parkinson’s disease (PD) and amyotrophic lateral sclerosis (ALS). Recently, however, improvements in the diagnostic certainty and advances in genetic technologies have promoted investigation of the etiological mechanisms of MSA. Although no hereditable mutations have been identified in MSA, some family history of genetic mutations as well as polymorphisms in unrelated patients with MSA have been found [9][10][11][12][13][14]. Recently, microRNA (miRNA) has emerged as a novel component of the gene expression regulation for maintaining brain functions [15]. Abnormalities in miRNA expression have been reported to cause several NDs, including MSA [16][17]. The first genome-wide miRNA analysis of MSA brain tissue revealed dysregulation of miRNAs, resulting in downregulation of the solute carrier family transporters for glutathione (GSH) and taurine, both of which are important antioxidants for neuroprotection against oxidative stress [18]. It is generally thought that the initiation and progression of NDs are likely induced by oxidative stress via an imbalance of oxidants and antioxidants [19][20]. GSH is known as a key protector against oxidative stress, and both nuclear magnetic resonance spectral and postmortem analyses have shown GSH depletion in the brains of patients with NDs [21][22][23][24][25][26][27][28]

2. The Association between GSH Dysregulation and MSA

The fact that mutations of COQ2, which is an important factor for regulating redox states, were found in specific populations of patients with MSA [29] suggests that oxidative stress in the brain is associated with the cause or progress of the disease. The brain might be inherently more vulnerable to oxidative damage due toROS, since the brain consumes more oxygen to produce energy—and consequently generates higher levels of toxic ROS—compared to other organs [30]. In addition, the brain contains an abundance of lipids with unsaturated fatty acids that act as a source of peroxidation [31]. Both features indicate the importance of neuroprotective antioxidants in the context of MSA and other NDs.

2.1. GSH Levels in Patients with MSA

GSH is one of the most effective antioxidants against oxidative stress in the brain [32]; other antioxidants, such as catalase, superoxide dismutase (SOD) and glutathione peroxidase (GPx), are present but expressed at lower levels there [33][34]. GSH detoxifies all types of ROS, including superoxide anions (O2), hydroxyl radicals (OH), hydroperoxyl radicals (HO2) and hydrogen peroxide (H2O2), and also acts as an electron donor, resulting in disulfide bond formation to produce oxidized glutathione (GSSG) [35]. GSSG is a substrate of the flavoenzyme glutathione reductase (GR), which transfers an electron from nicotinamide adenine dinucleotide phosphate (NADPH) to GSSG, thereby regenerating GSH and constituting a system for recycling GSH [36]. A trend of decreased levels of nigral GSH has been reported in the postmortem brains of patients with MSA compared to healthy controls, although the difference did not reach statistical significance [37][38]. Another report found a lower ratio of reduced GSH to oxidized GSSG, an index of oxidative stress, in the substantia nigra of patients with MSA, although, the change was not statistically significant [39]. These reports suggest that a decline in GSH exacerbates oxidative stress in the brains of MSA patients. Interestingly, in vitro experiments showed that the amyloid formation of α-syn was significantly facilitated by GSSG while GSH suppressed aggregation [40]. Furthermore, several studies have shown that increased intracellular GSH levels partially alleviated α-syn oligomerization and its cytotoxicity [41][42][43]. These results suggest that GSH depletion followed by oxidative stress promotes the pathological fibrillation of α-syn (Figure 1).
Figure 1. Association between the state of α-synuclein and conversion of GSH and GSSG.
The states of α-synuclein (α-syn) may depend on the redox states of glutathione, which are regulated by glutathione reductase (GR) and glutathione peroxidase (GPx). GR transfers an electron from nicotinamide adenine dinucleotide phosphate (NADPH) to GSSG and thereby catalyzes the reduction of GSSG to GSH. GPx reduces peroxide to a harmless compound by gathering the needed reducing equivalents from GSH. The aggregation ofα-syn is suppressed by ample GSH while the amyloid formation of α-syn is facilitated by GSSG. In addition, GPx markedly enhances the fibrillation of α-syn.

2.2. Possible Association of the Enzymes for GSH Synthesis and Metabolism with MSA

Synthesized GSH has been shown to react non-enzymatically with ROS or to function as an electron donor for the reduction of peroxides in the GPx reaction [44]. The formation of a disulfide bond between two GSH molecules gives rise to GSSG during this process. In a 1986 report, there were no significant alterations in GPx activity in the autopsied brains of patients with SND compared to healthy controls, suggesting that neuronal cell loss is unlikely to result from reduced activity of brain GPx [45]. However, there has been a report showing that α-syn enhanced in vitro GPx activity which in turn markedly enhanced fibrillation of α-syn [46]. Further studies at the cellular level are required to elucidate the role of GPx activity in the brains of patients with MSA.
Glutathione-S-transferases (GSTs) are a superfamily of phase 2 detoxification enzymes that detoxify both ROS and toxic xenobiotics, primarily by catalyzing GSH-dependent conjugation and redox reactions [47]. It has been reported that immunopositivity of GST in oligodendroglial inclusions did not show topographical linkage to neuronal degeneration [48]. In addition, a PubMed search revealed no studies showing dysregulation of the enzymes related to GSH synthesis and metabolism in the brains of patients with MSA. However, since the number of studies describing the physiological characteristics of MSA is still quite limited, further investigations are warranted to determine whether GSH and its related enzymes are dysregulated in the brains of patients with MSA.

2.3. Association between Transporters Related to GSH Biosynthesis and MSA

GSH is known to be ubiquitously distributed in many cells as a major non-protein thiol, which also functions as a storage and transport form of cysteine and an important player in antioxidative defense [49]. An excess amount of cysteine can be toxic to cells, because excess cysteine induces free radical generation and extracellular glutamate production [50][51]. Additionally, cysteine can impair mitochondrial respiration by limiting iron bioavailability through an oxidant-based mechanism [52].
GSH is a tripeptide composed of three amino acids: cysteine, glutamate, and glycine [32]. Among these amino acids, cysteine is the rate-limiting substrate for enzyme activity for GSH synthesis, because the other precursor amino acids, glutamate and glycine, have much higher intracellular concentrations than cysteine [49][53]. In agreement with these findings, it has been shown that GSH concentrations can only be elevated by increasing cytosolic cysteine availability [54]. The biosynthesis of GSH occurs via a two-step ATP-requiring enzymatic process that is catalyzed by glutamate-cysteine ligase (GCL; also known as γ-glutamylcysteine synthetase) and glutathione synthetase (GSS) [55][56]. GCL, which is composed of catalytic and modulatory subunits (GCLc and GCLm, respectively), catalyzes the formation of a dipeptide, γ-glutamylcysteine (γ-GluCys), from glutamate and cysteine, the rate-limiting step in GSH biosynthesis [55]. GSS is the critical enzyme for the second step in GSH biosynthesis, which couples γ-GluCys with glycine to generate GSH [56].
In neurons, cysteine uptake is mostly mediated through a neuronal sodium-dependent transporter known as excitatory amino acid carrier 1 (EAAC1; also known as EAAT3 or SLC1A1) [57]. EAAC1 is a member of the excitatory amino acid transporters (EAATs) that belong to the solute carrier family SLC1A. Acidic amino acid transport by the EAATs is coupled to the co-transport of three sodium ions and one proton, and the counter-transport of one potassium ion [58]. It has been reported that EAAC1 is expressed mainly in neurons and partially in subsets of oligodendrocytes, in immature oligodendrocytes, and in oligodendrocyte progenitor cells, but not expressed in mature astrocytes, in the brain [59]. EAAC1-deficient mice showed the age-dependent brain atrophy of both the hippocampal CA1 cell layer and the corpus callosum [60]. Interestingly, EAAC1 has been found to be downregulated in the postmortem brains of patients with MSA and in an animal model of MSA [18]. Alanine-serine-cysteine transporters (ASCTs) are also included among the SLC1A family members, which share sequence homology with the EAATs, and transport the neutral amino acids serine, alanine and cysteine [61]. ASCTs have been shown to participate in an amino acid antiport cycle, in which the exchange of neutral amino acids between the extracellular and intracellular sides is coupled to the co-transport of sodium ions from the extracellular side, without involvement of potassium ions or protons. Cysteine might act as a substrate of ASCT1 (also known as SLC1A4), which is mainly distributed in the astrocytes but also partially localized in neurons. Since cysteine uptake for GSH biosynthesis is mainly mediated by EAAC1, the contribution of ASCT1 to GSH generation might be limited, but could possibly support the function of EAAC1 [62] (Figure 2). Polymorphism of ASCT1 has been confirmed in some pathological conditions characterized by alterations of brain development and function [63][64][65][66][67][68]. Indeed, a study investigating oxidative-stress gene in MSA found significant associations between MSA and polymorphisms of ASCT1 [69], suggesting that the function of ASCT1 in neurons and oligodendrocytes is important for the maintenance of redox states.
Figure 2. Biosynthesis of intracellular glutathione in neurons, microglia and oligodendrocytes. (a) Glutathione (GSH) is comprised of three amino acids: cysteine (Cys), glutamate (Glu) and glycine (Gly), of which Cys is rate-limiting, with its uptake mainly mediated by EAAC1. ASCT1 also contributes to Cys uptake, but its ability might be more limited. Transported Cys is conjugated with Glu catalyzed by glutamate-cysteine ligase (GCL) and then couples with Gly to form GSH. (b) EAAC1 and ASCT1 are members of the SLC1A family, comprised of excitatory amino acid transporter (EAAT) and neutral amino acid transporter (ASCT) subtypes.

3. The Association of miRNA Dysregulation and MSA Pathology

Increasing lines of evidence suggest that over-expression or down-regulation of non-coding RNAs is involved in the onset and progression of NDs [70]. Among several kinds of non-coding RNAs, microRNAs (miRNAs) have been the most well-studied. Accumulating evidence indicates that several miRNAs are abnormally expressed in the serum, plasma, cerebrospinal fluid, cerebellum, pons, striatum, and frontal cortex of patients with MSA [18][71][72][73][74][75][76][77][78].

3.1. Molecular Mechanisms of miRNA Biogenesis

The function of miRNAs is basically to silence target gene expressions by binding to transcripts located mainly at the 3′-untranslated regions (3′-UTR), although there are also cases in which such silencing occurs by binding to transcripts in the 5′-UTR and coding region [79]. Clustered miRNAs can either be simultaneously transcribed from single polycistronic transcripts or independently transcribed. RNA polymerase II typically transcribes the primary miRNAs (pri-miRNAs), which are cleaved by a complex called a microprocessor consisting of an RNA-binding protein known as DiGeorge syndrome critical region 8 (DGCR8) and a ribonuclease III (RNase III) enzyme called Drosha [80]. DGCR8 recognizes an N6-methyladenylated GGAC and other motifs within the pri-miRNA, while Drosha cleaves the pri-miRNA duplex at the base of the characteristic hairpin structure of pri-miRNA [81]. The small, microprocessor-generated hairpin-shaped RNAs that form the 3′ overhang, called miRNA precursors (pre-miRNAs), are exported by exportin-5 in complex with RAN-GTP and are processed by a double-stranded RNase III enzyme termed Dicer, which is complexed with the trans-activation response RNA-binding protein (TRBP) [82]. The mature miRNA duplexes are loaded onto Argonaute (AGO) family proteins in an ATP-dependent manner to form an effector complex called the RNA-induced silencing complex (RISC) [83]. One strand of miRNA is then removed from the RISC to generate the mature RISC that induces gene silencing. In the earlier versions of miRBase, the removed strand of miRNA was called the “passenger” strand and represented as miRNA*. However, since it has been reported that miRNAs from the 5′ and 3′ arms of a pre-miRNA precursor both exist, these miRNAs are now represented as miRNA-5p and miRNA-3p, respectively. Post-transcriptional regulation by the RISC complex is mediated by incomplete base-paring of miRNA-mRNA interactions, likely due to the targeting of multiple transcripts, which contributes to the complexity or redundancy of miRNA systems. Recently, multiple non-canonical miRNA biogenesis pathways have been progressively identified, and have been grouped into Drosha/DGCR8-independent and Dicer-independent pathways [84] (Figure 3).
Figure 3. Canonical and non-canonical pathways of microRNA biogenesis.
In the canonical pathway, miRNA biogenesis begins with the transcription of primary miRNAs (pri-miRNAs), which a microprocessor composed of DGCR8 and Drosha cleaves to generate miRNA precursors (pre-miRNAs), which are hairpin forms of small RNA. Pre-miRNAs are exported by exportin-5 in complex with RAN-GTP and processed by Dicer, complexed with TRBP. In the Dicer-independent pathways, after the pre-miRNA, processed by DGCR8/Drosha, is exported, it is further processed via AGO-dependent cleavage but is not cleaved by Dicer. In the Drosha/DGCR8-independent pathway, miRNAs derived from snoRNA, tRNA, shRNA, splicing and others are exported without processing and then processed by Dicer. Through these processes, the mature miRNA is combined with members of the AGO family of proteins to form miRNA-induced silencing complexes (miRISCs).

3.2. Dysregulation of Genes and miRNAs in Patients with MSA

The first study of the miRNA profiles in MSA cases in comparison with controls and in transgenic (tg) models of MSA compared with non-tg mice was reported in 2014, much later than the corresponding studies in NDs such as AD, PD and ALS [18]. The screening of dysregulated miRNAs in both humans and animal models of MSA revealed that miR-96 is upregulated in these cases (probably miR-96-5p, since miR-96-3p was displayed as miR-96* in the earlier version of miRBase), resulting in downregulation of EAAC1 and a taurine transporter (TauT, also known as SLC6A6), another solute carrier family protein [18]. These proteins are particularly colocalized in neuronal and glial cells neighboring the α-syn-positive oligodendrocytes. Interestingly, researchers have shown that administration of an miR-96-5p inhibitor to the mouse brain improves neuroprotection against oxidative stress via upregulation of EAAC1 and the resulting increase in GSH levels [85][86]. In addition, several reports have shown that miR-96-5p expression is upregulated in the serum, striatum, and frontal cortex of MSA patients [18][87][88]. Moreover, the administration of FTY720-Mitoxy, which is known to increase the expression of neurotrophic factors in oligodendrocytes, reduced the expression of miR-96-5p in an MSA-mimicking mouse model [89]. These results suggest that agents that increase GSH levels in the brain and thereby manipulate miR-96-5p levels might be used to ameliorate the symptoms of MSA.
Another report has shown that both miR-339-5p and miR-96-5p are increased in the serum of patients with MSA [87]. In that study, miR-339-5p was shown to target strong mRNA-miRNA interactions with the RNA-binding protein neuro-oncological ventral antigen-1 (NOVA1). Interestingly, researchers have shown that NOVA1 regulates EAAC1 expression via glutamate transport-associated protein 3–18 (GTRAP3-18), which is an endoplasmic reticulum (ER)-localized protein and negative regulator of EAAC1, by trapping EAAC1 in the ER [86]. NOVA1 is also directly regulated by miR-96-5p, suggesting that the cysteine uptake system for GSH biosynthesis is impaired in patients with MSA.
There is another report that miR-202 (probably miR-202-3p, since miR-202-5p was displayed as miR-202* in the earlier version of miRBase) is upregulated in the cerebellums of patients with MSA, in correlation with reductions in the expression of the organic cation uptake transporter (Oct1, encoded by SLC22A1) [76]. Oct1 is known to regulate numerous target genes related to cellular stress. Deficiency in Oct1 expression results in hypersensitivity to hydrogen peroxide, which in turn elevates ROS levels. In the brain, Oct1 is expressed in the endothelial cells that form the blood–brain barrier (BBB) and may be involved in translocation of organic cations across the BBB in both directions [90]. The upregulation of miR-202-3p accompanied by a low level of Oct1 has been detected in the MSA cerebellum, which could result in decreased resistance of neurons to oxidative damage and subsequent cerebellar disease. Moreover, miR-202-3p has been reported to be linked to mitochondrial dysfunction, suggesting that increasing oxidative stress by miR-202-3p dysfunction contributes to the pathology of MSA [91].
Furthermore, miR-101 (probably miR-101-3p, since miR-101-5p was displayed as miR-101* in the earlier version of miRBase) has been observed to be upregulated in the striatum of MSA patients compared to controls [88]. Overexpression of miR-101-3p in an oligodendroglial cell line inhibited autophagy by altering the expression of autophagy-related genes that promote α-syn accumulation. In addition, researchers previously showed that EAAC1 expression is negatively regulated by miR-101-3p in a direct manner [85]. Interestingly, lentiviral delivery of an anti-miR-101-3p construct has been shown to reduce α-syn-induced autophagy deficits in a mouse model of MSA, indicating that anti-miR-101-3p is a promising therapeutic agent for MSA [88].

3.3. Candidate miRNA Biomarkers for MSA

Several studies have used miRNA microarrays to elucidate the profile of miRNA in patients with MSA, revealing the dysregulation of several miRNAs, some of which overlapped across the studies [92][93][94]. The most frequently reported miRNA to be dysregulated in MSA patients (6 reports) was miR-24-3p, which has also been reported to be dysregulated in AD and PD [95][96]. It has also been shown that inhibition of the miR-24-3p “sponge”, CircRtn4—one of the CircularRNAs, decreased ROS levels concomitant with increases in SOD and GSH levels [97]. In addition, miR-24-3p was also detected as a possible regulator of GPx3 in response to oxidative stress and related pathologies [98]. Four independent miRNA microarrays revealed several dysregulated miRNAs in MSA patients: miR-19b-3p, miR-25-3p and miR-92a-3p [18][71][72][73][74][78]. miR-19b-3p has been reported as a biomarker of PD in several studies [99][100]. Interestingly, both miR-25-3p and miR-19b-3p have been detected in the serum of patients with an isolated form of REM sleep behavior disorder, representing the prodromal state of the α-synucleinopathies, suggesting that these miRNAs could be potential prognostic biomarkers for α-synucleinopathies [101]. miR-92a-3p has been reported as a dysregulated miRNA in ALS [102][103]. miR-92a-3p has repeatedly been reported in connection to oxidative stress and could be a regulator of GPx3 [98][104]. Further investigation will be needed to clarify the functions of these miRNAs and whether they might have potential as either biomarkers or therapeutic agents for MSA.

This entry is adapted from the peer-reviewed paper 10.3390/ijms232315076

References

  1. Fanciulli, A.; Wenning, G.K. Multiple-system atrophy. N. Engl. J. Med. 2015, 372, 249–263.
  2. Palma, J.A.; Norcliffe-Kaufmann, L.; Kaufmann, H. Diagnosis of multiple system atrophy. Auton. Neurosci. Basic Clin. 2018, 211, 15–25.
  3. Schweighauser, M.; Shi, Y.; Tarutani, A.; Kametani, F.; Murzin, A.G.; Ghetti, B.; Matsubara, T.; Tomita, T.; Ando, T.; Hasegawa, K.; et al. Structures of α-synuclein filaments from multiple system atrophy. Nature 2020, 585, 464–469.
  4. Jellinger, K.A. Multiple System Atrophy: An Oligodendroglioneural Synucleinopathy1. J. Alzheimer’s Dis. JAD 2018, 62, 1141–1179.
  5. Kaji, S.; Maki, T.; Ishimoto, T.; Yamakado, H.; Takahashi, R. Insights into the pathogenesis of multiple system atrophy: Focus on glial cytoplasmic inclusions. Transl. Neurodegener. 2020, 9, 7.
  6. Mészáros, L.; Hoffmann, A.; Wihan, J.; Winkler, J. Current Symptomatic and Disease-Modifying Treatments in Multiple System Atrophy. Int. J. Mol. Sci. 2020, 21, 2775.
  7. Marmion, D.J.; Peelaerts, W.; Kordower, J.H. A historical review of multiple system atrophy with a critical appraisal of cellular and animal models. J. Neural Transm. 2021, 128, 1507–1527.
  8. Watanabe, H.; Saito, Y.; Terao, S.; Ando, T.; Kachi, T.; Mukai, E.; Aiba, I.; Abe, Y.; Tamakoshi, A.; Doyu, M.; et al. Progression and prognosis in multiple system atrophy: An analysis of 230 Japanese patients. Brain A J. Neurol. 2002, 125, 1070–1083.
  9. Multiple-System Atrophy Research Collaboration. Mutations in COQ2 in Familial and Sporadic Multiple-System Atrophy. N. Engl. J. Med. 2013, 369, 233–244.
  10. Ogaki, K.; Fujioka, S.; Heckman, M.G.; Rayaprolu, S.; Soto-Ortolaza, A.I.; Labbé, C.; Walton, R.L.; Lorenzo-Betancor, O.; Wang, X.; Asmann, Y.; et al. Analysis of COQ2 gene in multiple system atrophy. Mol. Neurodegener. 2014, 9, 44.
  11. Quinzii, C.M.; Hirano, M.; DiMauro, S. Mutant COQ2 in multiple-system atrophy. N. Engl. J. Med. 2014, 371, 81–82.
  12. Lin, C.H.; Tan, E.K.; Yang, C.C.; Yi, Z.; Wu, R.M. COQ2 gene variants associate with cerebellar subtype of multiple system atrophy in Chinese. Mov. Disord. Off. J. Mov. Disord. Soc. 2015, 30, 436–437.
  13. Zhao, Q.; Yang, X.; Tian, S.; An, R.; Zheng, J.; Xu, Y. Association of the COQ2 V393A variant with risk of multiple system atrophy in East Asians: A case-control study and meta-analysis of the literature. Neurol. Sci. Off. J. Ital. Neurol. Soc. Ital. Soc. Clin. Neurophysiol. 2016, 37, 423–430.
  14. Pasanen, P.; Myllykangas, L.; Siitonen, M.; Raunio, A.; Kaakkola, S.; Lyytinen, J.; Tienari, P.J.; Pöyhönen, M.; Paetau, A. Novel α-synuclein mutation A53E associated with atypical multiple system atrophy and Parkinson’s disease-type pathology. Neurobiol. Aging 2014, 35, e2181–e2185.
  15. Danka Mohammed, C.P.; Park, J.S.; Nam, H.G.; Kim, K. MicroRNAs in brain aging. Mech. Ageing Dev. 2017, 168, 3–9.
  16. Kinoshita, C.; Aoyama, K.; Nakaki, T. microRNA as a new agent for regulating neuronal glutathione synthesis and metabolism. AIMS Mol. Sci. 2015, 2, 124–143.
  17. Kinoshita, C.; Kubota, N.; Aoyama, K. Interplay of RNA-Binding Proteins and microRNAs in Neurodegenerative Diseases. Int. J. Mol. Sci. 2021, 22, 5292.
  18. Ubhi, K.; Rockenstein, E.; Kragh, C.; Inglis, C.; Spencer, B.; Michael, S.; Mante, M.; Adame, A.; Galasko, D.; Masliah, E. Widespread microRNA dysregulation in multiple system atrophy—Disease-related alteration in miR-96. Eur. J. Neurosci. 2014, 39, 1026–1041.
  19. Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative Stress: A Key Modulator in Neurodegenerative Diseases. Molecules 2019, 24, 1583.
  20. Shukla, D.; Mandal, P.K.; Mishra, R.; Punjabi, K.; Dwivedi, D.; Tripathi, M.; Badhautia, V. Hippocampal Glutathione Depletion and pH Increment in Alzheimer’s Disease: An in vivo MRS Study. J. Alzheimer’s Dis. JAD 2021, 84, 1139–1152.
  21. Mandal, P.K.; Dwivedi, D.; Shukla, D.; Samkaria, A.; Roy, R.G.; Arora, Y.; Jindal, K. Interplay Between Hippocampal Glutathione Depletion and pH Increment in Alzheimer’s Disease. J. Alzheimer’s Dis. JAD 2022, 88, 1–6.
  22. Chiang, G.C.; Mao, X.; Kang, G.; Chang, E.; Pandya, S.; Vallabhajosula, S.; Isaacson, R.; Ravdin, L.D.; Shungu, D.C. Relationships among Cortical Glutathione Levels, Brain Amyloidosis, and Memory in Healthy Older Adults Investigated In Vivo with (1)H-MRS and Pittsburgh Compound-B PET. AJNR Am. J. Neuroradiol. 2017, 38, 1130–1137.
  23. Jenner, P. Presymptomatic detection of Parkinson’s disease. Journal of neural transmission. Supplementum 1993, 40, 23–36.
  24. Pearce, R.K.B.; Owen, A.; Daniel, S.; Jenner, P.; Marsden, C.D. Alterations in the distribution of glutathione in the substantia nigra in Parkinson’s disease. J. Neural Transm. 1997, 104, 661–677.
  25. Andronesi, O.C.; Nicholson, K.; Jafari-Khouzani, K.; Bogner, W.; Wang, J.; Chan, J.; Macklin, E.A.; Levine-Weinberg, M.; Breen, C.; Schwarzschild, M.A.; et al. Imaging Neurochemistry and Brain Structure Tracks Clinical Decline and Mechanisms of ALS in Patients. Front. Neurol. 2020, 11, 590573.
  26. Chen, J.J.; Thiyagarajah, M.; Song, J.; Chen, C.; Herrmann, N.; Gallagher, D.; Rapoport, M.J.; Black, S.E.; Ramirez, J.; Andreazza, A.C.; et al. Altered central and blood glutathione in Alzheimer’s disease and mild cognitive impairment: A meta-analysis. Alzheimer’s Res. Ther. 2022, 14, 23.
  27. Sofic, E.; Lange, K.W.; Jellinger, K.; Riederer, P. Reduced and oxidized glutathione in the substantia nigra of patients with Parkinson’s disease. Neurosci. Lett. 1992, 142, 128–130.
  28. Fanciulli, A.; Stankovic, I.; Krismer, F.; Seppi, K.; Levin, J.; Wenning, G.K. Multiple system atrophy. Int. Rev. Neurobiol. 2019, 149, 137–192.
  29. Rink, C.; Khanna, S. Significance of brain tissue oxygenation and the arachidonic acid cascade in stroke. Antioxid. Redox Signal. 2011, 14, 1889–1903.
  30. Bazinet, R.P.; Layé, S. Polyunsaturated fatty acids and their metabolites in brain function and disease. Nat. Rev. Neurosci. 2014, 15, 771–785.
  31. Aoyama, K. Glutathione in the Brain. Int. J. Mol. Sci. 2021, 22, 5010.
  32. Shukla, G.S.; Hussain, T.; Srivastava, R.S.; Chandra, S.V. Glutathione peroxidase and catalase in liver, kidney, testis and brain regions of rats following cadmium exposure and subsequent withdrawal. Ind. Health 1989, 27, 59–69.
  33. Szymonik-Lesiuk, S.; Czechowska, G.; Stryjecka-Zimmer, M.; SŁomka, M.; MĄldro, A.; CeliŃski, K.; Wielosz, M. Catalase, superoxide dismutase, and glutathione peroxidase activities in various rat tissues after carbon tetrachloride intoxication. J. Hepato-Biliary-Pancreat. Surg. 2003, 10, 309–315.
  34. Dringen, R.; Hirrlinger, J. Glutathione Pathways in the Brain. Biol. Chem. 2003, 384, 505–516.
  35. Couto, N.; Wood, J.; Barber, J. The role of glutathione reductase and related enzymes on cellular redox homoeostasis network. Free. Radic. Biol. Med. 2016, 95, 27–42.
  36. Fitzmaurice, P.S.; Ang, L.; Guttman, M.; Rajput, A.H.; Furukawa, Y.; Kish, S.J. Nigral glutathione deficiency is not specific for idiopathic Parkinson’s disease. Mov. Disord. 2003, 18, 969–976.
  37. Jenner, P.; Dexter, D.T.; Sian, J.; Schapira, A.H.V.; Marsden, C.D. Oxidative stress as a cause of nigral cell death in Parkinson’s disease and incidental lewy body disease. Ann. Neurol. 1992, 32, S82–S87.
  38. Sian, J.; Dexter, D.T.; Lees, A.J.; Daniel, S.; Agid, Y.; Javoy-Agid, F.; Jenner, P.; Marsden, C.D. Alterations in glutathione levels in Parkinson’s disease and other neurodegenerative disorders affecting basal ganglia. Ann. Neurol. 1994, 36, 348–355.
  39. Paik, S.R.; Lee, D.; Cho, H.-J.; Lee, E.-N.; Chang, C.-S. Oxidized glutathione stimulated the amyloid formation of α-synuclein. FEBS Lett. 2003, 537, 63–67.
  40. Xu, B.; Wu, S.-W.; Lu, C.-W.; Deng, Y.; Liu, W.; Wei, Y.-G.; Yang, T.-Y.; Xu, Z.-F. Oxidative stress involvement in manganese-induced alpha-synuclein oligomerization in organotypic brain slice cultures. Toxicology 2013, 305, 71–78.
  41. Tanaka, K.-I.; Sonoda, K.; Asanuma, M. Effect of Alteration of Glutathione Content on Cell Viability in α-Synuclein-Transfected SH-SY5Y Cells. Adv. Park. Dis. 2017, 6, 93.
  42. Clark, J.; Clore, E.L.; Zheng, K.; Adame, A.; Masliah, E.; Simon, D.K. Oral N-acetyl-cysteine attenuates loss of dopaminergic terminals in alpha-synuclein overexpressing mice. PLoS ONE 2010, 5, e12333.
  43. Flohé, L. Glutathione peroxidase. Basic Life Sci. 1988, 49, 663–668.
  44. Kish, S.J.; Morito, C.L.; Hornykiewicz, O. Brain glutathione peroxidase in neurodegenerative disorders. Neurochem. Pathol. 1986, 4, 23–28.
  45. Koo, H.J.; Yang, J.E.; Park, J.H.; Lee, D.; Paik, S.R. α-Synuclein-mediated defense against oxidative stress via modulation of glutathione peroxidase. Biochim. Et Biophys. Acta 2013, 1834, 972–976.
  46. Hayes, J.D.; Flanagan, J.U.; Jowsey, I.R. Glutathione transferases. Annu. Rev. Pharmacol. Toxicol. 2005, 45, 51–88.
  47. Probst-Cousin, S.; Bergmann, M.; Kuchelmeister, K.; Schröder, R.; Schmid, K.W. Ubiquitin-positive inclusions in different types of multiple system atrophy: Distribution and specificity. Pathol. Res. Pract. 1996, 192, 453–461.
  48. Aoyama, K.; Nakaki, T. Impaired glutathione synthesis in neurodegeneration. Int. J. Mol. Sci. 2013, 14, 21021–21044.
  49. Janáky, R.; Varga, V.; Hermann, A.; Saransaari, P.; Oja, S.S. Mechanisms of L-cysteine neurotoxicity. Neurochem. Res. 2000, 25, 1397–1405.
  50. Paul, B.D.; Sbodio, J.I.; Snyder, S.H. Cysteine Metabolism in Neuronal Redox Homeostasis. Trends Pharmacol. Sci. 2018, 39, 513–524.
  51. Hughes, C.E.; Coody, T.K.; Jeong, M.Y.; Berg, J.A.; Winge, D.R.; Hughes, A.L. Cysteine Toxicity Drives Age-Related Mitochondrial Decline by Altering Iron Homeostasis. Cell 2020, 180, 296–310.e218.
  52. Richman, P.G.; Meister, A. Regulation of gamma-glutamyl-cysteine synthetase by nonallosteric feedback inhibition by glutathione. J. Biol. Chem. 1975, 250, 1422–1426.
  53. Lu, S.C. Regulation of glutathione synthesis. Mol. Asp. Med. 2009, 30, 42–59.
  54. Ferguson, G.; Bridge, W. Glutamate cysteine ligase and the age-related decline in cellular glutathione: The therapeutic potential of γ-glutamylcysteine. Arch. Biochem. Biophys. 2016, 593, 12–23.
  55. Njålsson, R. Glutathione synthetase deficiency. Cell. Mol. Life Sci. CMLS 2005, 62, 1938–1945.
  56. Aoyama, K.; Nakaki, T. Glutathione in Cellular Redox Homeostasis: Association with the Excitatory Amino Acid Carrier 1 (EAAC1). Molecules 2015, 20, 8742–8758.
  57. Divito, C.B.; Underhill, S.M. Excitatory amino acid transporters: Roles in glutamatergic neurotransmission. Neurochem. Int. 2014, 73, 172–180.
  58. Bianchi, M.G.; Bardelli, D.; Chiu, M.; Bussolati, O. Changes in the expression of the glutamate transporter EAAT3/EAAC1 in health and disease. Cell. Mol. Life Sci. 2014, 71, 2001–2015.
  59. Aoyama, K.; Suh, S.W.; Hamby, A.M.; Liu, J.; Chan, W.Y.; Chen, Y.; Swanson, R.A. Neuronal glutathione deficiency and age-dependent neurodegeneration in the EAAC1 deficient mouse. Nat. Neurosci. 2006, 9, 119–126.
  60. Kanai, Y.; Hediger, M.A. The glutamate and neutral amino acid transporter family: Physiological and pharmacological implications. Eur. J. Pharmacol. 2003, 479, 237–247.
  61. Watts, S.D.; Torres-Salazar, D.; Divito, C.B.; Amara, S.G. Cysteine Transport through Excitatory Amino Acid Transporter 3 (EAAT3). PLoS ONE 2014, 9, e109245.
  62. Sedláčková, L.; Laššuthová, P.; Štěrbová, K.; Vlčková, M.; Kudr, M.; Buksakowska, I.; Staněk, D.; Seeman, P. Severe neurodevelopmental disorder with intractable seizures due to a novel SLC1A4 homozygous variant. Eur. J. Med. Genet. 2021, 64, 104263.
  63. Abdelrahman, H.A.; Al-Shamsi, A.; John, A.; Ali, B.R.; Al-Gazali, L. A Novel SLC1A4 Mutation (p.Y191*) Causes Spastic Tetraplegia, Thin Corpus Callosum, and Progressive Microcephaly (SPATCCM) With Seizure Disorder. Child Neurol. Open 2019, 6, 2329048x19880647.
  64. Pironti, E.; Salpietro, V.; Cucinotta, F.; Granata, F.; Mormina, E.; Efthymiou, S.; Scuderi, C.; Gagliano, A.; Houlden, H.; Di Rosa, G. A novel SLC1A4 homozygous mutation causing congenital microcephaly, epileptic encephalopathy and spastic tetraparesis: A video-EEG and tractography—Case study. J. Neurogenet. 2018, 32, 316–321.
  65. Heimer, G.; Marek-Yagel, D.; Eyal, E.; Barel, O.; Oz Levi, D.; Hoffmann, C.; Ruzzo, E.K.; Ganelin-Cohen, E.; Lancet, D.; Pras, E.; et al. SLC1A4 mutations cause a novel disorder of intellectual disability, progressive microcephaly, spasticity and thin corpus callosum. Clin. Genet. 2015, 88, 327–335.
  66. Deng, X.; Sagata, N.; Takeuchi, N.; Tanaka, M.; Ninomiya, H.; Iwata, N.; Ozaki, N.; Shibata, H.; Fukumaki, Y. Association study of polymorphisms in the neutral amino acid transporter genes SLC1A4, SLC1A5 and the glycine transporter genes SLC6A5, SLC6A9 with schizophrenia. BMC Psychiatry 2008, 8, 58.
  67. Damseh, N.; Simonin, A.; Jalas, C.; Picoraro, J.A.; Shaag, A.; Cho, M.T.; Yaacov, B.; Neidich, J.; Al-Ashhab, M.; Juusola, J.; et al. Mutations in SLC1A4, encoding the brain serine transporter, are associated with developmental delay, microcephaly and hypomyelination. J. Med. Genet. 2015, 52, 541–547.
  68. Soma, H.; Yabe, I.; Takei, A.; Fujiki, N.; Yanagihara, T.; Sasaki, H. Associations between multiple system atrophy and polymorphisms of SLC1A4, SQSTM1, and EIF4EBP1 genes. Mov. Disord. Off. J. Mov. Disord. Soc. 2008, 23, 1161–1167.
  69. Kinoshita, C.; Aoyama, K. The Role of Non-Coding RNAs in the Neuroprotective Effects of Glutathione. Int. J. Mol. Sci. 2021, 22, 4245.
  70. Pérez-Soriano, A.; Bravo, P.; Soto, M.; Infante, J.; Fernández, M.; Valldeoriola, F.; Muñoz, E.; Compta, Y.; Tolosa, E.; Garrido, A.; et al. MicroRNA Deregulation in Blood Serum Identifies Multiple System Atrophy Altered Pathways. Mov. Disord. Off. J. Mov. Disord. Soc. 2020, 35, 1873–1879.
  71. Kume, K.; Iwama, H.; Deguchi, K.; Ikeda, K.; Takata, T.; Kokudo, Y.; Kamada, M.; Fujikawa, K.; Hirose, K.; Masugata, H.; et al. Serum microRNA expression profiling in patients with multiple system atrophy. Mol. Med. Rep. 2018, 17, 852–860.
  72. Uwatoko, H.; Hama, Y.; Iwata, I.T.; Shirai, S.; Matsushima, M.; Yabe, I.; Utsumi, J.; Sasaki, H. Identification of plasma microRNA expression changes in multiple system atrophy and Parkinson’s disease. Mol. Brain 2019, 12, 49.
  73. Kim, T.; Valera, E.; Desplats, P. Alterations in Striatal microRNA-mRNA Networks Contribute to Neuroinflammation in Multiple System Atrophy. Mol. Neurobiol. 2019, 56, 7003–7021.
  74. Wakabayashi, K.; Mori, F.; Kakita, A.; Takahashi, H.; Tanaka, S.; Utsumi, J.; Sasaki, H. MicroRNA expression profiles of multiple system atrophy from formalin-fixed paraffin-embedded samples. Neurosci. Lett. 2016, 635, 117–122.
  75. Lee, S.-T.; Chu, K.; Jung, K.-H.; Ban, J.-J.; Im, W.-S.; Jo, H.-Y.; Park, J.-H.; Lim, J.-Y.; Shin, J.-W.; Moon, J.; et al. Altered Expression of miR-202 in Cerebellum of Multiple-System Atrophy. Mol. Neurobiol. 2015, 51, 180–186.
  76. Vallelunga, A.; Ragusa, M.; Di Mauro, S.; Iannitti, T.; Pilleri, M.; Biundo, R.; Weis, L.; Di Pietro, C.; De Iuliis, A.; Nicoletti, A.; et al. Identification of circulating microRNAs for the differential diagnosis of Parkinson’s disease and Multiple System Atrophy. Front. Cell. Neurosci. 2014, 8, 156.
  77. Starhof, C.; Hejl, A.M.; Heegaard, N.H.H.; Carlsen, A.L.; Burton, M.; Lilje, B.; Winge, K. The biomarker potential of cell-free microRNA from cerebrospinal fluid in Parkinsonian Syndromes. Mov. Disord. Off. J. Mov. Disord. Soc. 2019, 34, 246–254.
  78. Seok, H.; Ham, J.; Jang, E.S.; Chi, S.W. MicroRNA Target Recognition: Insights from Transcriptome-Wide Non-Canonical Interactions. Mol. Cells 2016, 39, 375–381.
  79. Krol, J.; Loedige, I.; Filipowicz, W. The widespread regulation of microRNA biogenesis, function and decay. Nat. Rev. Genet. 2010, 11, 597–610.
  80. Alarcón, C.R.; Lee, H.; Goodarzi, H.; Halberg, N.; Tavazoie, S.F. N6-methyladenosine marks primary microRNAs for processing. Nature 2015, 519, 482–485.
  81. Michlewski, G.; Cáceres, J.F. Post-transcriptional control of miRNA biogenesis. RNA 2019, 25, 1–16.
  82. Iwakawa, H.O.; Tomari, Y. Life of RISC: Formation, action, and degradation of RNA-induced silencing complex. Mol. Cell 2022, 82, 30–43.
  83. Stavast, C.J.; Erkeland, S.J. The Non-Canonical Aspects of MicroRNAs: Many Roads to Gene Regulation. Cells 2019, 8, 1465.
  84. Kinoshita, C.; Aoyama, K.; Matsumura, N.; Kikuchi-Utsumi, K.; Watabe, M.; Nakaki, T. Rhythmic oscillations of the microRNA miR-96-5p play a neuroprotective role by indirectly regulating glutathione levels. Nat. Commun. 2014, 5, 3823.
  85. Kinoshita, C.; Kikuchi-Utsumi, K.; Aoyama, K.; Suzuki, R.; Okamoto, Y.; Matsumura, N.; Omata, D.; Maruyama, K.; Nakaki, T. Inhibition of miR-96-5p in the mouse brain increases glutathione levels by altering NOVA1 expression. Commun. Biol. 2021, 4, 182.
  86. Vallelunga, A.; Iannitti, T.; Capece, S.; Somma, G.; Russillo, M.C.; Foubert-Samier, A.; Laurens, B.; Sibon, I.; Meissner, W.G.; Barone, P.; et al. Serum miR-96-5P and miR-339-5P Are Potential Biomarkers for Multiple System Atrophy and Parkinson’s Disease. Front. Aging Neurosci. 2021, 13, 632891.
  87. Valera, E.; Spencer, B.; Mott, J.; Trejo, M.; Adame, A.; Mante, M.; Rockenstein, E.; Troncoso, J.C.; Beach, T.G.; Masliah, E.; et al. MicroRNA-101 Modulates Autophagy and Oligodendroglial Alpha-Synuclein Accumulation in Multiple System Atrophy. Front. Mol. Neurosci. 2017, 10, 329.
  88. Vidal-Martinez, G.; Segura-Ulate, I.; Yang, B.; Diaz-Pacheco, V.; Barragan, J.A.; De-Leon Esquivel, J.; Chaparro, S.A.; Vargas-Medrano, J.; Perez, R.G. FTY720-Mitoxy reduces synucleinopathy and neuroinflammation, restores behavior and mitochondria function, and increases GDNF expression in Multiple System Atrophy mouse models. Exp. Neurol. 2020, 325, 113120.
  89. Koepsell, H. General Overview of Organic Cation Transporters in Brain. In Organic Cation Transporters in the Central Nervous System; Daws, L.C., Ed.; Springer International Publishing: Cham, Switzerland, 2021; pp. 1–39.
  90. Baumgart, B.R.; Gray, K.L.; Woicke, J.; Bunch, R.T.; Sanderson, T.P.; Van Vleet, T.R. MicroRNA as biomarkers of mitochondrial toxicity. Toxicol. Appl. Pharmacol. 2016, 312, 26–33.
  91. Chang, L.; Xia, J. MicroRNA Regulatory Network Analysis Using miRNet 2.0. In Transcription Factor Regulatory Networks; Song, Q., Tao, Z., Eds.; Springer US: New York, NY, USA, 2022; pp. 185–204.
  92. Huang, Z.; Shi, J.; Gao, Y.; Cui, C.; Zhang, S.; Li, J.; Zhou, Y.; Cui, Q. HMDD v3.0: A database for experimentally supported human microRNA-disease associations. Nucleic Acids Res. 2019, 47, D1013–D1017.
  93. Liu, L.; Liu, L.; Lu, Y.; Zhang, T.; Zhao, W. Serum aberrant expression of miR-24-3p and its diagnostic value in Alzheimer’s disease. Biomark. Med. 2021, 15, 1499–1507.
  94. Chen, Y.; Wang, X. miRDB: An online database for prediction of functional microRNA targets. Nucleic acids research 2019, 48, D127–D131.
  95. Yousefi, M.; Peymani, M.; Ghaedi, K.; Irani, S.; Etemadifar, M. Significant modulations of linc001128 and linc0938 with miR-24-3p and miR-30c-5p in Parkinson disease. Sci. Rep. 2022, 12, 2569.
  96. Di, G.; Yang, X.; Cheng, F.; Liu, H.; Xu, M. CEBPA-AS1 Knockdown Alleviates Oxygen-Glucose Deprivation/Reperfusion-Induced Neuron Cell Damage by the MicroRNA 24-3p/BOK Axis. Mol. Cell. Biol. 2021, 41, e0006521.
  97. Matoušková, P.; Hanousková, B.; Skálová, L. MicroRNAs as Potential Regulators of Glutathione Peroxidases Expression and Their Role in Obesity and Related Pathologies. Int. J. Mol. Sci. 2018, 19, 1199.
  98. Chis, A.R.; Moatar, A.I.; Dijmarescu, C.; Rosca, C.; Vorovenci, R.J.; Krabbendam, I.; Dolga, A.; Bejinar, C.; Marian, C.; Sirbu, I.O.; et al. Plasma hsa-mir-19b is a potential LevoDopa therapy marker. J. Cell. Mol. Med. 2021, 25, 8715–8724.
  99. Cao, X.Y.; Lu, J.M.; Zhao, Z.Q.; Li, M.C.; Lu, T.; An, X.S.; Xue, L.J. MicroRNA biomarkers of Parkinson’s disease in serum exosome-like microvesicles. Neurosci. Lett. 2017, 644, 94–99.
  100. Soto, M.; Iranzo, A.; Lahoz, S.; Fernández, M.; Serradell, M.; Gaig, C.; Melón, P.; Martí, M.J.; Santamaría, J.; Camps, J.; et al. Serum MicroRNAs Predict Isolated Rapid Eye Movement Sleep Behavior Disorder and Lewy Body Diseases. Mov. Disord. Off. J. Mov. Disord. Soc. 2022, 37, 2086–2098.
  101. Joilin, G.; Gray, E.; Thompson, A.G.; Bobeva, Y.; Talbot, K.; Weishaupt, J.; Ludolph, A.; Malaspina, A.; Leigh, P.N.; Newbury, S.F.; et al. Identification of a potential non-coding RNA biomarker signature for amyotrophic lateral sclerosis. Brain Commun. 2020, 2, fcaa053.
  102. Arakawa, Y.; Itoh, S.; Fukazawa, Y.; Ishiguchi, H.; Kohmoto, J.; Hironishi, M.; Ito, H.; Kihira, T. Association between oxidative stress and microRNA expression pattern of ALS patients in the high-incidence area of the Kii Peninsula. Brain Res. 2020, 1746, 147035.
  103. Zhou, Y.; Wei, W.; Shen, J.; Lu, L.; Lu, T.; Wang, H.; Xue, X. Alisol A 24-acetate protects oxygen-glucose deprivation-induced brain microvascular endothelial cells against apoptosis through miR-92a-3p inhibition by targeting the B-cell lymphoma-2 gene. Pharm. Biol. 2021, 59, 513–524.
  104. Eisele, Y.S.; Monteiro, C.; Fearns, C.; Encalada, S.E.; Wiseman, R.L.; Powers, E.T.; Kelly, J.W. Targeting protein aggregation for the treatment of degenerative diseases. Nat. Rev. Drug Discov. 2015, 14, 759–780.
More
This entry is offline, you can click here to edit this entry!
Video Production Service