Abstract
Hydrogen sulfide (H2S) and hydrogen polysulfides are recognized as important signaling molecules that are generated physiologically in the body, including the central nervous system (CNS). Studies have shown that these two molecules are involved in cytoprotection against oxidative stress and inflammatory response. In the brain system, H2S and polysulfides exert multiple functions in both health and diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington's disease (HD), memory decline, and glioma. Mechanistically, S-Persulfidation (also known as S-sulfuration or S-sulfhydration) of target proteins is believed to be a fundamental mechanism that underlies H2S-regulated signaling pathways. Cysteine S-Persulfidation is an important paradigm of post translational protein modification in the process of H2S signaling. This model is established as a critical redox mechanism to regulate numerous biological functions, especially in H2S-mediated neuroprotection and neurogenesis. Although the current research of S-Persulfidation is still in its infancy, accumulative evidence suggests that protein S-Persulfidation may share similar characteristics with protein S-nitrosylation. In this review, we will provide a comprehensive insight into the S-Persulfidation biology of H2S and polysulfides in neurological ailments and presume potential avenues for therapeutic development in these disorders based on S-Persulfidation of target proteins.
Keywords: Hydrogen sulfide, polysulfides, cysteine, S-Persulfidation, neurodegeneration, central nervous system
1. INTRODUCTION
Hydrogen sulfide (H2S) is previously known as an environmental hazard with a rotten egg smell [1]. However, mounting evidence suggests that H2S, independently of any transporters, confers a diversity of physiological actions on various systems [2-7], including the central nervous system (CNS) [8-11]. H2S is distinctly expressed in mammalian tissues where it could freely cross through the cell membranes [12]. It has now been well accepted that H2S acts as the third gaseous signal molecule in conjunction with nitric oxide (NO) and carbon monoxide (CO) [13]. As a gasotransmitter, H2S is taken as a key regulator in a wide spectrum of physiological and pathological processes in the brain tissues [10]. Studies of H2S in the CNS were initiated by the discovery of sulfides in the brain [14]. In this study, the authors demonstrated that inhalation of H2S led to increased brain sulfide deposition; this may be associated with the mortality in rats [14]. After that, H2S biosynthesis is detected in the brain, and H2S is found to facilitate long-term hippocampal poten-tiation (LTP) by raising N-methyl-D-aspartic acid receptors (NMDARs)-induced responses [15]. Besides, it has been unveiled that H2S could directly modulate pH homeostasis and intracellular Ca2+ release in microglial cells, neurons, and astrocytes [16-20]. Most importantly, impaired H2S synthesis participates in the development of various neurological diseases, such as ischemic stroke, Alzheimer’s disease (AD), and Parkinson’s disease (PD) [9].
Polysulfides are H2S-derived endogenous molecules with a distinct number of inner sulfur atoms [21]. In brief, H2S is sequentially oxidized to polysulfides until the number of sulfur atoms reaches eight, and the sulfur molecules cyclize and separate from polysulfides [22-24]. In comparison with H2S, polysulfides grant a greater potency toward ion channels, transcription factors, or tumor suppressors [1]. Similar to the effects of H2S, recent studies have demonstrated that polysulfides also exhibit neuroprotective effects by sulfurating the target proteins, such as Kelch-like ECH-associating protein 1 (Keap1), transient receptor potential cation channel subfamily A member 1 (TRPA1) channels, and phosphatase and tensin homolog (PTEN), much more potently than H2S [18, 25]. The significance of polysulfides in the regulation of neurobiology has been gradually recognized, especially their roles in S-Persulfidation of target protein cysteine sites [26, 27]. The additional sulfur of polysulfides could be incorporated into the cysteine residues, termed as S-Persulfidation, an important post-translational modification [25, 28]. For example, polysulfides-induced S-Persulfidation of parkin, a neuroprotective ubiquitin E3 ligase, is remarkably depleted in the brain tissues of subjects suffering from with PD, hinting that this collapse might play a critical role in the pathophysiology of PD [29].
S-Persulfidation, a chemical modification, is characterized by adding sulfur atoms to specific cysteine residues of target proteins, thus generating persulfide adducts on both small molecules and proteins [30, 31]. This process is considered to be a critical step for the biological functions of reactive sulfur species, including H2S and polysulfides [32]. Similar to S-nitrosation, S-Persulfidation of target proteins could be reversed by the thioredoxin system [26, 32], which is closely linked to various neurological diseases [32, 33]. In this regard, we will summarize the current studies of protein S-Persulfidation induced by H2S and polysulfides in the CNS, and discuss the advanced mechanistic concepts that underpin the signaling events of protein S-Persulfidation in neurodegenerative disorders.
2. PRODUCTION OF H2S AND POLYSULFIDES IN THE CNS
H2S, a weak acid, is slightly dissolved in water and easily dissociated into H+, HS−, and S2− [15]. It is estimated that less than 20% of H2S exists as H2S and the remaining 80% as HS− and S2− under physiological conditions [11]. The expression of H2S in mammalian brain tissues was first detected in 1989 [14, 34, 35]. Compelling evidence has demonstrated that H2S biosynthesis is mainly regulated by cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE) or 3-mercaptopyruvate sulfurtransferase (3-MST) together with cysteine aminotransferase (CAT) [3, 21, 36-40]. Excepting the classic H2S-generating pathway, D-cysteine can be metabolized by D-amino acid oxidase (DAO) to 3-mercaptopyruvate (3-MP), which is decomposed to H2S by 3-MST [41]. The fourth pathway is predominantly observed in the kidney and cerebellum, where D-cysteine-derived H2S protects the kidney tissues from the ischemia-reperfusion injury and cerebellar neurons from oxidative stress [42-45]. In the embryonic brain, the expression of CBS is low, and it will increase significantly from the prenatal period to the early postpartum period, and then reduces in the adult brain tissues [46]. The expressions of CBS are ubiquitously localized to the radial glia/astrocyte lineage, and CBS is indispensable for the differentiation of glial cells and astrocytes during brain development [46]. In the presence of various insults, such as ischemia-reperfusion injury or oxidative stress, the elevated CBS and H2S in astrocytes may contribute to the recovery of injured neurons [46-49]. It is likely that the levels of CSE in the brain are very low; thus its contribution to neuronal H2S production might be extremely minimal [50]. As CSE is not observed in the brain tissues, CBS seems to be the H2S-generating enzyme in the brain tissues. Nevertheless, the production of H2S is still detectable in the brain tissues from mice with a deficiency of CBS [51, 52]. This leads to a third pathway that the brain H2S production can be regulated by 3-MST along with CAT [53, 54]. 3-MST is found to be expressed in pyramidal neurons in the cerebral cortex, Purkinje cells in the cerebellum, mitral cells in the olfactory bulb, and also in the hippocampus and retinal neurons [55, 56]. Although 3-MST is localized in both mitochondria and cytosol, the 3-MST/CAT pathway might mainly give rise to H2S in the mitochondria [57, 58]. Interestingly, overexpression of 3-MST along with CAT may noticeably yield more bound sulfane sulfur, a cellular storage form of H2S, but not increased in neuron cells expressing functionally defective mutant enzymes [55]. In addition, CBS overexpression only slightly raises the content of bound sulfane sulfur [55, 59]. These findings suggest that the 3-MST/CAT pathway rather than CBS may be a major resource for H2S production in the brain. These published papers provide a novel perspective on the modulation of brain H2S generation.
As to the metabolic ways of H2S, it has been recommended that the fate of H2S’s catabolism may comprise several pathways, including methylation to methanethiol and dimethyl sulfide, reactions with metalloor cysteine-containing proteins in cells [60], and oxidation to sulfate (Fig. 1) [11, 61]. As aforementioned, H2S could also be stored as bound sulfane sulfur in cells, such as polysulfides and persulfides [18, 59]. However, this may be taken as an internal storage of H2S rather than its metabolic catabolism. Compared with the biosynthesis of H2S, the metabolic approaches of this gaseous mediator are not fully understood yet. Therefore, further validation is required to examine the exact metabolic turnover of H2S in experimental settings of physiological and pathophysiological conditions.
Fig. (1).
The metabolic turnover of H2S. The endogenous synthesis of H2S is synthesized by CBS, CSE, 3-MST, D-cysteine with DAO. In the mitochondria, H2S could be oxidized to persulfides by sulfide quinone oxidoreductase (SQR). The persulfides is then oxidized to sulfite (H2SO3) using sulfur dioxygenase, and H2SO3 is then metabolized to H2S2O3 with the aid of rhodanese. The appropriate production and clearance of H2S is essential for normal cellular functions. CBS, cystathionine β-synthase; CSE, cystathionine γ-lyase; 3-MST, 3-mercaptopyruvatesulfurtransferase; CAT, cysteine aminotransferase; DAO, D-amino acidoxidase; 3-mercapto pyruvate, 3-MP; SQR, sulfide quinone oxidoreductase. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
Polysulfides, novel H2S-derived molecules, are generated in mammal cells via either enzymatic or non-enzymatic pathways [21]. Besides, polysulfides could also be formed by the chemical interactions between H2S and NO [62-64]. However, this crosstalk between H2S and NO might be responsible for the production of polysulfides in mammalian tissues as such a reaction could occur under physiological conditions [63]. In the brain, oxidation of H2S-derived polysulfides is revealed to activate the TRPA1 channels approximately 300 times more potent than H2S in astrocytes [18]. Intriguingly, the same group also demonstrates that interactions of H2S with NO generate polysulfides to activate the TRPA1 channels [65]. This study offers a novel insight into the potential mechanisms for polysulfides production upon an interaction between H2S and NO. It is noted that the proportion of polysulfides produced by these pathways remains undefined, and the regulatory mechanism of polysulfides formation have yet to be fully elucidated. Therefore, a better understanding of polysulfide production and functions will certainly facilitate the therapeutic potential of H2S-related compounds in neurological diseases.
3. A BRIEF OVERVIEW OF PROTEIN S-PERSULFIDATION
S-Persulfidation (also termed as S-perthiolation or S-sulfhydration) is a modification of specific cysteine residues of target proteins by H2S or polysulfides [24]. In the process of S-Persulfidation, the thiols could be transformed to persulfides, whereby R may be small molecules or proteins [31]. More studies have revealed that S-Persulfidation is a crucial post-translational modification that contributes to H2S-mediated signal transduction in mammalian systems [44, 49]. In view of its chemistry characteristics, H2S is directly unable to react with protein cysteine residues to give rise to persulfides. However, persulfides could be generated by the interactions of H2S with oxidative cysteine residues, including disulfides, sulfenic acid, and nitrosothiol. Notably, the reactions of H2S occur majorly through its anion (HS-), the main form of H2S in aqueous solutions. The reactions between H2S and low molecular weight disulfides (including cysteine and glutathione disulfide) may produce mixture products in a slow and reversible manner [66]. It should be emphasized that the concentrations of protein disulfides and low molecular weight disulfides in the cytosol are quite low. As a result, the formation of polysulfides may be predominantly located in the endoplasmic reticulum rather than the cytosol [67].
A negative correlation is identified between cysteine residue activities and their acid dissociation constant (pKa) [23]. Typically, the S-Persulfidation sites are demonstrated to exist at cysteine residue sites with low pKa, albeit some S-Persulfidation modifications occur on cysteine residue sites with higher pKa [23]. In other words, the cysteine residue sites with low pKa seem to be more reactive with S-Persulfidation since they are present in the form of thiolate anions (S-) under physiological circumstances. However, the low pKa cysteines are more susceptible to oxidants such as hydrogen peroxide (H2O2) that are responsible for the formation of sulfenic acid (SOH), sulfinic acid (SO2H), or sulfonic acid (SO3H) derivatives [68-70]. Under the H2O2 challenge, the intracellular persulfide contents are upregulated; this rise could be prevented by inhibition of CBS or CSE [71]. In this process, the production of protein sulfenic acids (P-SOH) could be further oxidized to sulfinic acids (P-SO2H) and sulfonic acids (P-SO3H), which are irreversible oxidized products of the original protein cysteine adducts [72, 73]. S-Persulfidation of target proteins might induce the formation of P-SSO2H (perthiosulfinic) and P-SSO3H (perthiosulfonic) under an oxidative environment [71]. Both of them could be reduced to thiols, thereby recovering their physiological functions in cells [31, 74]. Polysulfide species (such as H2Sn or RSnSH) are important molecules that could deliver the sulfur atom to the specific cysteine residues, causing protein S-Persulfidation and consecutive signal transduction events [18, 67, 75]. Kimura and colleagues have suggested that the endogenous polysulfides are primarily generated from 3-MP by 3-MST [76]. The same group further demonstrated that the endogenous polysulfides could be produced from H2S by 3-MST and rhodanese [77]. Besides 3-MST, other sulfur transfer enzymes are also responsible for the production of endogenous persulfide species; such enzymes include sulfide quinone oxidoreductase (SQR), CSE, CBS, and cysteine desulfurase [31, 78, 79]. The cysteine could also be catalyzed by prokaryotic and mammalian cysteinyl-tRNA synthetases (CARSs), contributing to the formation of cysteine persulfide and polysulfides [80]. The production of polysulfides by CARSs could act as central mediators to induce endogenous protein S-Persulfidation.
In addition to S-Persulfidation, the cysteine residues might undergo other modifications, such as glutathionylation, palmitoylation, and nitrosylation. Therefore, it is of utmost importance to effectively distinguish the distinct post-translational modifications. Despite that, it is still a challenge to differentiate the persulfide group from the thiol group due to their analogous reactivity. There are several strategies for the detection of S-Persulfidation, such as modified biotin switch assay, maleimide assay, tag-switch method, or mass spectrometry assay, and these methods are well-reviewed [1, 6, 12, 50, 81-83]. As of yet, the specificity and sensitivity of the currently available methods for detecting S-Persulfidation might be questionable. Hence, it is needed to develop more novel and specific approaches for the exact identification of S-Persulfidation. To date, it is highly possible that a combination of the currently available methods, together with mass spectrometry may provide accurate avenues for the detection of target protein S-Persulfidation.
4. S-PERSULFIDATION BY H2S IN THE CNS
A variety of high-quality review papers have documented that H2S protects against neurologic disorders, including traumatic brain injury, ischemia-reperfusion injury, stroke, AD, PD, Huntington’s disease (HD), and Down syndrome [84-91]. It is expected that H2S therapy might become a potential treatment regimen in the near future on the basis of pre-clinic and clinical studies. The favorable effects of H2S on neurological diseases might be dependent on various mechanisms involving anti-oxidative, anti-inflammatory and anti-apoptotic effects, inhibition of endoplasmic reticulum stress and calcium overload [50]. Also, S-Persulfidation is recommended as an important model of action of H2S where H2S adds a sulfur atom to the cysteine residues of target proteins, which is being employed to investigate H2S-mediated signaling pathways in various brain disorders [82]. Therefore, we will next focus on the roles of H2S-mediated S-Persulfidation of target proteins in neuropathology.
4.1. S-Persulfidation by H2S in PD
PD, a neurodegenerative disorder in the middle-elderly population, is manifested by motor system abnormalities [92, 93]. A growing number of studies have identified that the pathologies of PD involve dopamine neuron degeneration in the substantia nigra, early reduction of dopaminergic uptake in the frontal lobes, the cholinergic disturbance in both brainstem and corticostriatal pathways [94-97]. However, to date, there is no consensus on the etiopathogenesis of PD [98]. The ambiguous pathogenesis of PD leads to limited clinical treatment options, thus causing immeasurable socioeconomic burdens and family suffering [99, 100]. Therefore, it is urgent to investigate the potential mechanisms that underlie the evolution of PD, and it is essential to establish more standardized management of PD.
Accumulating lines of evidence demonstrate a favorable role of H2S in the pathologies of PD, suggesting that H2S may be a new frontier for the treatment of PD [8, 81, 101, 102]. The endogenous levels of H2S are found to be downregulated in the substantia nigra from 6-hydroxydopamine (6-OHDA)-induced PD rats or 1-methy-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice [102, 103]. The decreased H2S production may be ascribed to the diminished H2S generating enzyme, CBS, in the substantia nigra of PD models [102]. Evidence for H2S-mediated therapies against PD is achieved from the exogenous administration of H2S-releasing donors [8, 81, 101, 102, 104-108]. Multiple mechanisms are speculated to be responsible for the therapeutic roles of H2S in PD [107, 109, 110]. Among which, dysregulation of protein S-Persulfidation contributes to the pathogenesis of PD [12, 81]. As such, treatment with H2S donors may be beneficial in the treatment of PD by stimulating the transsulfuration pattern [8]. As an E3 ligase, parkin is able to ubiquitinate the target proteins for proteasome-induced degradation; mutations of parkin lead to dopaminergic cell death during the progression of PD [111, 112]. Snyder and coworkers have confirmed that S-Persulfidated parkin is remarkably depleted in the brain tissues from PD patients [29], implying that disrupted parkin activities might be pathogenic in the development of PD. Its S-Persulfidation is associated with its normal catalytic activity, whereas nitrosylated parkin impairs its activity [29, 113]. It can be proposed that the restored S-Persulfidation of parkin by H2S enhances its catalytic activity, thus exerting neuroprotective effects against PD (Fig. 2) [29]. The protein p66Shc acts as a critical modulator in mitochondrial redox signaling, and its dysregulation is involved in the pathophysiology of PD [114-116]. Our group found that H2S inhibited overproduction of mitochondrial reactive oxygen species (ROS) in H2O2/D-galactose-incubated SH-SY5Y cells by S-Persulfidation of p66Shc as mutation of cystein-59 within p66Shc abolished the antagonistic effects of H2S on mitochondrial ROS production [117]. Given the importance of oxidative stress in the evolution of PD [118], our results suggest that p66Shc S-Persulfidation mediates the antioxidant actions of H2S, thus protecting against the development of PD. Although the roles of H2S-mediated protein S-Persulfidation in the pathogenesis of PD are still in its early stage, it is believed that intensive investigations will be inspired by such findings.
Fig. (2).
Effects of S-Persulfidation on parkin in PD. Under healthy conditions, an E3 ubiquitin ligase, the parkin activity is enhanced by S-Persulfidation and induces target protein degradation caused by ubiquitination. In PD, the activity of parkin is decreased by S-Persulfidation of reactive cysteine residues within parkin, thus causing accumulation of target protein, such as a-synuclein, and subsequent neurotoxicity. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
4.2. S-Persulfidation by H2S in HD
HD is characterized by the expansion of polyglutamine repeats in the protein huntingtin within the corpus striatum [119, 120]. Mutant huntingtin triggers its aggregates, leading to disrupted cognitive and motor functions along with psychiatric disturbances [121]. The transcriptional factor specificity protein 1 (Sp1) is a potent regulator of CSE [122, 123], and this transcriptional factor is sequestered and inactivated by mutant huntingtin in early HD [124], resulting in cell redox imbalance because of depleted cysteine biosynthesis. Indeed, the expression of CSE is diminished in HD brain tissues, indicating that H2S might play an essential role in the pathophysiology of HD [125, 126]. Aside from the deficiency of cysteine biosynthesis, the dysfunction of cysteine and cystine transporters is also observed in HD, all of which are responsible for elevated ROS production in cells due to a cysteine deficit [127, 128]. In response to amino acid limitation and endoplasmic reticulum stress, CSE is also modulated by activating transcription factor 4 (ATF4), this signaling pathway is impaired in HD as aberrant cysteine biosynthesis and transport contribute to oxidative stress-induced neurotoxicity [129]. In addition, the expression of CSE, a critical enzyme in the metabolism of cysteine, is downregulated in brain tissues of spinocerebellar ataxia type 3 patients, whereas thevoverexpression of CSE suppresses the detrimental effects of spinocerebellar ataxia type 3, a disease that is caused by a CAG repeat expansion in the ataxin-3 (ATXN3) gene [130]. Most importantly, CSE overexpression recovers protein S-Persulfidation and inhibits oxidative stress, thereby improving spinocerebellar ataxia type 3-associated tissue degeneration [130]. On the basis of these observations, we speculated that appropriate regulation of the reverse transsulfuration pathway is essential for the maintenance of cellular redox homeostasis, thus conferring neuroprotection. Although not conducted experimentally, S-Persulfidation of cysteine residues within the target proteins, such as huntingtin and ATXN3, most likely mediates the protective effects of H2S against HD, which may merit further investigation.
4.3. S-Persulfidation by H2S in AD
AD is a prevalent neurodegenerative disease that leads to cognitive dysfunction and memory loss in affected individuals [131, 132]. The pathophysiology of AD is closely associated with the generation of neurofibrillary tangles and accumulation of amyloid plaques, especially in the cerebral cortex and the hippocampus [133, 134]. Aggregation of β-amyloid (Aβ) and Tau proteins could induce the formation of amyloid plaques and neurofibrillary tangles, respectively [135, 136]. Mutations of the amyloid precursor protein (APP), presenilin-1 and 2 are frequently encountered in AD [137]. APP undergoes sequential proteolysis with the aid of proteases (also termed secretases), including α, β and γ secretases [138]. Similar to other several neurodegenerative diseases, AD is also tightly associated with elevated oxidative stress [139-141]. Increased lipid peroxidation, DNA damage and protein nitration are found to be engaged in the progression of AD [142-145]. Besides, mitochondrial dysfunction, transcriptional dysregulation, and aberrant nitrosylation occur at multiple levels in the progression of AD [33]. Regardless of the intensive research on the underlying mechanisms of AD, its etiologies are not fully elucidated.
H2S is proposed to be an important contributor to the development of AD [146-148]. It is reported that the high levels of homocysteine (a precursor of cysteine when acted on by CBS and CSE) are taken as a high risk factor for AD progression. It is reasonable that the decreased CBS activity accounts for the abnormal homocysteine deposition [149, 150]. The CBS-mediated homocysteine transsulfuration pathway is responsible for the development of AD [151], and the formation of H2S is actually hampered because of the disturbed CBS-mediated homocysteine transsulfuration in patients with AD [149, 152, 153]. As a matter of fact, the plasma level of H2S tends to be lower in AD patients and the decreased H2S level may be correlated with the severity of AD [151]. A number of studies have confirmed that H2S plays a protective role in the management of AD. Schreier et al., have found that H2S exerts a strong ability to counteract the cytotoxic lipid oxidation product 4-hydroxynonenal (HNE) in SH-SY5Y neuronal cells [154], an elevated cytotoxic in the brain tissues of severe AD patients [155, 156]. H2S donor sodium hydrosulfide (NaHS) is documented to improve spatial learning and memory impairment in a mouse model of AD [157]. Furthermore, the administration of NaHS slows down the development of experimental AD models via regulation of oxidative and nitrosative stress, inflammation and apoptosis [158]. A myriad of studies have also demonstrated that H2S influences Aβ formation and toxicity through various ways, such as APP glycosylation, γ secretase, cell cycle re-entry, inflammation response, and mitochondrial member potential [52, 159-162]. Thus, H2S might represent a new entity to delay AD progression through numerous signaling pathways. Nevertheless, it is noteworthy to mention that the precise roles of H2S in the pathogenesis of AD remain largely uncertain. Therefore, more studies are needed before its transformation to a clinical therapy for AD.
Neuroinflammation and excessive Aβ deposition synergistically contribute to the development and progression of AD [163]. The intervention, neuroinflammation and Aβ accumulation might provide a potential approach for AD therapy [163, 164]. Signal transducer and activator of transcription 3 (STAT3) is revealed to participate in neuroinflammation and Aβ pathogenesis during the progression of AD [165, 166]. Cathepsin S (Cat S) is predominantly expressed in the microglial cells and its inhibition produces neuroprotective effects in AD [167, 168]. We recently found that H2S attenuated adenosine triphosphate (ATP)-induced ROS production, inflammation response, and Aβ1-42 generation via inactivation of STAT3 and Cat S in both BV-2 and primary cultured microglial cells [169]. Moreover, we demonstrated that the S-Persulfidation of Cat S at cysteine-25 was required for H2S-mediated effects in the context of ATP [169]. Our results provided a novel understanding of the possible contribution S-Persulfidation of Cat S to the neuroprotective effects of H2S. In addition to the direct S-Persulfidation of Cat S, the Akt is S-Persulfidated at cysteine-77 by H2S, and Akt S-Persulfidation is also detected in the postmortem brains from AD patients [170]. Very recently, in a transgenic knockin mouse that lacked sulfhydrated Akt, decreased dendritic spine loss and improved cognitive dysfunctions were observed by reducing dendritic localization of human Tau is phosphorylated at S199 [170], representing a novel posttranslational modification of Akt, which primarily contributes to synaptic dysfunction in AD. These above findings suggest that S-Persulfidated proteins, such as Cat S and Akt, might exert a vital role in the pathological functions of AD.
A common feature of AD is aggravated oxidative burden in neuronal cells, and supplementation of H2S donor prevents neuronal cell death induced by oxidative stress [47, 171, 172]. H2S is also able to relieve oxidative injury by stimulating glutathione biosynthesis and aldehyde dehydrogenase 2 expressions [173, 174]. Additionally, the upregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) by H2S plays a vital role in the maintenance of cellular redox balance [175, 176]. As described earlier, H2S induces the increased Nrf2 activation by inducing S-Persulfidation of Keap1 and subsequent Keap1/Nrf2 disassociation [177-182]. However, whether Keap1 S-Persulfidation by H2S could be extrapolated to the treatment of AD requires in-depth research.
The α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptors (AMPARs) are key elements for fast excitatory synaptic transmission, and their dynamic regulation is majorly responsible for adaption of the CNS to environment stimulations [183, 184]. The dysfunction of AMPARs is one of the pathological factors for the development of AD [185]. A downregulation of AMPAR GluR1 subunit expression is a hallmark in pathological molecular alterations in AD [186]. Restoration of AMPAR GluR1 subunit or inhibition of AMPAR endocytosis may be promising new strategies to improve spatial memory deficits in the Tg2576 AD mouse model [187, 188]. Bath application of H2S and Na2S4 obviously promotes the surface insertion of AMPAR GluR1 subunit in the hippocampal tissues, and this is abolished in the presence of dithiothreitol (DTT), hinting an involvement of the S-Persulfidation-dependent mechanism [189]. However, AMPAR is not directly S-Persulfidated by H2S, but the phosphorylation levels of GluR1 at serine-831 and serine-845 sites are activated by H2S [189]. Despite this, H2S could directly increase the S-Persulfidation levels of postsynaptic signal molecules that control GluR1 phosphorylation, including protein phosphatase type 2A (PP2A), protein kinase A (PKA), protein kinase C, and calcium/calmodulin-dependent protein kinases II (CaMKII) [189]. This observation suggests that H2S promotes the surface delivery of AMPARs via S-Persulfidation-mediated mechanisms. In this regard, H2S-mediated S-Persulfidation of specific reactive thiols in target postsynaptic proteins indirectly regulates AMPARs in the hippocampus area, which may provide a new perspective of the pathophysiological functions of H2S in AD. Although the mechanisms that underlie S-Persulfidation-dependent regulation of kinase activity remain unclear, H2S may stimulate phosphorylation of these kinases via S-Persulfidation of themselves.
4.4. S-Persulfidation by H2S in Brain Memory Functions
Memory impairment is present in several neurodegenerative disorders, which is induced by numerous pathophysiological mechanisms, including neuroinflammation and aging [190-193]. IL-1β, a well-known pro-inflammatory cytokine, is widely observed in the brain [194, 195], and brain-derived IL-1β plays a dispensable role in the process of learning and memory via regulation of postsynaptic density 95 (PSD95), a critical scaffold protein that regulates synaptic stability, strength, and plasticity [196-199]. The brain memory impairment in response to IL-1β is mediated by CBS-generated H2S production as gene ablation of CBS ameliorates IL-1β-induced neurological impairments in mice [200]. At the molecular level, the induction of H2S by IL-1β modifies glyceraldehyde- 3-phosphate dehydrogenase (GAPDH) essentially via S-Persulfidation at cysteine-150, which enhances the binding of the E3 ligase Siah to the S-Persulfidated GAPDH, thereby inducing ubiquitination-mediated degradation of PSD95 in IL-1β-induced cognitive dysfunction [200]. GAPDH can not be S-Persulfidated by IL-1β when CBS is absent; the degradation of PSD95 will be less and IL-1β-triggered synaptic dysfunction and memory impairment are markedly relieved [200]. This study establishes a novel signaling pathway whereby IL-1β evokes the degradation of neuronal PSD95 by GAPDH S-Persulfidation in an H2S-dependent manner (Fig. 3). The S-Persulfidation of GAPDH by H2S may provide a therapeutic strategy for the prevention and treatment of neurological disorders-related memory impairment whereby the overproduction of IL-1β manifests the pathologies of such diseases.
Fig. (3).
S-Persulfidation of GAPDH mediates synaptic function. The proinflammtory cytokine IL-1β upregulates CBS-generating H2S production, and leads to S-Persulfidation of GAPDH. This event results in the binding of GAPDH to siah1, an E3 ubiquitin ligase, which triggers PSD95 toward for degradation. The dysregulation of PSD95 plays an important role in learning and memory dysfunction. CBS, cystathionine β-synthase; IL-1β: interleukin-1β; GAPDH: glycolytic enzyme glyceraldehyde 3-phosphate dehydrogenase; PSD95: post-synaptic density 95 protein; Siah1: seven in absentia homolog-1. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
LTP, a cellular model for memory, is regulated by various redox signaling molecules in which they act as double-edged swords [201-203]. In the presence of several thiol agents, such as DTT and glutathione, the LTP is increased and the aging-associated synaptic dysfunctions are enormously reversed [204-206], suggesting that the endogenous reducing agents might serve as a physiological mediator in synaptic plasticity. The D-serine released from astrocytes is one of the governing coagonists of synaptic NMDARs by stimulating its glycine modulatory sites [207-209]. Serine racemase (SR) is one of the main synthetases of D-serine [210]. The sulfide generation and protein S-Persulfidation are induced after high-frequency stimulation, which are necessary for NMDARs-dependent induction of LTP via the maintenance of D-serine [211]. The S-Persulfidation and disinhibition of SR by both H2S and polysulfides stimulate NMDARs-dependent LTP; this may be beneficial for hippocampus-dependent memory (Fig. 4) [211]. Actually, the levels of H2S and SR S-Persulfidation are diminished significantly in aged rats, and exogenous administration of H2S restores the S-Persulfidation of SR, followed by upregulation of D-serine and improvement of age-related deficits in hippocampus LTP [211]. In summary, this study suggests that H2S-induced posttranslational modification of SR appears to play a pivotal role in NMDARs-dependent synaptic plasticity, and H2S-based therapies may be effective for the management of memory loss in aging animals (Fig. 4). Excepting the S-Persulfidation of SR, other mechanisms underlying H2S-mediated regulation of D-serine await in-depth research. For example, activation of TRPA1 by polysulfides may promote the release of D-serine and NMDARs-dependent LTP [18, 212, 213]. These findings allow us to suppose a possible role for H2S-linked TRPA1 S-Persulfidation in synaptic plasticity and memory function; this hypothesis is further confirmed by a finding that polysulfides activate the TRPA1 channels by S-Persulfidating cysteine residues within the channels, thereby inducing Ca2+ influx in rat astrocytes (Fig. 4) [18, 28, 44].
Fig. (4).
A putative mechanism of H2S/polysulfides involving in the induction of LTP. The cysteine disulfide bond of NMDARs is reduced by H2S, thus enhancing its activity. Polysulfides derived from H2S further induce the formation of bound sulfane sulfur in the cysteine residues of NMDARs and further facilitate NMDARs-dependent induction of LTP. In astrocyte, activation of TRPA1 by polysulfides derived from H2S may promote the release of D-serine and NMDARs-dependent LTP, this effect may be mediated by S-Persulfidating the cysteine-422 and cysteine-622 of TRPA1. Also, the S-Persulfidation and disinhibition of SR by both H2S and polysulfides stimulate NMDARs-dependent induction of LTP, and SR is one of the main synthetases of D-serine. NMDARs: N-methyl-D-aspartate subtype glutamate receptors; TRPA1: transient receptor potential cation channel subfamily A member 1; CBS, cystathionine β-synthase; 3-MST, 3-mercaptopyruvatesulfurtransferase; CAT, cysteine aminotransferase; SR: serine racemase; LTP: long-term potentiation. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
4.5. Roles of H2S in other Neurological Disease
Amyotrophic lateral sclerosis (ALS), a detrimental neurodegenerative disease, is an important cause of the selective degeneration of upper and lower motor neurons [214]. Recently published results have revealed that abnormal autophagy, vesicle trafficking, RNA metabolism, and cytoskeleton dynamics act as functional pathways in ALS pathogenesis [215]. Human genetic studies have demonstrated that mutation of Cu/Zn superoxide dismutase (SOD1) gene mainly aggregates and causes motor neurons death in the process of ALS [216, 217]. The levels of H2S are higher in the spinal fluid of ALS patients, and the upregulated levels of H2S are also observed in the tissues from mice bearing the familial ALS mutation SOD1G93A [218]. In spinal cord cultures, H2S is toxic for motor neurons through increasing intracellular Ca2+ levels [218]. Interestingly, pharmacological inhibition of H2S obviously enhances the lifespan of female mice bearing the familial ALS mutation SOD1G93A, but not in male ALS mice [219]. This observation suggests that the relationship between gender and H2S needs to be adequately considered in the development of ALS. These studies unravel H2 S as an important mediator of motor neuron damage in the setting of ALS.
In accordance with the results obtained from ALS, the elevated H2S generation is also detected in Down syndrome, a disease manifested by trisomy of chromosome 21 (a chromosome on which CBS is located) [220]. The levels of urine thiosulfate, a catabolic product of H2S, are also upregulated in subjects with Down syndrome [221]. The expression of CBS is localized in astrocytes adjacent to the senile plaques, indicating involvement of H2S in this disease process [220]. A recent report has shown that overproduction of H2S disrupts Complex IV activity, mitochondrial electron transport, ATP synthesis and fibroblasts cell proliferation, inducing significant destruction in mitochondrial function of Down syndrome [222]. As a consequence, inhibition of CBS-derived H2S offers an attractive approach for the pharmacological treatment of Down syndrome-associated mitochondrial dysfunction. It is concluded that higher concentrations of H2S may be detrimental in both ALS and Down syndrome. However, the S-Persulfidation-related mechanisms by H2S are scarce in both diseases. At the molecular level, we should develop proteomic approaches to solve the sites of H2S-mediated S-Persulfidation and its interplay with the pathogenesis of ALS and Down syndrome.
It has been accepted that H2S promotes tumor proliferation in some types of cancer, while it inhibits tumor cell growth in other cancer types [61]. Glioblastoma is a malignant brain tumor type with a poor prognosis [223]. Currently, effective and sufficient therapeutic strategies for glioblastoma are unavailable due to the poor understanding of its pathological mechanisms [224]. The roles of H2S in glioblastoma development and progression are recently disclosed by cell and animal experiments. Injection of H2S donor NaHS aggravates the physical symptoms of glioma-bearing rats via upregulating hypoxia-inducible factor 1α (HIF-1α) expression and neovascular formation [225]. Incubation of C6 glioma cells with NaHS (400 µM) facilitates the cell proliferation and inhibits cell apoptosis, and this may be mediated by the p38 MAPK/ERK1/2-COX-2 pathway [226]. The tumor-promoting effects of H2S are challenged by a finding that the administration of NaHS inhibits cell proliferation and induces apoptosis of C6 gliomas cells through the p38 MAPK signaling pathway [227]. In accordance, the silencing of CBS, a major H2S-generating enzyme in the brain, promotes the growth of human glioma tumor cells [228]. However, whether the level of CBS is altered in human glioma tissues is not examined. Considering that 3-MST expression is obviously enhanced in gliomas tissue [229], we can not exclude a possibility that 3-MST, a crucial enzyme for H2S production in the brain system, may be upregulated on CBS silencing, thus compensatory H2S from 3-MST might promote the growth of glioma cells in the absence of CBS. However, the direct effects of 3-MST on glioma behaviors are unknown. Therefore, the exact roles of CBS and 3-MST in glioma development will be an interesting field to explore in the near future. Doxorubicin exerts cytotoxicity toward rat C6 glioma cells [230], this effect may be attributed to an elevation of H2S contents as the pro-apoptotic effects of H2S at high millimolar doses are accompanied by an increase in the generation of ROS and a decrease in the gluthatione concentration, leading to activation of the caspase 3-mediated apoptotic pathway [231-233]. All in all, the effects of H2S on glioma biology remain controversial, and these unsolved matters need to be answered in further studies. Overwhelming evidence suggests that H2S-mediated S-Persulfidation of target proteins are demonstrated to be involved in a plethora of signaling transduction during the development and progression of different types of cancer [61]. Unfortunately, this post-translational modification paradigm is unexplored in H2S-mediated effects on glioblastoma. Consequently, H2S-mediated protein S-Persulfidation that is relevant to glioblastoma biology will become a focus of future research.
In agreement with the above findings, although H2S is an important dominator in the pathologies of stroke and traumatic brain injury from accumulative studies [10, 52, 88], no extensive investigations are performed to determine the possible effects of H2S-mediated S-Persulfidation of target proteins in both neurodegenerative diseases. It is shown that sulfane sulfur in astrocytes is obviously lower in stroke-prone spontaneously hypertensive rats, and this decrease is further diminished by CBS inhibitor [234], implying that the formation of S-Persulfidation by H2S may be involved in this disease development. More original experiments are warranted to provide novel insights into how potential S-Persulfidation on specific cysteine residues by H2S could induce benefits to the pathological changes in stroke and traumatic brain injury.
5. S-PERSULFIDATION BY POLYSULFIDES IN THE CNS
Polysulfides are identified to be abundantly expressed in the brain, and exhibit a higher oxidation capability than the sulfur atom in H2S as they contain sulfane sulfur [25, 65]. The levels of polysulfides in the brain are found to be in micromolar concentrations using high-performance liquid chromatography (HPLC) analysis, this dose of polysulfides is enough to activate the TRPA1 channels [18]. Even though it is still unclear whether polysulfides are actively transported into cells, polysulfides are known to easily pass through the plasma membranes [18, 23]. Similar to H2S, polysulfides are also involved in neurodegenerative diseases, including PD, HD, ethylmalonyl encephalopathy, and even in brain cancer [29, 235, 236].
Studies have established that polysulfides may directly target several molecules, such as Keap1/Nrf2 complex [237], a tumor suppresser PTEN [23], GAPDH, an enzyme that catalyzes glycolysis [238], and a vascular tension regulator protein kinase G1α [239]. Apart from being important signaling molecules, polysulfides also function as a neuroprotective modulator by activating the channels, enzymes, and transcription factors through S-Persulfidation of the target proteins [28, 42, 50, 240]. Polysulfides activate the TRPA1 channels by S-Persulfidated cysteine residues within the channels, thereby inducing Ca2+ influx in rat astrocytes (Fig. 4) [18]. The actions of polysulfides are attenuated by inhibitors of TRPA1 or silencing of TRPA1, implying that the responses of polysulfides are attributed to TRPA1 activation. Once activated in astrocytes, the release of D-serine to the synapse enhances the activity of NMDARs and facilitates the induction of LTP [212, 241].
The imbalance in cellular redox state is an important reason for oxidative stress, which is also a common etiological factor in neurological diseases [242, 243]. Under normal homeostatic conditions, Keap1 is a redox-sensitive ubiquitin ligase substrate adaptor that represses the activity of the transcription factor Nrf2 [178, 181, 244]. Upon oxidative stress, Keap1 S-sulfhydration by polysulfides induces Nrf2 dissociation from Keap1, which upregulates the nuclear translocation of Nrf2 and the subsequent expressions of antioxidant genes [237], thus conferring beneficial effects against oxidative injury in Neuro2A cells. In addition, the activation of the PI3K/Akt pathway is necessary for polysulfides for the translocation of Nrf2 to the nucleus [237]. With the growing identification of anti-oxidative protein, S-sulfhydration, by polysulfides, significant advancements in the roles and mechanisms of polysulfides in modulating cellular responses to oxidative stress in the neurodegenerative process will be made.
A recent study has detected the higher levels of polysulfides in glioblastoma-bearing ipsilateral hemispheres, but not in glioblastoma-free control hemispheres using surface-enhanced Raman spectroscopy [235]. Although it is still unknown, for the accurate molecular entities of polysulfides in glioma cells, they might be involved in the pathologies of glioma. GAPDH is an enzyme that catalyzes glycolysis, an orchestrated process in the development of chemotherapy resistance in some types of malignancies, including glioma [245-248]. For this reason, inhibition of GAPDH activity has gained considerable attention as an attractive strategy for cancer therapy [249, 250]. Interestingly, polysulfides treatment significantly inhibits the activity of GAPDH, by S-Persulfidation of GAPDH at cysteine-156 and cysteine-247 [238]. Accordingly, it will be exciting to know whether targeting S-Persulfidation of GAPDH by polysulfides alters the glycolysis process in brain gliomas. This hypothesis, therefore, requires further research. Furthermore, polysulfides are shown to inhibit the activity of PTEN by inducing the generation of a cysteine disulfide bond [23]. The development and progression of glioblastoma are intimately related to abnormal PTEN expression [251], as dysregulated PTEN may participate in glioma initiation, progression, and treatment resistance [252, 253]. On these grounds, the protein S-Persulfidation by polysulfides may be an attractive therapeutic avenue for the prevention of brain gliomas. Further investigations are needed to verify this assumption.
CONCLUSION AND FUTURE PERSPECTIVES
Over the last decade, an important post-translational modification induced by H2S and polysulfides, named S-Persulfidation, has been well accepted. S-Persulfidation is a novel redox pathway to exhibit diverse biological processes in H2S/polysulfides signaling. There is no doubt that the studies on protein S-Persulfidation are increasingly proposed as the future research direction in the field of gasotransmitters for the coming years. However, there are still several issues to be resolved. (1) The production and metabolism pathways of polysulfides in the brain are largely unknown. Solving this problem might provide novel insights into the biochemistry of H2S and facilitate the therapeutic application of H2S-derived compounds. (2) In the process of protein S-Persulfidation, both small-molecule based persulfides and protein persulfides are correspondingly generated, and such species are highly reactive. The metabolic regulation of these species is largely unknown. (3) It is interestingly to know how brain cells differentially use H2S and polysulfides at the appropriate time points. (4) More scientific approaches with higher specificity and sensitivity are urgently required to detect protein S-Persulfidation. (5) More protein cysteine sites of S-Persulfidation are necessary to be clarified in the CNS. (6) The interactions of S-Persulfidation with other post-translational modifications, such as S-nitrosylation, deserve to be elucidated in neuropathy. (7) The clinical relevance of S-Persulfidation in neurological disorders needs to be explored in detail.
It is anticipated that a comprehensive understanding of protein S-Persulfidation will be helpful in identifying the underlying mechanisms in which S-Persulfidation could benefit various neurological disorders. Importantly, the S-Persulfidated proteins could serve as potential targets for the therapeutic intervention of neurological disorders, thus advancing the development of H2S/polysulfides-based agents in the near future.
ACKNOWLEDGEMENTS
Hai-Jian Sun, Zhi-Yuan Wu, Xiao-Wei Nie, and Jin-Song Bian were responsible for the manuscript writing, proofreading and editing.
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
This work was supported by the Ministry of Education of Singapore Tier 2 Research grant (MOE2017-T2-2-029).
CONFLICT OF INTEREST
The authors declare no conflict of interest.
REFERENCES
- 1.Kimura H. Hydrogen sulfide and polysulfides as signaling molecules. Proc. Jpn. Acad., Ser. B, Phys. Biol. Sci. 2015;91(4):131–159. doi: 10.2183/pjab.91.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Paul B.D., Snyder S.H. Modes of physiologic H2S signaling in the brain and peripheral tissues. Antioxid. Redox Signal. 2015;22(5):411–423. doi: 10.1089/ars.2014.5917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Sun H.J., Wu Z.Y., Cao L., Zhu M.Y., Liu T.T., Guo L., Lin Y., Nie X.W., Bian J.S. Hydrogen sulfide: recent progression and perspectives for the treatment of diabetic nephropathy. Molecules. 2019;24(15):2857. doi: 10.3390/molecules24152857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Liu Y.H., Lu M., Hu L.F., Wong P.T., Webb G.D., Bian J.S. Hydrogen sulfide in the mammalian cardiovascular system. Antioxid. Redox Signal. 2012;17(1):141–185. doi: 10.1089/ars.2011.4005. [DOI] [PubMed] [Google Scholar]
- 5.Sun H.J., Wu Z.Y., Nie X.W., Bian J.S. Role of endothelial dysfunction in cardiovascular diseases: the link between inflammation and hydrogen sulfide. Front. Pharmacol. 2019;10:1568. doi: 10.3389/fphar.2019.01568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Meng G., Zhao S., Xie L., Han Y., Ji Y. Protein S-sulfhydration by hydrogen sulfide in cardiovascular system. Br. J. Pharmacol. 2018;175(8):1146–1156. doi: 10.1111/bph.13825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wang R. Signaling pathways for the vascular effects of hydrogen sulfide. Curr. Opin. Nephrol. Hypertens. 2011;20(2):107–112. doi: 10.1097/MNH.0b013e3283430651. [DOI] [PubMed] [Google Scholar]
- 8.Paul B.D., Snyder S.H. Gasotransmitter hydrogen sulfide signaling in neuronal health and disease. Biochem. Pharmacol. 2018;149:101–109. doi: 10.1016/j.bcp.2017.11.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Zhang X., Bian J.S. Hydrogen sulfide: a neuromodulator and neuroprotectant in the central nervous system. ACS Chem. Neurosci. 2014;5(10):876–883. doi: 10.1021/cn500185g. [DOI] [PubMed] [Google Scholar]
- 10.Tan B.H., Wong P.T., Bian J.S. Hydrogen sulfide: a novel signaling molecule in the central nervous system. Neurochem. Int. 2010;56(1):3–10. doi: 10.1016/j.neuint.2009.08.008. [DOI] [PubMed] [Google Scholar]
- 11.Hu L.F., Lu M., Hon Wong P.T., Bian J.S. Hydrogen sulfide: neurophysiology and neuropathology. Antioxid. Redox Signal. 2011;15(2):405–419. doi: 10.1089/ars.2010.3517. [DOI] [PubMed] [Google Scholar]
- 12.Paul B.D., Snyder S.H. H2S: A Novel gasotransmitter that signals by sulfhydration. Trends Biochem. Sci. 2015;40(11):687–700. doi: 10.1016/j.tibs.2015.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Cirino G., Vellecco V., Bucci M. Nitric oxide and hydrogen sulfide: the gasotransmitter paradigm of the vascular system. Br. J. Pharmacol. 2017;174(22):4021–4031. doi: 10.1111/bph.13815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Warenycia M.W., Goodwin L.R., Benishin C.G., Reiffenstein R.J., Francom D.M., Taylor J.D., Dieken F.P. Acute hydrogen sulfide poisoning. Demonstration of selective uptake of sulfide by the brainstem by measurement of brain sulfide levels. Biochem. Pharmacol. 1989;38(6):973–981. doi: 10.1016/0006-2952(89)90288-8. [DOI] [PubMed] [Google Scholar]
- 15.Abe K., Kimura H. The possible role of hydrogen sulfide as an endogenous neuromodulator. J. Neurosci. 1996;16(3):1066–1071. doi: 10.1523/JNEUROSCI.16-03-01066.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lee S.W., Hu Y.S., Hu L.F., Lu Q., Dawe G.S., Moore P.K., Wong P.T., Bian J.S. Hydrogen sulphide regulates calcium homeostasis in microglial cells. Glia. 2006;54(2):116–124. doi: 10.1002/glia.20362. [DOI] [PubMed] [Google Scholar]
- 17.Nagai Y., Tsugane M., Oka J., Kimura H. Hydrogen sulfide induces calcium waves in astrocytes. FASEB J. 2004;18(3):557–559. doi: 10.1096/fj.03-1052fje. [DOI] [PubMed] [Google Scholar]
- 18.Kimura Y., Mikami Y., Osumi K., Tsugane M., Oka J., Kimura H. Polysulfides are possible H2S-derived signaling molecules in rat brain. FASEB J. 2013;27(6):2451–2457. doi: 10.1096/fj.12-226415. [DOI] [PubMed] [Google Scholar]
- 19.Yong Q.C., Choo C.H., Tan B.H., Low C.M., Bian J.S. Effect of hydrogen sulfide on intracellular calcium homeostasis in neuronal cells. Neurochem. Int. 2010;56(3):508–515. doi: 10.1016/j.neuint.2009.12.011. [DOI] [PubMed] [Google Scholar]
- 20.Lu M., Choo C.H., Hu L.F., Tan B.H., Hu G., Bian J.S. Hydrogen sulfide regulates intracellular pH in rat primary cultured glia cells. Neurosci. Res. 2010;66(1):92–98. doi: 10.1016/j.neures.2009.09.1713. [DOI] [PubMed] [Google Scholar]
- 21.Cao X., Zhang W., Moore P.K., Bian J. Protective smell of hydrogen sulfide and polysulfide in cisplatin-induced nephrotoxicity. Int. J. Mol. Sci. 2019;20(2):313. doi: 10.3390/ijms20020313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Nagy P., Winterbourn C.C. Rapid reaction of hydrogen sulfide with the neutrophil oxidant hypochlorous acid to generate polysulfides. Chem. Res. Toxicol. 2010;23(10):1541–1543. doi: 10.1021/tx100266a. [DOI] [PubMed] [Google Scholar]
- 23.Greiner R., Palinkas Z., Basell K., Becher D., Antelmann H., Nagy P., Dick T.P. Polysulfides link H2S to protein thiol oxidation. Antioxid. Redox Signal. 2013;19(15):1749–1765. doi: 10.1089/ars.2012.5041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Toohey J.I. Sulfur signaling: is the agent sulfide or sulfane? Anal. Biochem. 2011;413(1):1–7. doi: 10.1016/j.ab.2011.01.044. [DOI] [PubMed] [Google Scholar]
- 25.Kimura H. Physiological roles of hydrogen sulfide and polysulfides. Handb. Exp. Pharmacol. 2015;230:61–81. doi: 10.1007/978-3-319-18144-8_3. [DOI] [PubMed] [Google Scholar]
- 26.Mustafa A.K., Gadalla M.M., Sen N., Kim S., Mu W., Gazi S.K., Barrow R.K., Yang G., Wang R., Snyder S.H. H2S signals through protein S-sulfhydration. Sci. Signal. 2009;2(96):ra72. doi: 10.1126/scisignal.2000464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Iciek M., Kowalczyk-Pachel D., Bilska-Wilkosz A., Kwiecien I., Gorny M., Wlodek L. S-sulfhydration as a cellular redox regulation. Biosci. Rep. 2015;36(2) doi: 10.1042/BSR20150147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kimura H. Hydrogen sulfide and polysulfides as biological mediators. Molecules. 2014;19(10):16146–16157. doi: 10.3390/molecules191016146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Vandiver M.S., Paul B.D., Xu R., Karuppagounder S., Rao F., Snowman A.M., Ko H.S., Lee Y.I., Dawson V.L., Dawson T.M., Sen N., Snyder S.H. Sulfhydration mediates neuroprotective actions of parkin. Nat. Commun. 2013;4:1626. doi: 10.1038/ncomms2623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zivanovic J., Kouroussis E., Kohl J.B., Adhikari B., Bursac B., Schott-Roux S., Petrovic D., Miljkovic J.L., Thomas-Lopez D., Jung Y., Miler M., Mitchell S., Milosevic V., Gomes J.E., Benhar M., Gonzalez-Zorn B., Ivanovic-Burmazovic I., Torregrossa R., Mitchell J.R., Whiteman M., Schwarz G., Snyder S.H., Paul B.D., Carroll K.S., Filipovic M.R. Selective persulfide detection reveals evolutionarily conserved antiaging effects of s-sulfhydration. Cell Metab. 2019;30(6):1152–1170.e1113. doi: 10.1016/j.cmet.2019.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Yang C.T., Devarie-Baez N.O., Hamsath A., Fu X.D., Xian M. S-Persulfidation: chemistry, chemical biology, and significance in health and disease. Antioxid. Redox Signal. 2020;33(15):1092–1114. doi: 10.1089/ars.2019.7889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Wedmann R., Onderka C., Wei S., Szijarto I.A., Miljkovic J.L., Mitrovic A., Lange M., Savitsky S., Yadav P.K., Torregrossa R., Harrer E.G., Harrer T., Ishii I., Gollasch M., Wood M.E., Galardon E., Xian M., Whiteman M., Banerjee R., Filipovic M.R. Improved tag-switch method reveals that thioredoxin acts as depersulfidase and controls the intracellular levels of protein persulfidation. Chem. Sci. (Camb.) 2016;7(5):3414–3426. doi: 10.1039/C5SC04818D. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Sbodio J.I., Snyder S.H., Paul B.D. Redox mechanisms in neurodegeneration: from disease outcomes to therapeutic opportunities. Antioxid. Redox Signal. 2019;30(11):1450–1499. doi: 10.1089/ars.2017.7321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Goodwin L.R., Francom D., Dieken F.P., Taylor J.D., Warenycia M.W., Reiffenstein R.J., Dowling G. Determination of sulfide in brain tissue by gas dialysis/ion chromatography: postmortem studies and two case reports. J. Anal. Toxicol. 1989;13(2):105–109. doi: 10.1093/jat/13.2.105. [DOI] [PubMed] [Google Scholar]
- 35.Savage J.C., Gould D.H. Determination of sulfide in brain tissue and rumen fluid by ion-interaction reversed-phase high-performance liquid chromatography. J. Chromatogr. A. 1990;526(2):540–545. doi: 10.1016/S0378-4347(00)82537-2. [DOI] [PubMed] [Google Scholar]
- 36.Yang C.T., Lai Z.Z., Zheng Z.H., Kang J.M., Xian M., Wang R.Y., Shi K., Meng F.H., Li X., Chen L., Zhang H. A novel pH-controlled hydrogen sulfide donor protects gastric mucosa from aspirin-induced injury. J. Cell. Mol. Med. 2017;21(10):2441–2451. doi: 10.1111/jcmm.13166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Chappell M.C. Hydrogen sulfide, then nitric oxide and vasoprotection. J. Hypertens. 2018;36(3):493–494. doi: 10.1097/HJH.0000000000001617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Yang C.T., Wang Y., Marutani E., Ida T., Ni X., Xu S., Chen W., Zhang H., Akaike T. Data-driven identification of hydrogen sulfide scavengers. Angew. Chem. Int. Ed. Engl. 2019;58(32):10898–10902. doi: 10.1002/anie.201905580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yang C.T., Chen L., Xu S., Day J.J., Li X., Xian M. Recent development of hydrogen sulfide releasing/stimulating reagents and their potential applications in cancer and glycometabolic disorders. Front. Pharmacol. 2017;8:664. doi: 10.3389/fphar.2017.00664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zhao Y., Biggs T.D., Xian M. Hydrogen sulfide (H2S) releasing agents: chemistry and biological applications. Chem. Commun. (Camb.) 2014;50(80):11788–11805. doi: 10.1039/C4CC00968A. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Shibuya N., Koike S., Tanaka M., Ishigami-Yuasa M., Kimura Y., Ogasawara Y., Fukui K., Nagahara N., Kimura H. A novel pathway for the production of hydrogen sulfide from D-cysteine in mammalian cells. Nat. Commun. 2013;4:1366. doi: 10.1038/ncomms2371. [DOI] [PubMed] [Google Scholar]
- 42.Kimura H. The physiological role of hydrogen sulfide and beyond. Nitric Oxide. 2014;41:4–10. doi: 10.1016/j.niox.2014.01.002. [DOI] [PubMed] [Google Scholar]
- 43.Shibuya N., Kimura H. Production of hydrogen sulfide from d-cysteine and its therapeutic potential. Front. Endocrinol. (Lausanne) 2013;4:87. doi: 10.3389/fendo.2013.00087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kimura H. Signaling molecules: hydrogen sulfide and polysulfide. Antioxid. Redox Signal. 2015;22(5):362–376. doi: 10.1089/ars.2014.5869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Seki T. Availability of D-cysteine as a protectant for cerebellar neurons. Nippon Yakurigaku Zasshi. 2019;154(3):133–137. doi: 10.1254/fpj.154.133. [DOI] [PubMed] [Google Scholar]
- 46.Enokido Y., Suzuki E., Iwasawa K., Namekata K., Okazawa H., Kimura H. Cystathionine beta-synthase, a key enzyme for homocysteine metabolism, is preferentially expressed in the radial glia/astrocyte lineage of developing mouse CNS. FASEB J. 2005;19(13):1854–1856. doi: 10.1096/fj.05-3724fje. [DOI] [PubMed] [Google Scholar]
- 47.Kimura Y., Kimura H. Hydrogen sulfide protects neurons from oxidative stress. FASEB J. 2004;18(10):1165–1167. doi: 10.1096/fj.04-1815fje. [DOI] [PubMed] [Google Scholar]
- 48.Kimura Y., Dargusch R., Schubert D., Kimura H. Hydrogen sulfide protects HT22 neuronal cells from oxidative stress. Antioxid. Redox Signal. 2006;8(3-4):661–670. doi: 10.1089/ars.2006.8.661. [DOI] [PubMed] [Google Scholar]
- 49.Kimura H. Hydrogen sulfide: from brain to gut. Antioxid. Redox Signal. 2010;12(9):1111–1123. doi: 10.1089/ars.2009.2919. [DOI] [PubMed] [Google Scholar]
- 50.Kimura H. Physiological role of hydrogen sulfide and polysulfide in the central nervous system. Neurochem. Int. 2013;63(5):492–497. doi: 10.1016/j.neuint.2013.09.003. [DOI] [PubMed] [Google Scholar]
- 51.Cao X., Cao L., Ding L., Bian J.S. A new hope for a devastating disease: hydrogen sulfide in Parkinson’s disease. Mol. Neurobiol. 2018;55(5):3789–3799. doi: 10.1007/s12035-017-0617-0. [DOI] [PubMed] [Google Scholar]
- 52.Qu K., Lee S.W., Bian J.S., Low C.M., Wong P.T. Hydrogen sulfide: neurochemistry and neurobiology. Neurochem. Int. 2008;52(1-2):155–165. doi: 10.1016/j.neuint.2007.05.016. [DOI] [PubMed] [Google Scholar]
- 53.Wang J.F., Li Y., Song J.N., Pang H.G. Role of hydrogen sulfide in secondary neuronal injury. Neurochem. Int. 2014;64:37–47. doi: 10.1016/j.neuint.2013.11.002. [DOI] [PubMed] [Google Scholar]
- 54.Chen X., Jhee K.H., Kruger W.D. Production of the neuromodulator H2S by cystathionine beta-synthase via the condensation of cysteine and homocysteine. J. Biol. Chem. 2004;279(50):52082–52086. doi: 10.1074/jbc.C400481200. [DOI] [PubMed] [Google Scholar]
- 55.Shibuya N., Tanaka M., Yoshida M., Ogasawara Y., Togawa T., Ishii K., Kimura H. 3-Mercaptopyruvate sulfurtransferase produces hydrogen sulfide and bound sulfane sulfur in the brain. Antioxid. Redox Signal. 2009;11(4):703–714. doi: 10.1089/ars.2008.2253. [DOI] [PubMed] [Google Scholar]
- 56.Mikami Y., Shibuya N., Kimura Y., Nagahara N., Yamada M., Kimura H. Hydrogen sulfide protects the retina from light-induced degeneration by the modulation of Ca2+ influx. J. Biol. Chem. 2011;286(45):39379–39386. doi: 10.1074/jbc.M111.298208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Griffith O.W. Biologic and pharmacologic regulation of mammalian glutathione synthesis. Free Radic. Biol. Med. 1999;27(9-10):922–935. doi: 10.1016/S0891-5849(99)00176-8. [DOI] [PubMed] [Google Scholar]
- 58.Cao X., Wu Z., Xiong S., Cao L., Sethi G., Bian J.S. The role of hydrogen sulfide in cyclic nucleotide signaling. Biochem. Pharmacol. 2018;149:20–28. doi: 10.1016/j.bcp.2017.11.011. [DOI] [PubMed] [Google Scholar]
- 59.Ishigami M., Hiraki K., Umemura K., Ogasawara Y., Ishii K., Kimura H. A source of hydrogen sulfide and a mechanism of its release in the brain. Antioxid. Redox Signal. 2009;11(2):205–214. doi: 10.1089/ars.2008.2132. [DOI] [PubMed] [Google Scholar]
- 60.Kabil O., Banerjee R. Enzymology of H2S biogenesis, decay and signaling. Antioxid. Redox Signal. 2014;20(5):770–782. doi: 10.1089/ars.2013.5339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Cao X., Ding L., Xie Z.Z., Yang Y., Whiteman M., Moore P.K., Bian J.S. A review of hydrogen sulfide synthesis, metabolism, and measurement: is modulation of hydrogen sulfide a novel therapeutic for cancer? Antioxid. Redox Signal. 2019;31(1):1–38. doi: 10.1089/ars.2017.7058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kimura H. Hydrogen polysulfide (H2S n) signaling along with hydrogen sulfide (H2S) and nitric oxide (NO). J. Neural Transm. (Vienna) 2016;123(11):1235–1245. doi: 10.1007/s00702-016-1600-z. [DOI] [PubMed] [Google Scholar]
- 63.Nagpure B.V., Bian J.S. Interaction of hydrogen sulfide with nitric oxide in the cardiovascular system. Oxid. Med. Cell. Longev. 2016;2016:6904327. doi: 10.1155/2016/6904327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Sun H.J., Lee W.T., Leng B., Wu Z.Y., Yang Y., Bian J.S. Nitroxyl as a potential theranostic in the cancer arena. Antioxid. Redox Signal. 2020;32(5):331–349. doi: 10.1089/ars.2019.7904. [DOI] [PubMed] [Google Scholar]
- 65.Miyamoto R., Koike S., Takano Y., Shibuya N., Kimura Y., Hanaoka K., Urano Y., Ogasawara Y., Kimura H. Polysulfides (H2Sn) produced from the interaction of hydrogen sulfide (H2S) and nitric oxide (NO) activate TRPA1 channels. Sci. Rep. 2017;7:45995. doi: 10.1038/srep45995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Francoleon N.E., Carrington S.J., Fukuto J.M. The reaction of H(2)S with oxidized thiols: generation of persulfides and implications to H(2)S biology. Arch. Biochem. Biophys. 2011;516(2):146–153. doi: 10.1016/j.abb.2011.09.015. [DOI] [PubMed] [Google Scholar]
- 67.Filipovic M.R., Zivanovic J., Alvarez B., Banerjee R. Chemical biology of H2S signaling through persulfidation. Chem. Rev. 2018;118(3):1253–1337. doi: 10.1021/acs.chemrev.7b00205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Poole L.B., Nelson K.J. Discovering mechanisms of signaling-mediated cysteine oxidation. Curr. Opin. Chem. Biol. 2008;12(1):18–24. doi: 10.1016/j.cbpa.2008.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Klomsiri C., Karplus P.A., Poole L.B. Cysteine-based redox switches in enzymes. Antioxid. Redox Signal. 2011;14(6):1065–1077. doi: 10.1089/ars.2010.3376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Finkel T. From sulfenylation to sulfhydration: what a thiolate needs to tolerate. Sci. Signal. 2012;5(215):pe10. doi: 10.1126/scisignal.2002943. [DOI] [PubMed] [Google Scholar]
- 71.Cuevasanta E., Lange M., Bonanata J., Coitino E.L., Ferrer-Sueta G., Filipovic M.R., Alvarez B. Reaction of hydrogen sulfide with disulfide and sulfenic acid to form the strongly nucleophilic persulfide. J. Biol. Chem. 2015;290(45):26866–26880. doi: 10.1074/jbc.M115.672816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Woo H.A., Chae H.Z., Hwang S.C., Yang K.S., Kang S.W., Kim K., Rhee S.G. Reversing the inactivation of peroxiredoxins caused by cysteine sulfinic acid formation. Science. 2003;300(5619):653–656. doi: 10.1126/science.1080273. [DOI] [PubMed] [Google Scholar]
- 73.Wood Z.A., Poole L.B., Karplus P.A. Peroxiredoxin evolution and the regulation of hydrogen peroxide signaling. Science. 2003;300(5619):650–653. doi: 10.1126/science.1080405. [DOI] [PubMed] [Google Scholar]
- 74.Cuevasanta E., Moller M.N., Alvarez B. Biological chemistry of hydrogen sulfide and persulfides. Arch. Biochem. Biophys. 2017;617:9–25. doi: 10.1016/j.abb.2016.09.018. [DOI] [PubMed] [Google Scholar]
- 75.Greiner R., Dick T.P. Real-time assays for monitoring the influence of sulfide and sulfane sulfur species on protein thiol redox states. Methods Enzymol. 2015;555:57–77. doi: 10.1016/bs.mie.2014.11.020. [DOI] [PubMed] [Google Scholar]
- 76.Kimura Y., Toyofuku Y., Koike S., Shibuya N., Nagahara N., Lefer D., Ogasawara Y., Kimura H. Identification of H2S3 and H2S produced by 3-mercaptopyruvate sulfurtransferase in the brain. Sci. Rep. 2015;5:14774. doi: 10.1038/srep14774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kimura Y., Koike S., Shibuya N., Lefer D., Ogasawara Y., Kimura H. 3-Mercaptopyruvate sulfurtransferase produces potential redox regulators cysteine- and glutathione-persulfide (Cys-SSH and GSSH) together with signaling molecules H2S2, H2S3 and H2S. Sci. Rep. 2017;7(1):10459. doi: 10.1038/s41598-017-11004-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ida T., Sawa T., Ihara H., Tsuchiya Y., Watanabe Y., Kumagai Y., Suematsu M., Motohashi H., Fujii S., Matsunaga T., Yamamoto M., Ono K., Devarie-Baez N.O., Xian M., Fukuto J.M., Akaike T. Reactive cysteine persulfides and S-polythiolation regulate oxidative stress and redox signaling. Proc. Natl. Acad. Sci. USA. 2014;111(21):7606–7611. doi: 10.1073/pnas.1321232111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Ono K., Akaike T., Sawa T., Kumagai Y., Wink D.A., Tantillo D.J., Hobbs A.J., Nagy P., Xian M., Lin J., Fukuto J.M. Redox chemistry and chemical biology of H2S, hydropersulfides, and derived species: implications of their possible biological activity and utility. Free Radic. Biol. Med. 2014;77:82–94. doi: 10.1016/j.freeradbiomed.2014.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Akaike T., Ida T., Wei F.Y., Nishida M., Kumagai Y., Alam M.M., Ihara H., Sawa T., Matsunaga T., Kasamatsu S., Nishimura A., Morita M., Tomizawa K., Nishimura A., Watanabe S. Cysteinyl-tRNA synthetase governs cysteine polysulfidation and mitochondrial bioenergetics. Nat. Commun. 2017;8(1):1177. doi: 10.1038/s41467-017-01311-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Sen N. Functional and molecular insights of hydrogen sulfide signaling and protein sulfhydration. J. Mol. Biol. 2017;429(4):543–561. doi: 10.1016/j.jmb.2016.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Lu C., Kavalier A., Lukyanov E., Gross S.S. S-sulfhydration/desulfhydration and S-nitrosylation/denitrosylation: a common paradigm for gasotransmitter signaling by H2S and NO. Methods. 2013;62(2):177–181. doi: 10.1016/j.ymeth.2013.05.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Dongo E., Beliczai-Marosi G., Dybvig A.S., Kiss L. The mechanism of action and role of hydrogen sulfide in the control of vascular tone. Nitric Oxide. 2018;81:75–87. doi: 10.1016/j.niox.2017.10.010. [DOI] [PubMed] [Google Scholar]
- 84.Sandu R.E., Dumbrava D., Surugiu R., Glavan D.G., Gresita A., Petcu E.B. Cellular and molecular mechanisms underlying non-pharmaceutical ischemic stroke therapy in aged subjects. Int. J. Mol. Sci. 2017;19(1):99. doi: 10.3390/ijms19010099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Zhang J.Y., Ding Y.P., Wang Z., Kong Y., Gao R., Chen G. Hydrogen sulfide therapy in brain diseases: from bench to bedside. Med. Gas Res. 2017;7(2):113–119. doi: 10.4103/2045-9912.208517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Tabassum R., Jeong N.Y., Jung J. Therapeutic importance of hydrogen sulfide in age-associated neurodegenerative diseases. Neural Regen. Res. 2020;15(4):653–662. doi: 10.4103/1673-5374.266911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Che X., Fang Y., Si X., Wang J., Hu X., Reis C., Chen S. The role of gaseous molecules in traumatic brain injury: an updated review. Front. Neurosci. 2018;12:392. doi: 10.3389/fnins.2018.00392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Chan S.J., Wong P.T. Hydrogen sulfide in stroke: Protective or deleterious? Neurochem. Int. 2017;105:1–10. doi: 10.1016/j.neuint.2016.11.015. [DOI] [PubMed] [Google Scholar]
- 89.Wu D., Wang J., Li H., Xue M., Ji A., Li Y. Role of hydrogen sulfide in ischemia-reperfusion injury. Oxid. Med. Cell. Longev. 2015;2015:186908. doi: 10.1155/2015/186908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Rumbeiha W., Whitley E., Anantharam P., Kim D.S., Kanthasamy A. Acute hydrogen sulfide-induced neuropathology and neurological sequelae: challenges for translational neuroprotective research. Ann. N. Y. Acad. Sci. 2016;1378(1):5–16. doi: 10.1111/nyas.13148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Dou Y., Wang Z., Chen G. The role of hydrogen sulfide in stroke. Med. Gas Res. 2016;6(2):79–84. doi: 10.4103/2045-9912.184717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Grayson M. Parkinson’s disease. Nature. 2016;538(7626):S1. doi: 10.1038/538S1a. [DOI] [PubMed] [Google Scholar]
- 93.Huntley G.W., Benson D.L. Origins of Parkinson’s disease in brain development: insights from early and persistent effects of LRRK2-G2019S on striatal circuits. Front. Neurosci. 2020;14:265. doi: 10.3389/fnins.2020.00265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kalia L.V., Lang A.E. Parkinson’s disease. Lancet. 2015;386(9996):896–912. doi: 10.1016/S0140-6736(14)61393-3. [DOI] [PubMed] [Google Scholar]
- 95.Bruck A., Aalto S., Nurmi E., Bergman J., Rinne J.O. Cortical 6-[18F]fluoro-L-dopa uptake and frontal cognitive functions in early Parkinson’s disease. Neurobiol. Aging. 2005;26(6):891–898. doi: 10.1016/j.neurobiolaging.2004.07.014. [DOI] [PubMed] [Google Scholar]
- 96.Bohnen N.I., Albin R.L. The cholinergic system and Parkinson disease. Behav. Brain Res. 2011;221(2):564–573. doi: 10.1016/j.bbr.2009.12.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Perry E.K., Curtis M., Dick D.J., Candy J.M., Atack J.R., Bloxham C.A., Blessed G., Fairbairn A., Tomlinson B.E., Perry R.H. Cholinergic correlates of cognitive impairment in Parkinson’s disease: comparisons with Alzheimer’s disease. J. Neurol. Neurosurg. Psychiatry. 1985;48(5):413–421. doi: 10.1136/jnnp.48.5.413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Fang C., Lv L. Cognition Deficits in Parkinson’s disease: mechanisms and treatment. Parkinsons Dis. 2020;2020:2076942. doi: 10.1155/2020/2076942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Hidalgo-Agudo R.D., Lucena-Anton D. Additional physical interventions to conventional physical therapy in Parkinson’s disease: a systematic review and meta-analysis of randomized clinical trials. J. Clin. Med. 2020;9(4):1038. doi: 10.3390/jcm9041038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Bellucci A., Bubacco L., Longhena F., Parrella E., Faustini G., Porrini V., Bono F., Missale C., Pizzi M. Nuclear factor-kappa b dysregulation and alpha-synuclein pathology: critical interplay in the pathogenesis of Parkinson’s disease. Front. Aging Neurosci. 2020;12:68. doi: 10.3389/fnagi.2020.00068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Gong Q.H., Shi X.R., Hong Z.Y., Pan L.L., Liu X.H., Zhu Y.Z. A new hope for neurodegeneration: possible role of hydrogen sulfide. J. Alzheimers Dis. 2011;24(Suppl. 2):173–182. doi: 10.3233/JAD-2011-110128. [DOI] [PubMed] [Google Scholar]
- 102.Yuan Y.Q., Wang Y.L., Yuan B.S., Yuan X., Hou X.O., Bian J.S., Liu C.F., Hu L.F. Impaired CBS-H2S signaling axis contributes to MPTP-induced neurodegeneration in a mouse model of Parkinson’s disease. Brain Behav. Immun. 2018;67:77–90. doi: 10.1016/j.bbi.2017.07.159. [DOI] [PubMed] [Google Scholar]
- 103.Hu L.F., Lu M., Tiong C.X., Dawe G.S., Hu G., Bian J.S. Neuroprotective effects of hydrogen sulfide on Parkinson’s disease rat models. Aging Cell. 2010;9(2):135–146. doi: 10.1111/j.1474-9726.2009.00543.x. [DOI] [PubMed] [Google Scholar]
- 104.Hou X., Yuan Y., Sheng Y., Yuan B., Wang Y., Zheng J., Liu C.F., Zhang X., Hu L.F. GYY4137, an H2S Slow-Releasing Donor, Prevents Nitrative Stress and alpha-Synuclein Nitration in an MPTP Mouse Model of Parkinson’s Disease. Front. Pharmacol. 2017;8:741. doi: 10.3389/fphar.2017.00741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Xie L., Hu L.F., Teo X.Q., Tiong C.X., Tazzari V., Sparatore A., Del Soldato P., Dawe G.S., Bian J.S. Therapeutic effect of hydrogen sulfide-releasing L-Dopa derivative ACS84 on 6-OHDA-induced Parkinson’s disease rat model. PLoS One. 2013;8(4):e60200. doi: 10.1371/journal.pone.0060200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Hu L.F., Lu M., Wu Z.Y., Wong P.T., Bian J.S. Hydrogen sulfide inhibits rotenone-induced apoptosis via preservation of mitochondrial function. Mol. Pharmacol. 2009;75(1):27–34. doi: 10.1124/mol.108.047985. [DOI] [PubMed] [Google Scholar]
- 107.Shefa U., Kim M.S., Jeong N.Y. Antioxidant and Cell-Signaling Functions of Hydrogen Sulfide in the Central Nervous System. Oxid. Med. Cell. Longev. 2018;2018:1873962. doi: 10.1155/2018/1873962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Tabassum R., Jeong N.Y. Potential for therapeutic use of hydrogen sulfide in oxidative stress-induced neurodegenerative diseases. Int. J. Med. Sci. 2019;16(10):1386–1396. doi: 10.7150/ijms.36516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Kida K., Ichinose F. Hydrogen sulfide and neuroinflammation. Handb. Exp. Pharmacol. 2015;230:181–189. doi: 10.1007/978-3-319-18144-8_9. [DOI] [PubMed] [Google Scholar]
- 110.Yang S.Q., Tian Q., Li D., He S.Q., Hu M., Liu S.Y., Zou W., Chen Y.J., Zhang P., Tang X.Q. Leptin mediates protection of hydrogen sulfide against 6-hydroxydopamine-induced Parkinson’s disease: Involving enhancement in Warburg effect. Neurochem. Int. 2020;135:104692. doi: 10.1016/j.neuint.2020.104692. [DOI] [PubMed] [Google Scholar]
- 111.Chia S.J., Tan E.K., Chao Y.X. Historical perspective: models of Parkinson’s disease. Int. J. Mol. Sci. 2020;21(7):2464. doi: 10.3390/ijms21072464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kunath T., Natalwala A., Chan C., Chen Y., Stecher B., Taylor M., Khan S., Muqit M.M.K. Are PARKIN patients ideal candidates for dopaminergic cell replacement therapies? Eur. J. Neurosci. 2019;49(4):453–462. doi: 10.1111/ejn.14314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Chung K.K., Thomas B., Li X., Pletnikova O., Troncoso J.C., Marsh L., Dawson V.L., Dawson T.M. S-nitrosylation of parkin regulates ubiquitination and compromises parkin’s protective function. Science. 2004;304(5675):1328–1331. doi: 10.1126/science.1093891. [DOI] [PubMed] [Google Scholar]
- 114.Lizama-Manibusan B., McLaughlin B. Redox modification of proteins as essential mediators of CNS autophagy and mitophagy. FEBS Lett. 2013;587(15):2291–2298. doi: 10.1016/j.febslet.2013.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Savino C., Pelicci P., Giorgio M. The P66Shc/mitochondrial permeability transition pore pathway determines neurodegeneration. Oxid. Med. Cell. Longev. 2013;2013:719407. doi: 10.1155/2013/719407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Yamamori T., Mizobata A., Saito Y., Urano Y., Inanami O., Irani K., Noguchi N. Phosphorylation of p66shc mediates 6-hydroxydopamine cytotoxicity. Free Radic. Res. 2011;45(3):342–350. doi: 10.3109/10715762.2010.532496. [DOI] [PubMed] [Google Scholar]
- 117.Xie Z.Z., Shi M.M., Xie L., Wu Z.Y., Li G., Hua F., Bian J.S. Sulfhydration of p66Shc at cysteine59 mediates the antioxidant effect of hydrogen sulfide. Antioxid. Redox Signal. 2014;21(18):2531–2542. doi: 10.1089/ars.2013.5604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Lotankar S., Prabhavalkar K.S., Bhatt L.K. Biomarkers for Parkinson’s disease: recent advancement. Neurosci. Bull. 2017;33(5):585–597. doi: 10.1007/s12264-017-0183-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Lieberman A.P., Shakkottai V.G., Albin R.L. Polyglutamine repeats in neurodegenerative diseases. Annu. Rev. Pathol. 2019;14:1–27. doi: 10.1146/annurev-pathmechdis-012418-012857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Pandey M., Rajamma U. Huntington’s disease: the coming of age. J. Genet. 2018;97(3):649–664. doi: 10.1007/s12041-018-0957-1. [DOI] [PubMed] [Google Scholar]
- 121.Soares T.R., Reis S.D., Pinho B.R., Duchen M.R., Oliveira J.M.A. Targeting the proteostasis network in Huntington’s disease. Ageing Res. Rev. 2019;49:92–103. doi: 10.1016/j.arr.2018.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Ishii I., Akahoshi N., Yu X.N., Kobayashi Y., Namekata K., Komaki G., Kimura H. Murine cystathionine gamma-lyase: complete cDNA and genomic sequences, promoter activity, tissue distribution and developmental expression. Biochem. J. 2004;381(Pt 1):113–123. doi: 10.1042/BJ20040243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Yang G., Pei Y., Teng H., Cao Q., Wang R. Specificity protein-1 as a critical regulator of human cystathionine gamma-lyase in smooth muscle cells. J. Biol. Chem. 2011;286(30):26450–26460. doi: 10.1074/jbc.M111.266643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Dunah A.W., Jeong H., Griffin A., Kim Y.M., Standaert D.G., Hersch S.M., Mouradian M.M., Young A.B., Tanese N., Krainc D. Sp1 and TAFII130 transcriptional activity disrupted in early Huntington’s disease. Science. 2002;296(5576):2238–2243. doi: 10.1126/science.1072613. [DOI] [PubMed] [Google Scholar]
- 125.Paul B.D., Sbodio J.I., Xu R., Vandiver M.S., Cha J.Y., Snowman A.M., Snyder S.H. Cystathionine gamma-lyase deficiency mediates neurodegeneration in Huntington’s disease. Nature. 2014;509(7498):96–100. doi: 10.1038/nature13136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Paul B.D., Snyder S.H. Neurodegeneration in Huntington’s disease involves loss of cystathionine gamma-lyase. Cell Cycle. 2014;13(16):2491–2493. doi: 10.4161/15384101.2014.950538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Li X., Valencia A., McClory H., Sapp E., Kegel K.B., Difiglia M. Deficient Rab11 activity underlies glucose hypometabolism in primary neurons of Huntington’s disease mice. Biochem. Biophys. Res. Commun. 2012;421(4):727–730. doi: 10.1016/j.bbrc.2012.04.070. [DOI] [PubMed] [Google Scholar]
- 128.Frederick N.M., Bertho J., Patel K.K., Petr G.T., Bakradze E., Smith S.B., Rosenberg P.A. Dysregulation of system xc(-) expression induced by mutant huntingtin in a striatal neuronal cell line and in R6/2 mice. Neurochem. Int. 2014;76:59–69. doi: 10.1016/j.neuint.2014.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Sbodio J.I., Snyder S.H., Paul B.D. Transcriptional control of amino acid homeostasis is disrupted in Huntington’s disease. Proc. Natl. Acad. Sci. USA. 2016;113(31):8843–8848. doi: 10.1073/pnas.1608264113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Snijder P.M., Baratashvili M., Grzeschik N.A., Leuvenink H.G.D., Kuijpers L., Huitema S., Schaap O., Giepmans B.N.G., Kuipers J., Miljkovic J.L., Mitrovic A., Bos E.M., Szabo C., Kampinga H.H., Dijkers P.F., Bos E.M., Szabo C., Kampinga H.H., Dijkers P.F., Dunnen W., Filipovic M.R., Goor H.V., Sibon O.C.M. Overexpression of cystathionine gamma-lyase suppresses detrimental effects of spinocerebellar ataxia type. Mol. Med. 2016;21(1):758–768. doi: 10.2119/molmed.2015.00221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Fessel J. Alzheimer’s disease combination treatment. Neurobiol. Aging. 2018;63:165. doi: 10.1016/j.neurobiolaging.2017.10.022. [DOI] [PubMed] [Google Scholar]
- 132.Krantic S. Editorial: From current diagnostic tools and therapeutics for Alzheimer’s disease towards earlier diagnostic markers and treatment targets. Curr. Alzheimer Res. 2017;14(1):2–5. doi: 10.2174/156720501401161201104858. [DOI] [PubMed] [Google Scholar]
- 133.Rad S.K., Arya A., Karimian H., Madhavan P., Rizwan F., Koshy S., Prabhu G. Mechanism involved in insulin resistance via accumulation of beta-amyloid and neurofibrillary tangles: link between type 2 diabetes and Alzheimer’s disease. Drug Des. Devel. Ther. 2018;12:3999–4021. doi: 10.2147/DDDT.S173970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Metaxas A., Kempf S.J. Neurofibrillary tangles in Alzheimer’s disease: elucidation of the molecular mechanism by immunohistochemistry and tau protein phospho-proteomics. Neural Regen. Res. 2016;11(10):1579–1581. doi: 10.4103/1673-5374.193234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Schwarzman A.L., Sarantseva S.V. Transmission of pathogenic protein aggregates in Alzheimer’s disease. Mol. Biol. (Mosk.) 2017;51(3):418–422. doi: 10.7868/S0026898417030144. [DOI] [PubMed] [Google Scholar]
- 136.Brody D.L., Jiang H., Wildburger N., Esparza T.J. Non-canonical soluble amyloid-beta aggregates and plaque buffering: controversies and future directions for target discovery in Alzheimer’s disease. Alzheimers Res. Ther. 2017;9(1):62. doi: 10.1186/s13195-017-0293-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hunter S., Brayne C. Understanding the roles of mutations in the amyloid precursor protein in Alzheimer disease. Mol. Psychiatry. 2018;23(1):81–93. doi: 10.1038/mp.2017.218. [DOI] [PubMed] [Google Scholar]
- 138.Haass C., Kaether C., Thinakaran G., Sisodia S. Trafficking and proteolytic processing of APP. Cold Spring Harb. Perspect. Med. 2012;2(5):a006270. doi: 10.1101/cshperspect.a006270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Cadet J., Ravanat J.L., Martinez G.R., Medeiros M.H., Di Mascio P. Singlet oxygen oxidation of isolated and cellular DNA: product formation and mechanistic insights. Photochem. Photobiol. 2006;82(5):1219–1225. doi: 10.1562/2006-06-09-IR-914. [DOI] [PubMed] [Google Scholar]
- 140.Butterfield D.A., Boyd-Kimball D. Amyloid beta-peptide(1-42) contributes to the oxidative stress and neurodegeneration found in Alzheimer disease brain. Brain Pathol. 2004;14(4):426–432. doi: 10.1111/j.1750-3639.2004.tb00087.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Butterfield D.A., Boyd-Kimball D. Oxidative stress, Amyloid-beta Peptide, and altered key molecular pathways in the pathogenesis and progression of Alzheimer’s disease. J. Alzheimers Dis. 2018;62(3):1345–1367. doi: 10.3233/JAD-170543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Mecocci P., Boccardi V., Cecchetti R., Bastiani P., Scamosci M., Ruggiero C., Baroni M. A long journey into aging, brain aging, and Alzheimer’s disease following the oxidative stress Tracks. J. Alzheimers Dis. 2018;62(3):1319–1335. doi: 10.3233/JAD-170732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Tonnies E., Trushina E. Oxidative stress, synaptic dysfunction, and Alzheimer’s disease. J. Alzheimers Dis. 2017;57(4):1105–1121. doi: 10.3233/JAD-161088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Radi E., Formichi P., Battisti C., Federico A. Apoptosis and oxidative stress in neurodegenerative diseases. J. Alzheimers Dis. 2014;42(Suppl. 3):S125–S152. doi: 10.3233/JAD-132738. [DOI] [PubMed] [Google Scholar]
- 145.Bradley-Whitman M.A., Timmons M.D., Beckett T.L., Murphy M.P., Lynn B.C., Lovell M.A. Nucleic acid oxidation: an early feature of Alzheimer’s disease. J. Neurochem. 2014;128(2):294–304. doi: 10.1111/jnc.12444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Beltowski J. Hydrogen sulfide in pharmacology and medicine--An update. Pharmacol. Rep. 2015;67(3):647–658. doi: 10.1016/j.pharep.2015.01.005. [DOI] [PubMed] [Google Scholar]
- 147.Vandini E., Ottani A., Zaffe D., Calevro A., Canalini F., Cavallini G.M., Rossi R., Guarini S., Giuliani D. Mechanisms of hydrogen sulfide against the progression of severe Alzheimer’s disease in transgenic mice at different ages. Pharmacology. 2019;103(1-2):50–60. doi: 10.1159/000494113. [DOI] [PubMed] [Google Scholar]
- 148.Kumar M., Sandhir R. Hydrogen sulfide in physiological and pathological mechanisms in brain. CNS Neurol. Disord. Drug Targets. 2018;17(9):654–670. doi: 10.2174/1871527317666180605072018. [DOI] [PubMed] [Google Scholar]
- 149.Clarke R., Smith A.D., Jobst K.A., Refsum H., Sutton L., Ueland P.M. Folate, vitamin B12, and serum total homocysteine levels in confirmed Alzheimer disease. Arch. Neurol. 1998;55(11):1449–1455. doi: 10.1001/archneur.55.11.1449. [DOI] [PubMed] [Google Scholar]
- 150.Dwyer B.E., Raina A.K., Perry G., Smith M.A. Homocysteine and Alzheimer’s disease: a modifiable risk? Free Radic. Biol. Med. 2004;36(11):1471–1475. doi: 10.1016/j.freeradbiomed.2004.03.009. [DOI] [PubMed] [Google Scholar]
- 151.Liu X.Q., Liu X.Q., Jiang P., Huang H., Yan Y. Plasma levels of endogenous hydrogen sulfide and homocysteine in patients with Alzheimer’s disease and vascular dementia and the significance thereof. Zhonghua Yi Xue Za Zhi. 2008;88(32):2246–2249. [PubMed] [Google Scholar]
- 152.Kamat P.K., Kyles P., Kalani A., Tyagi N. Hydrogen sulfide ameliorates homocysteine-induced Alzheimer’s disease-like pathology, blood-brain barrier disruption, and synaptic disorder. Mol. Neurobiol. 2016;53(4):2451–2467. doi: 10.1007/s12035-015-9212-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Ingenbleek Y., Bernstein L.H. Downsizing of lean body mass is a key determinant of Alzheimer’s disease. J. Alzheimers Dis. 2015;44(3):745–754. doi: 10.3233/JAD-141950. [DOI] [PubMed] [Google Scholar]
- 154.Schreier S.M., Muellner M.K., Steinkellner H., Hermann M., Esterbauer H., Exner M., Gmeiner B.M., Kapiotis S., Laggner H. Hydrogen sulfide scavenges the cytotoxic lipid oxidation product 4-HNE. Neurotox. Res. 2010;17(3):249–256. doi: 10.1007/s12640-009-9099-9. [DOI] [PubMed] [Google Scholar]
- 155.Zarkovic K. 4-hydroxynonenal and neurodegenerative diseases. Mol. Aspects Med. 2003;24(4-5):293–303. doi: 10.1016/S0098-2997(03)00024-4. [DOI] [PubMed] [Google Scholar]
- 156.Islam M.T. Oxidative stress and mitochondrial dysfunction-linked neurodegenerative disorders. Neurol. Res. 2017;39(1):73–82. doi: 10.1080/01616412.2016.1251711. [DOI] [PubMed] [Google Scholar]
- 157.Xuan A., Long D., Li J., Ji W., Zhang M., Hong L., Liu J. Hydrogen sulfide attenuates spatial memory impairment and hippocampal neuroinflammation in beta-amyloid rat model of Alzheimer’s disease. J. Neuroinflammation. 2012;9:202. doi: 10.1186/1742-2094-9-202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Giuliani D., Ottani A., Zaffe D., Galantucci M., Strinati F., Lodi R., Guarini S. Hydrogen sulfide slows down progression of experimental Alzheimer’s disease by targeting multiple pathophysiological mechanisms. Neurobiol. Learn. Mem. 2013;104:82–91. doi: 10.1016/j.nlm.2013.05.006. [DOI] [PubMed] [Google Scholar]
- 159.Nagpure B.V., Bian J.S. Hydrogen sulfide inhibits A2A adenosine receptor agonist induced beta-amyloid production in SH-SY5Y neuroblastoma cells via a cAMP dependent pathway. PLoS One. 2014;9(2):e88508. doi: 10.1371/journal.pone.0088508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Liu Y.Y., Bian J.S. Hydrogen sulfide protects amyloid-beta induced cell toxicity in microglia. J. Alzheimers Dis. 2010;22(4):1189–1200. doi: 10.3233/JAD-2010-101002. [DOI] [PubMed] [Google Scholar]
- 161.Selkoe D.J. Amyloid protein and Alzheimer's disease. Sci. Am. 1991;265(5):68–71. doi: 10.1038/scientificamerican1191-68. [DOI] [PubMed] [Google Scholar]
- 162.Zhang H., Gao Y., Zhao F., Dai Z., Meng T., Tu S., Yan Y. Hydrogen sulfide reduces mRNA and protein levels of beta-site amyloid precursor protein cleaving enzyme 1 in PC12 cells. Neurochem. Int. 2011;58(2):169–175. doi: 10.1016/j.neuint.2010.11.010. [DOI] [PubMed] [Google Scholar]
- 163.Cai Z., Hussain M.D., Yan L.J. Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer’s disease. Int. J. Neurosci. 2014;124(5):307–321. doi: 10.3109/00207454.2013.833510. [DOI] [PubMed] [Google Scholar]
- 164.Doens D., Fernandez P.L. Microglia receptors and their implications in the response to amyloid beta for Alzheimer’s disease pathogenesis. J. Neuroinflammation. 2014;11:48. doi: 10.1186/1742-2094-11-48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Reichenbach N., Delekate A., Plescher M., Schmitt F., Krauss S., Blank N., Halle A., Petzold G.C. Inhibition of Stat3-mediated astrogliosis ameliorates pathology in an Alzheimer’s disease model. EMBO Mol. Med. 2019;11(2):e9665. doi: 10.15252/emmm.201809665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Chandra S.R. Alzheimer’s disease: An alternative approach. Indian J. Med. Res. 2017;145(6):723–729. doi: 10.4103/ijmr.IJMR_74_17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Schechter I., Ziv E., Cathepsins S. B and L with aminopeptidases display beta-secretase activity associated with the pathogenesis of Alzheimer’s disease. Biol. Chem. 2011;392(6):555–569. doi: 10.1515/bc.2011.054. [DOI] [PubMed] [Google Scholar]
- 168.Lowry J.R., Klegeris A. Emerging roles of microglial cathepsins in neurodegenerative disease. Brain Res. Bull. 2018;139:144–156. doi: 10.1016/j.brainresbull.2018.02.014. [DOI] [PubMed] [Google Scholar]
- 169.Cao L., Cao X., Zhou Y., Nagpure B.V., Wu Z.Y., Hu L.F., Yang Y., Sethi G., Moore P.K., Bian J.S. Hydrogen sulfide inhibits ATP-induced neuroinflammation and Abeta1-42 synthesis by suppressing the activation of STAT3 and cathepsin S. Brain Behav. Immun. 2018;73:603–614. doi: 10.1016/j.bbi.2018.07.005. [DOI] [PubMed] [Google Scholar]
- 170.Sen T., Saha P., Jiang T., Sen N. Sulfhydration of AKT triggers Tau-phosphorylation by activating glycogen synthase kinase 3beta in Alzheimer’s disease. Proc. Natl. Acad. Sci. USA. 2020;117(8):4418–4427. doi: 10.1073/pnas.1916895117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Whiteman M., Cheung N.S., Zhu Y.Z., Chu S.H., Siau J.L., Wong B.S., Armstrong J.S., Moore P.K. Hydrogen sulphide: a novel inhibitor of hypochlorous acid-mediated oxidative damage in the brain? Biochem. Biophys. Res. Commun. 2005;326(4):794–798. doi: 10.1016/j.bbrc.2004.11.110. [DOI] [PubMed] [Google Scholar]
- 172.Whiteman M., Armstrong J.S., Chu S.H., Jia-Ling S., Wong B.S., Cheung N.S., Halliwell B., Moore P.K. The novel neuromodulator hydrogen sulfide: an endogenous peroxynitrite ‘scavenger’? J. Neurochem. 2004;90(3):765–768. doi: 10.1111/j.1471-4159.2004.02617.x. [DOI] [PubMed] [Google Scholar]
- 173.Kimura Y., Goto Y., Kimura H. Hydrogen sulfide increases glutathione production and suppresses oxidative stress in mitochondria. Antioxid. Redox Signal. 2010;12(1):1–13. doi: 10.1089/ars.2008.2282. [DOI] [PubMed] [Google Scholar]
- 174.Chen Y., Zhou C.F., Xiao F., Huang H.L., Zhang P., Gu H.F., Tang X.Q. Inhibition of ALDH2 protects PC12 cells against formaldehyde-induced cytotoxicity: involving the protection of hydrogen sulphide. Clin. Exp. Pharmacol. Physiol. 2017;44(5):595–601. doi: 10.1111/1440-1681.12741. [DOI] [PubMed] [Google Scholar]
- 175.Xin L., Junhua W., Long L., Jun Y., Yang X. Exogenous hydrogen sulfide protects SH-SY5Y cells from OGD/RInduced Injury. Curr. Mol. Med. 2017;17(8):563–567. doi: 10.2174/1566524018666180222121643. [DOI] [PubMed] [Google Scholar]
- 176.Yang G., Zhao K., Ju Y., Mani S., Cao Q., Puukila S., Khaper N., Wu L., Wang R. Hydrogen sulfide protects against cellular senescence via S-sulfhydration of Keap1 and activation of Nrf2. Antioxid. Redox Signal. 2013;18(15):1906–1919. doi: 10.1089/ars.2012.4645. [DOI] [PubMed] [Google Scholar]
- 177.Huang P., Shen Z., Liu J., Huang Y., Chen S., Yu W., Wang S., Ren Y., Li X., Tang C., Du J., Jin H. Hydrogen sulfide inhibits high-salt diet-induced renal oxidative stress and kidney injury in dahl rats. Oxid. Med. Cell. Longev. 2016;2016:2807490. doi: 10.1155/2016/2807490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Xie L., Gu Y., Wen M., Zhao S., Wang W., Ma Y., Meng G., Han Y., Wang Y., Liu G., Moore P.K., Wang X., Wang H., Zhang Z., Yu Y., Ferro A., Huang Z., Ji Y. Hydrogen sulfide induces keap1 s-sulfhydration and suppresses diabetes-accelerated atherosclerosis via nrf2 activation. Diabetes. 2016;65(10):3171–3184. doi: 10.2337/db16-0020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Meng W., Pei Z., Feng Y., Zhao J., Chen Y., Shi W., Xu Q., Lin F., Sun M., Xiao K. Neglected role of hydrogen sulfide in sulfur mustard poisoning: Keap1 S-sulfhydration and subsequent Nrf2 pathway activation. Sci. Rep. 2017;7(1):9433. doi: 10.1038/s41598-017-09648-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Liu Z., Wang X., Li L., Wei G., Zhao M. Hydrogen sulfide protects against paraquat-induced acute liver injury in rats by regulating oxidative stress, mitochondrial function, and inflammation. Oxid. Med. Cell. Longev. 2020;2020:6325378. doi: 10.1155/2020/6325378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Zhao S., Song T., Gu Y., Zhang Y., Cao S., Miao Q., Zhang X., Chen H., Gao Y., Zhang L., Han Y., Wang H., Pu J., Xie L., Ji Y. Hydrogen sulfide alleviates liver injury via S-sulfhydrated-Keap1/Nrf2/LRP1 pathway. Hepatology. 2020;73(1):282–302. doi: 10.1002/hep.31247. [DOI] [PubMed] [Google Scholar]
- 182.Guo C., Liang F., Shah Masood W., Yan X. Hydrogen sulfide protected gastric epithelial cell from ischemia/reperfusion injury by Keap1 s-sulfhydration, MAPK dependent anti-apoptosis and NF-kappaB dependent anti-inflammation pathway. Eur. J. Pharmacol. 2014;725:70–78. doi: 10.1016/j.ejphar.2014.01.009. [DOI] [PubMed] [Google Scholar]
- 183.Ceprian M., Fulton D. Glial cell AMPA receptors in nervous system health, injury and disease. Int. J. Mol. Sci. 2019;20(10):2450. doi: 10.3390/ijms20102450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Sekhar V.C., Viswanathan G., Baby S. Insights into the molecular aspects of neuroprotective bacoside A and Bacopaside I. Curr. Neuropharmacol. 2019;17(5):438–446. doi: 10.2174/1570159X16666180419123022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Yan Y., Yang H., Xie Y., Ding Y., Kong D., Yu H. Research progress on Alzheimer’s disease and resveratrol. Neurochem. Res. 2020;45(5):989–1006. doi: 10.1007/s11064-020-03007-0. [DOI] [PubMed] [Google Scholar]
- 186.Zhang Y., Guo O., Huo Y., Wang G., Man H.Y. Amyloid-beta induces AMPA receptor ubiquitination and degradation in primary neurons and human brains of Alzheimer’s disease. J. Alzheimers Dis. 2018;62(4):1789–1801. doi: 10.3233/JAD-170879. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Gao L., Tian M., Zhao H.Y., Xu Q.Q., Huang Y.M., Si Q.C., Tian Q., Wu Q.M., Hu X.M., Sun L.B., McClintock S.M., Zeng Y. TrkB activation by 7, 8-dihydroxyflavone increases synapse AMPA subunits and ameliorates spatial memory deficits in a mouse model of Alzheimer’s disease. J. Neurochem. 2016;136(3):620–636. doi: 10.1111/jnc.13432. [DOI] [PubMed] [Google Scholar]
- 188.Du Y., Fu M., Huang Z., Tian X., Li J., Pang Y., Song W., Wang T. Y.; Dong, Z. TRPV1 activation alleviates cognitive and synaptic plasticity impairments through inhibiting AMPAR endocytosis in APP23/PS45 mouse model of Alzheimer’s disease. Aging Cell. 2020;19(3):e13113. doi: 10.1111/acel.13113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Li Y.L., Zhou J., Zhang H., Luo Y., Long L.H., Hu Z.L., Chen J.G., Wang F., Wu P.F. Hydrogen sulfide promotes surface insertion of hippocampal AMPA Receptor GluR1 subunit via Phosphorylating at Serine-831/Serine-845 sites through a sulfhydration-dependent mechanism. CNS Neurosci. Ther. 2016;22(9):789–798. doi: 10.1111/cns.12585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Sodhi R.K., Singh N. Liver X receptors: emerging therapeutic targets for Alzheimer’s disease. Pharmacol. Res. 2013;72:45–51. doi: 10.1016/j.phrs.2013.03.008. [DOI] [PubMed] [Google Scholar]
- 191.Cotman C.W., Berchtold N.C., Christie L.A. Exercise builds brain health: key roles of growth factor cascades and inflammation. Trends Neurosci. 2007;30(9):464–472. doi: 10.1016/j.tins.2007.06.011. [DOI] [PubMed] [Google Scholar]
- 192.Nyberg L. Neuroimaging in aging: brain maintenance. F1000 Res. 2017;6:1215. doi: 10.12688/f1000research.11419.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Nyberg L., Pudas S. Successful memory aging. Annu. Rev. Psychol. 2019;70:219–243. doi: 10.1146/annurev-psych-010418-103052. [DOI] [PubMed] [Google Scholar]
- 194.Coleman L.G., Jr, Zou J., Qin L., Crews F.T. HMGB1/IL-1beta complexes regulate neuroimmune responses in alcoholism. Brain Behav. Immun. 2018;72:61–77. doi: 10.1016/j.bbi.2017.10.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Patterson S.L. Immune dysregulation and cognitive vulnerability in the aging brain: Interactions of microglia, IL-1beta, BDNF and synaptic plasticity. Neuropharmacology. 2015;96(Pt A):11–18. doi: 10.1016/j.neuropharm.2014.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Ralay Ranaivo H., Craft J.M., Hu W., Guo L., Wing L.K., Van Eldik L.J., Watterson D.M. Glia as a therapeutic target: selective suppression of human amyloid-beta-induced upregulation of brain proinflammatory cytokine production attenuates neurodegeneration. J. Neurosci. 2006;26(2):662–670. doi: 10.1523/JNEUROSCI.4652-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Han Q., Lin Q., Huang P., Chen M., Hu X., Fu H., He S., Shen F., Zeng H., Deng Y. Microglia-derived IL-1beta contributes to axon development disorders and synaptic deficit through p38-MAPK signal pathway in septic neonatal rats. J. Neuroinflammation. 2017;14(1):52. doi: 10.1186/s12974-017-0805-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Jones M.E., Paniccia J.E., Lebonville C.L., Reissner K.J., Lysle D.T. Chemogenetic manipulation of dorsal hippocampal astrocytes protects against the development of stress-enhanced fear learning. Neuroscience. 2018;388:45–56. doi: 10.1016/j.neuroscience.2018.07.015. [DOI] [PubMed] [Google Scholar]
- 199.Zhu X., Liu J., Chen S., Xue J., Huang S., Wang Y., Chen O. Isoliquiritigenin attenuates lipopolysaccharide-induced cognitive impairment through antioxidant and anti-inflammatory activity. BMC Neurosci. 2019;20(1):41. doi: 10.1186/s12868-019-0520-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Mir S., Sen T., Sen N. Cytokine-induced GAPDH sulfhydration affects PSD95 degradation and memory. Mol. Cell. 2014;56(6):786–795. doi: 10.1016/j.molcel.2014.10.019. [DOI] [PubMed] [Google Scholar]
- 201.Cai F., Wang F., Lin F.K., Liu C., Ma L.Q., Liu J., Wu W.N., Wang W., Wang J.H., Chen J.G. Redox modulation of long-term potentiation in the hippocampus via regulation of the glycogen synthase kinase-3beta pathway. Free Radic. Biol. Med. 2008;45(7):964–970. doi: 10.1016/j.freeradbiomed.2008.06.014. [DOI] [PubMed] [Google Scholar]
- 202.Kamsler A., Segal M. Hydrogen peroxide modulation of synaptic plasticity. J. Neurosci. 2003;23(1):269–276. doi: 10.1523/JNEUROSCI.23-01-00269.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Zhang C., Yi F., Xia M., Boini K.M., Zhu Q., Laperle L.A., Abais J.M., Brimson C.A., Li P.L. NMDA receptor-mediated activation of NADPH oxidase and glomerulosclerosis in hyperhomocysteinemic rats. Antioxid. Redox Signal. 2010;13(7):975–986. doi: 10.1089/ars.2010.3091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Bodhinathan K., Kumar A., Foster T.C. Intracellular redox state alters NMDA receptor response during aging through Ca2+/calmodulin-dependent protein kinase II. J. Neurosci. 2010;30(5):1914–1924. doi: 10.1523/JNEUROSCI.5485-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Wang Y., Guan X.L., Wu P.F., Wang C.M., Cao H., Li L., Guo X.J., Wang F., Xie N., Jiang F.C., Chen J.G. Multifunctional mercapto-tacrine derivatives for treatment of age-related neurodegenerative diseases. J. Med. Chem. 2012;55(7):3588–3592. doi: 10.1021/jm300124p. [DOI] [PubMed] [Google Scholar]
- 206.Yang Y.J., Wu P.F., Long L.H., Yu D.F., Wu W.N., Hu Z.L., Fu H., Xie N., Jin Y., Ni L., Wang J.Z., Wang F., Chen J.G. Reversal of aging-associated hippocampal synaptic plasticity deficits by reductants via regulation of thiol redox and NMDA receptor function. Aging Cell. 2010;9(5):709–721. doi: 10.1111/j.1474-9726.2010.00595.x. [DOI] [PubMed] [Google Scholar]
- 207.Henneberger C., Papouin T., Oliet S.H., Rusakov D.A. Long-term potentiation depends on release of D-serine from astrocytes. Nature. 2010;463(7278):232–236. doi: 10.1038/nature08673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Panatier A., Theodosis D.T., Mothet J.P., Touquet B., Pollegioni L., Poulain D.A., Oliet S.H. Glia-derived D-serine controls NMDA receptor activity and synaptic memory. Cell. 2006;125(4):775–784. doi: 10.1016/j.cell.2006.02.051. [DOI] [PubMed] [Google Scholar]
- 209.Papouin T., Ladepeche L., Ruel J., Sacchi S., Labasque M., Hanini M., Groc L., Pollegioni L., Mothet J.P., Oliet S.H. Synaptic and extrasynaptic NMDA receptors are gated by different endogenous coagonists. Cell. 2012;150(3):633–646. doi: 10.1016/j.cell.2012.06.029. [DOI] [PubMed] [Google Scholar]
- 210.Wolosker H., Blackshaw S., Snyder S.H. Serine racemase: a glial enzyme synthesizing D-serine to regulate glutamate-N-methyl-D-aspartate neurotransmission. Proc. Natl. Acad. Sci. USA. 1999;96(23):13409–13414. doi: 10.1073/pnas.96.23.13409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Li Y.L., Wu P.F., Chen J.G., Wang S., Han Q.Q., Li D., Wang W., Guan X.L., Li D., Long L.H., Huang J.G., Wang F. Activity-dependent sulfhydration signal Controls N-Methyl-D-Aspartate subtype glutamate receptor-dependent synaptic plasticity via increasing d-serine availability. Antioxid. Redox Signal. 2017;27(7):398–414. doi: 10.1089/ars.2016.6936. [DOI] [PubMed] [Google Scholar]
- 212.Shigetomi E., Jackson-Weaver O., Huckstepp R.T., O’Dell T.J., Khakh B.S. TRPA1 channels are regulators of astrocyte basal calcium levels and long-term potentiation via constitutive D-serine release. J. Neurosci. 2013;33(24):10143–10153. doi: 10.1523/JNEUROSCI.5779-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Hatakeyama Y., Takahashi K., Tominaga M., Kimura H., Ohta T. Polysulfide evokes acute pain through the activation of nociceptive TRPA1 in mouse sensory neurons. Mol. Pain. 2015;11:24. doi: 10.1186/s12990-015-0023-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Taylor J.P., Brown R.H., Jr, Cleveland D.W. Decoding ALS: from genes to mechanism. Nature. 2016;539(7628):197–206. doi: 10.1038/nature20413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Weishaupt J.H., Hyman T., Dikic I. Common molecular pathways in amyotrophic lateral sclerosis and frontotemporal dementia. Trends Mol. Med. 2016;22(9):769–783. doi: 10.1016/j.molmed.2016.07.005. [DOI] [PubMed] [Google Scholar]
- 216.Gurney M.E., Pu H., Chiu A.Y., Dal Canto M.C., Polchow C.Y., Alexander D.D., Caliendo J., Hentati A., Kwon Y.W., Deng H.X., et al. Motor neuron degeneration in mice that express a human Cu,Zn superoxide dismutase mutation. Science. 1994;264(5166):1772–1775. doi: 10.1126/science.8209258. [DOI] [PubMed] [Google Scholar]
- 217.Rosen D.R., Siddique T., Patterson D., Figlewicz D.A., Sapp P., Hentati A., Donaldson D., Goto J., O’Regan J.P., Deng H.X., et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993;362(6415):59–62. doi: 10.1038/362059a0. [DOI] [PubMed] [Google Scholar]
- 218.Davoli A., Greco V., Spalloni A., Guatteo E., Neri C., Rizzo G.R., Cordella A., Romigi A., Cortese C., Bernardini S., Sarchielli P., Cardaioli G., Calabresi P., Mercuri N.B., Urbani A., Longone P. Evidence of hydrogen sulfide involvement in amyotrophic lateral sclerosis. Ann. Neurol. 2015;77(4):697–709. doi: 10.1002/ana.24372. [DOI] [PubMed] [Google Scholar]
- 219.Spalloni A., Greco V., Ciriminna G. Impact of pharmacological inhibition of hydrogen sulphide production in the SOD1G93A-ALS mouse model. Int. J. Mol. Sci. 2019;20(10):2550. doi: 10.3390/ijms20102550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Kamoun P., Belardinelli M.C., Chabli A., Lallouchi K., Chadefaux-Vekemans B. Endogenous hydrogen sulfide overproduction in Down syndrome. Am. J. Med. Genet. A. 2003;116a(3):310–311. doi: 10.1002/ajmg.a.10847. [DOI] [PubMed] [Google Scholar]
- 221.Belardinelli M.C., Chabli A., Chadefaux-Vekemans B., Kamoun P. Urinary sulfur compounds in Down syndrome. Clin. Chem. 2001;47(8):1500–1501. doi: 10.1093/clinchem/47.8.1500. [DOI] [PubMed] [Google Scholar]
- 222.Panagaki T., Randi E.B., Augsburger F., Szabo C. Overproduction of H2S, generated by CBS, inhibits mitochondrial Complex IV and suppresses oxidative phosphorylation in Down syndrome. Proc. Natl. Acad. Sci. USA. 2019;116(38):18769–18771. doi: 10.1073/pnas.1911895116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Fehervari Z. Glioma immune evasion. Nat. Immunol. 2017;18(5):487. doi: 10.1038/ni.3736. [DOI] [PubMed] [Google Scholar]
- 224.Dong Z., Cui H. The emerging roles of RNA modifications in glioblastoma. Cancers (Basel) 2020;12(3):736. doi: 10.3390/cancers12030736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Li Z.Y., Liu S.C., Xu P.J., Yang Z., Zhang T. Hydrogen sulfide stimulates the development of rat glioblastoma. Zhonghua Zhong Liu Za Zhi. 2012;34(4):254–258. doi: 10.3760/cma.j.issn.0253-3766.2012.04.004. [DOI] [PubMed] [Google Scholar]
- 226.Zhen Y., Zhang W., Liu C., He J., Lu Y., Guo R., Feng J., Zhang Y., Chen J. Exogenous hydrogen sulfide promotes C6 glioma cell growth through activation of the p38 MAPK/ERK1/2-COX-2 pathways. Oncol. Rep. 2015;34(5):2413–2422. doi: 10.3892/or.2015.4248. [DOI] [PubMed] [Google Scholar]
- 227.Zhao L., Wang Y., Yan Q., Lv W., Zhang Y., He S. Exogenous hydrogen sulfide exhibits anti-cancer effects though p38 MAPK signaling pathway in C6 glioma cells. Biol. Chem. 2015;396(11):1247–1253. doi: 10.1515/hsz-2015-0148. [DOI] [PubMed] [Google Scholar]
- 228.Szabo C., Ransy C., Modis K., Andriamihaja M., Murghes B., Coletta C., Olah G., Yanagi K., Bouillaud F. Regulation of mitochondrial bioenergetic function by hydrogen sulfide. Part I. Biochemical and physiological mechanisms. Br. J. Pharmacol. 2014;171(8):2099–2122. doi: 10.1111/bph.12369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Wrobel M., Czubak J., Bronowicka-Adamska P., Jurkowska H., Adamek D., Papla B. Is development of high-grade gliomas sulfur-dependent? Molecules. 2014;19(12):21350–21362. doi: 10.3390/molecules191221350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.capital Ka, C.o.C.L.I.; Boiko, N.M.; Panchuk, R.R.; Grytsyna, II; Klyuchivska, O.Y.; Biletska, L.P.; Lesyk, R.B.; Zsmall i, U.B.S.; Stoika, R.S. Putative anticancer potential of novel 4-thiazolidinone derivatives: cytotoxicity toward rat C6 glioma in vitro and correlation of general toxicity with the balance of free radical oxidation in rats. Croat. Med. J. 2016;57(2):151–163. doi: 10.3325/cmj.2016.57.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Wang G., Zhang T., Sun W., Wang H., Yin F., Wang Z., Zuo D., Sun M., Zhou Z., Lin B., Xu J., Hua Y., Li H., Cai Z. Arsenic sulfide induces apoptosis and autophagy through the activation of ROS/JNK and suppression of Akt/mTOR signaling pathways in osteosarcoma. Free Radic. Biol. Med. 2017;106:24–37. doi: 10.1016/j.freeradbiomed.2017.02.015. [DOI] [PubMed] [Google Scholar]
- 232.Sriram N., Kalayarasan S., Ashokkumar P., Sureshkumar A., Sudhandiran G. Diallyl sulfide induces apoptosis in Colo 320 DM human colon cancer cells: involvement of caspase-3, NF-kappaB, and ERK-2. Mol. Cell. Biochem. 2008;311(1-2):157–165. doi: 10.1007/s11010-008-9706-8. [DOI] [PubMed] [Google Scholar]
- 233.Kalpage H.A., Bazylianska V., Recanati M.A., Fite A., Liu J., Wan J., Mantena N., Malek M.H., Podgorski I., Heath E.I., Vaishnav A., Edwards B.F., Grossman L.I., Sanderson T.H., Lee I., Huttemann M. Tissue-specific regulation of cytochrome c by post-translational modifications: respiration, the mitochondrial membrane potential, ROS, and apoptosis. FASEB J. 2019;33(2):1540–1553. doi: 10.1096/fj.201801417R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Juman S., Nara Y., Yasui N., Negishi H., Okuda H., Takado N., Miki T. Reduced Production of Hydrogen Sulfide and Sulfane Sulfur Due to Low Cystathionine beta-Synthase Levels in Brain Astrocytes of Stroke-Prone Spontaneously Hypertensive Rats. Biol. Pharm. Bull. 2016;39(12):1932–1938. doi: 10.1248/bpb.b16-00334. [DOI] [PubMed] [Google Scholar]
- 235.Shiota M., Naya M., Yamamoto T. Gold-nanofeve surface-enhanced Raman spectroscopy visualizes hypotaurine as a robust anti-oxidant consumed in cancer survival. Nat. Commun. 2018;9(1):1561. doi: 10.1038/s41467-018-03899-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Kimura H. Signaling by hydrogen sulfide (H2S) and polysulfides (H2Sn) in the central nervous system. Neurochem. Int. 2019;126:118–125. doi: 10.1016/j.neuint.2019.01.027. [DOI] [PubMed] [Google Scholar]
- 237.Koike S., Ogasawara Y., Shibuya N., Kimura H., Ishii K. Polysulfide exerts a protective effect against cytotoxicity caused by t-buthylhydroperoxide through Nrf2 signaling in neuroblastoma cells. FEBS Lett. 2013;587(21):3548–3555. doi: 10.1016/j.febslet.2013.09.013. [DOI] [PubMed] [Google Scholar]
- 238.Jarosz A.P., Wei W., Gauld J.W., Auld J., Ozcan F., Aslan M., Mutus B. Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) is inactivated by S-sulfuration in vitro. Free Radic. Biol. Med. 2015;89:512–521. doi: 10.1016/j.freeradbiomed.2015.09.007. [DOI] [PubMed] [Google Scholar]
- 239.Stubbert D., Prysyazhna O., Rudyk O., Scotcher J., Burgoyne J.R., Eaton P. Protein kinase G Ialpha oxidation paradoxically underlies blood pressure lowering by the reductant hydrogen sulfide. Hypertension. 2014;64(6):1344–1351. doi: 10.1161/HYPERTENSIONAHA.114.04281. [DOI] [PubMed] [Google Scholar]
- 240.Paul B.D., Snyder S.H.H. (2)S signalling through protein sulfhydration and beyond. Nat. Rev. Mol. Cell Biol. 2012;13(8):499–507. doi: 10.1038/nrm3391. [DOI] [PubMed] [Google Scholar]
- 241.Shigetomi E., Tong X., Kwan K.Y., Corey D.P., Khakh B.S. TRPA1 channels regulate astrocyte resting calcium and inhibitory synapse efficacy through GAT-3. Nat. Neurosci. 2011;15(1):70–80. doi: 10.1038/nn.3000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Finelli M.J., Oliver P.L. TLDc proteins: new players in the oxidative stress response and neurological disease. Mamm. Genome. 2017;28(9-10):395–406. doi: 10.1007/s00335-017-9706-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Blesa J., Trigo-Damas I., Quiroga-Varela A., Jackson-Lewis V.R. Oxidative stress and Parkinson’s disease. Front. Neuroanat. 2015;9:91. doi: 10.3389/fnana.2015.00091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Joung E.J., Li M.H., Lee H.G., Somparn N., Jung Y.S., Na H.K., Kim S.H., Cha Y.N., Surh Y.J. Capsaicin induces heme oxygenase-1 expression in HepG2 cells via activation of PI3K-Nrf2 signaling: NAD(P)H:quinone oxidoreductase as a potential target. Antioxid. Redox Signal. 2007;9(12):2087–2098. doi: 10.1089/ars.2007.1827. [DOI] [PubMed] [Google Scholar]
- 245.Butera G., Pacchiana R., Mullappilly N., Margiotta M., Bruno S., Conti P., Riganti C., Donadelli M. Mutant p53 prevents GAPDH nuclear translocation in pancreatic cancer cells favoring glycolysis and 2-deoxyglucose sensitivity. Biochim. Biophys. Acta Mol. Cell Res. 2018;1865(12):1914–1923. doi: 10.1016/j.bbamcr.2018.10.005. [DOI] [PubMed] [Google Scholar]
- 246.Grillon E., Farion R., Reuveni M., Glidle A., Remy C., Coles J.A. Spatial profiles of markers of glycolysis, mitochondria, and proton pumps in a rat glioma suggest coordinated programming for proliferation. BMC Res. Notes. 2015;8:207. doi: 10.1186/s13104-015-1191-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Appelskog I.B., Ammerpohl O., Svechnikova I.G., Lui W.O., Almqvist P.M., Ekstrom T.J. Histone deacetylase inhibitor 4-phenylbutyrate suppresses GAPDH mRNA expression in glioma cells. Int. J. Oncol. 2004;24(6):1419–1425. [PubMed] [Google Scholar]
- 248.Dai S., Peng Y., Zhu Y., Xu D., Zhu F., Xu W., Chen Q., Zhu X., Liu T., Hou C., Wu J., Miao Y. Glycolysis promotes the progression of pancreatic cancer and reduces cancer cell sensitivity to gemcitabine. Biomed. Pharmacother. 2020;121:109521. doi: 10.1016/j.biopha.2019.109521. [DOI] [PubMed] [Google Scholar]
- 249.Butera G., Mullappilly N., Masetto F., Palmieri M., Scupoli M.T., Pacchiana R., Donadelli M. Regulation of autophagy by nuclear GAPDH and its aggregates in cancer and neurodegenerative disorders. Int. J. Mol. Sci. 2019;20(9):2062. doi: 10.3390/ijms20092062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Krasnov G.S., Dmitriev A.A., Snezhkina A.V., Kudryavtseva A.V. Deregulation of glycolysis in cancer: glyceraldehyde-3-phosphate dehydrogenase as a therapeutic target. Expert Opin. Ther. Targets. 2013;17(6):681–693. doi: 10.1517/14728222.2013.775253. [DOI] [PubMed] [Google Scholar]
- 251.Waker C.A., Lober R.M. Brain tumors of glial origin. Adv. Exp. Med. Biol. 2019;1190:281–297. doi: 10.1007/978-981-32-9636-7_18. [DOI] [PubMed] [Google Scholar]
- 252.Zeng T., Cui D., Gao L. Glioma: an overview of current classifications, characteristics, molecular biology and target therapies. Front. Biosci. 2015;20:1104–1115. doi: 10.2741/4362. [DOI] [PubMed] [Google Scholar]
- 253.Cohen A.L., Colman H. Glioma biology and molecular markers. Cancer Treat. Res. 2015;163:15–30. doi: 10.1007/978-3-319-12048-5_2. [DOI] [PubMed] [Google Scholar]