Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Oct 25.
Published in final edited form as: Free Radic Biol Med. 2021 Apr 18;169:74–83. doi: 10.1016/j.freeradbiomed.2021.03.043

Oxidative Stress Sensing and Response in Neural Stem Cell Fate

Inah Hwang 1,3,#, Deanna Tang 2,#, Jihye Paik 3,*
PMCID: PMC9594080  NIHMSID: NIHMS1695217  PMID: 33862161

Abstract

Neural stem/progenitor cells (NSPCs) contribute to the physiological cellular turnover of the adult brain and make up its regenerative potential. It is thus essential to understand how different factors influence their proliferation and differentiation to gain better insight into potential therapeutic targets in neurodegenerative diseases and traumatic brain injuries. Recent evidences indicate the roles of redox stress sensing and coping mechanisms in mediating the balance between NSPC self-renewal and differentiation. Such mechanisms involve direct cysteine modification, signaling and metabolic reprogramming, epigenetic alterations and transcription changes leading to adaptive responses like autophagy. Here, we discuss emerging findings on the involvement of redox sensors and effectors and their mechanisms in influencing changes in cellular redox potential and NSPC fate.

Graphical Abstract

graphic file with name nihms-1695217-f0001.jpg

1. Introduction

The persistence of neural stem/progenitor cells (NSPCs) ensures the regenerative potential of the adult brain [1]; [2]. NSPC fate changes depending on pathophysiological variables [3]. Disease conditions can involve defective antioxidant systems and radiation, [4], metabolism [5], and inflammation [4]; [6]. Aging, in particular, influences NSPC fate [7]. Biologically, the cellular milieu of the aging brain exhibits increased signs of impaired bioenergetics and adaptive neuroplasticity, aberrant neuronal activity and calcium homeostasis, the accumulation of oxidatively damaged molecules and organelles, and inflammation [8]. The brain is particularly vulnerable to oxidative stresses due to its high oxygen consumption, polyunsaturated fatty acids enriched in the neuronal membrane, and the presence of excitotoxic amino acids [9].

Reactive oxygen species (ROS) are unstable and reactive derivatives of oxygen such as hydroxyl radicals (•OH), superoxide anion radicals (O2•-), hydrogen peroxide (H2O2) and peroxynitrite (ONOO-) in a cell. These are by-products of cellular activities like mitochonrial respiration and fatty acid oxidation which contribute to the oxidative stress [10]. The major sources of ROS are cellular organelles including mitochondria, peroxisomes, endoplasmic reticulum, and lysosomes as well as enzymatic reactions like those of NADPH oxidases (NOXs) and cyclooxygenase [10]. Cellular signal transduction, such as activation of receptor tyrosine kinases, also increases cellular ROS levels [11, 12]. Therefore, NSPCs have different levels of ROS based on cellular contexts like proliferation and differentiation.

The cellular redox state also regulates the balance between the self-renewal and differentiation of NSPCs (Figure 1). Redox signaling has been found to play a significant role in regulating differentiation of NSPCs and clonal expansion in their proliferative niches [1316]. Under normal physiological conditions, the production of ROS is tightly regulated by the anti-oxidative scavenging system to maintain stemness properties of the NSPCs. Excessive levels of ROS accumulate and lead to oxidative stress conditions when ROS generation outpaces the scavenging system. High level of ROS can induce NSPC death and quiescence [13, 17]. On the other hand, elevated ROS are associated with increased production of their immediate progenitors [18]. Growth signaling-activated proliferative and multipotent NSPCs were shown to maintain a high ROS status for self-renewal and neurogenesis [14]. For example, NOX promotes maintaining self-renewal of NSPCs [14, 19]. Pharmacological or genetic studies showed NOX2 or NOX4 are necessary for NSPCs proliferation by inducing H2O2 production and PI3K/AKT pathway activation, suggesting that the basal ROS production is required for proliferation of NSPCs [14, 20, 21]. More recently, quiescent adult hippocampul NSPCs have been characterized to maintain high intracellular ROS levels which causally decrease as cells become activated [13]. Physiological stimuli induced a NOX2-mediated transient surge of ROS for the transition to activation, a finding emphasizing the dynamic coordination of ROS levels and NSPC fate. The reversible balance between NSPC quiescence and activation is important for adult neurogenesis and ROS dynamics surely matter in this fate decision.

Figure 1. Redox potential determines NSPCs fate.

Figure 1.

Schema for NSPC responses upon redox potential changes. ER: endoplasmic reticulum, NOX: NADPH oxidase, COX: cyclooxygenase, LOX: lipoxygenase, FA oxidation: fatty acid oxidation, PPP: pentose phosphate pathway, GPx: glutathione peroxidase, Trx: thioredoxin, Prx: peroxiredoxin

A number of genetic and pharmacological strategies were employed to reach the consensus that redox homeostasis and stress steer NSPC fate. Here, we will review current understanding of redox sensors and effectors and their mechanisms of action on NSPC fate change. We will further discuss redox signaling intermediates and redox responsive transcription factors as direct sensors and effctors of redox challenge, and ROS-induced epigenetic alterations, activation of autophagy, and metabolic reprogramming as adaptive responses to cope with redox-stress in NSPCs (Figure 1).

2. Redox Sensors and Effectors in NSPCs

Redox sensing is achieved by the biochemical and molecular alterations on specific sensor molecules. Intracellular ROS exist either as the reactive superoxide anion or its reduced hydrogen peroxide and hydroxyl radicals which can participate in reduction–oxidation-dependent signaling mechanisms [22]. The list of redox sensors is in Table 1. NSPCs respond to redox changes in the form of proximal signaling, intermediate epigenetic alterations and adaptive gene expression leading to reconfiguration of organelles and longer-term changes. We will discuss these changes characterized in NSPCs below.

Table 1.

The list of redox-sensing proteins

Molecule Family/Class Modification Effect of the modification Reference
AMPK Kinase Phosphorylation Activation [23]
ATM Kinase Phosphorylation Activation [24]
ASK1 Kinase Phosphorylation Activation [25]
ERK Kinase Phosphorylation Activation [26]
IKK Kinase Phosphorylation Activation [27]
Cysteine oxidation Inactivation [28]
JNK Kinase Phosphorylation Activation [29]
P38 MAPK Kinase Phosphorylation Activation [30]
mTORC1 Kinase Cysteine oxidation Activation [31]
PI3K/AKT Kinase Phosphorylation Activation [32],[33]
PKGIα Kinase Cysteine oxidation Activation [34]
Src Kinase Cysteine oxidation Inactivation [35]
FOXO Transcription factor Acetylation, Phosphorylation, Ubiquitination Activation [36]
HIF1 Transcription factor Deubiquitination Enhanced DNA binding [37]
P53 Transcription factor Cysteine oxidation, Phosphorylation Enhanced DNA binding [38]
NF-κB (p50) Transcription factor Cysteine oxidation Reduced DNA binding [39]
NF-κB (p65) Transcription factor Phosphorylation Enhanced DNA binding [40]
NF-κB/RelA Transcription factor Phosphorylation Reduced DNA binding [41]
Nrf2/Keap1 Transcription factor Deubiquitination Nrf2-ARE binding; activation [42],[43]
PTPs Phosphatase Cysteine oxidation Inactivation [44]
PTP1B Phosphatase Cysteine oxidation Inactivation [45]
PTEN Phosphatase Cysteine oxidation Inactivation [46]

2.1. Redox signaling

Cysteine oxidation/sulfenylation (-SOH) mediated activity change of signaling effectors is a well conserved redox responsive mechanism. It is direct and reversible sensing of redox potential within cellular milieu. It also provides rapid response to adjust the effect of growth signaling in order to cope with unfavorable environment. Notably, a number of phosphorylation consensus motifs are adjacent to cysteine residues that are subject to oxidation. For example, oxidized cysteines interfere with AKT-dependent phosphorylation of its substrate FOXO3 and override inhibitory PI3K/AKT signaling under oxidative stress [47]. As a result, FOXO3 is activated even in the presence of activated PI3K/AKT signaling in NSPCs. Similarly, reversible cysteine oxidation within the activating segment of Aurora kinase A interferes with adjacent threonine phosphorylation [48]. This mechanism is predicted to be dominant and evolutionarily conserved. Growing list of proteins that are subject to cysteine oxidation-mediated signaling is shown in Table 2. Below, we discuss a few examples of signaling effectors responsive to redox stress in NSPCs.

Table 2.

The list of molecules regulated by reversible cysteine oxidation

Molecule Residue Cell/Tissue type Family/Class Function Reference
Bcl-2 C158 C229 human lung epithelial cells (H460) Adaptor protein Interfering interaction with ERK; apoptosis [49]
DJ1 C106 H1299 HUVEC Adaptor protein Induced interaction with p53 [50, 51]
hSESN2 C125 HEK293 Adaptor protein mTORC1 activation [52]
STING C148 HEK293, Murine BMDM Adaptor protein Inhibition STING dimerization [53]
Calbindin D28K C187 C219 C257 Calcium binding protein Activation of myo-inositol monophosphatase, Reduction of Ca2+-binding [54]
Recoverin C39 Calcium binding protein Reduction of Ca2+-binding [55]
DLDH Unknown Rat brain Dehydrogenase Inactivation [56]
GAPDH C149 Rat ventricular myocyte Dehydrogenase Inhibition of glycolysis [57]
A20 C103 Deubiquitinase Reduced enzyme activity [58]
USP1 Unknown U2OS,T98 Deubiquitinase Reduced enzyme activity [59]
MGL C201 C208 Rat primary cortical neurons Hydrolase Reduced enzyme activity [60]
SGFH C60 Saccharomyces cerevisiae Hydrolase Reduced enzyme activity [61]
AKT2 C124 NIH-3T3 Kinase Enhanced kinase activity [62]
ASK1 C250 HEK293 Kinase T838 phosphorylation; JNK activation [63]
ATM C2991 Human primary fibroblasts (GM08399 B) Kinase Form S-S dimer, Activate downstream signaling [24]
Aurora A C290 HeLa Kinase Reduced kinase activity [48]
IKK0α Kinase Reduced kinase activity [28]
JNK2 C222 Human articular chondrocytes Kinase Enhanced kinase activity [64]
MEKK1 C1238 Kinase Inhibition of ATP-binding site [65]
Rheb unknown HEK293 Kinase Activation [31]
p56lck unknown Peripheral blood T lymphocytes (PBL blasts) Kinase Enhanced kinase activity [66]
Pin1 C113 Human brain SH-SY5Y Kinase Reduced kinase activity [67], [68]
PKA C358 HeLa Kinase Reduced kinase activity [48]
PKIG1a C42 Mouse cardiomyocytes Kinase Reduced kinase activity [34]
Src C277 C185 NCI-H292 Kinase Y416, Y527 phosphorylation; activation [69], [35]
Aldose Reductase C298 C303 Rat heart Oxidoreductase Reduced enzyme activity [70]
MsrA C72 Oxidoreductase Reduced enzyme activity [71]
Prx-1 C52 C83 C173 Oxidoreductase Eliminates hydrogen peroxide [72]
PTEN C124 NIH-3T3, HeLa Phosphatase Suppressed enzyme activity [73], [46]
PTPσ C1589 Phosphatase Reduced enzyme activity [74]
PTP1B C215 A431 (human epidermoid carcinoma) Phosphatase Enzyme inactivation [45], [75],[76]
SHP-1/2 C455 C459 A20 (Murine B-Cell lymphoma) Phosphatase Enzyme inactivation [77], [78]
Cathepsin K C25 Protease Reduced maturation of precathepsin K [79]
EGFR C797 A431 Receptor kinase Enhanced kinase activity [80]
FGFR1 C488 Receptor kinase Enhanced kinase activity [69]
AP-1 Transcription Factor Decreased DNA binding [81]
FOXO3 C31 NSPC Transcription Factor Inhibit T32 phosphorylation; activation [47]
FOXO4 C477 HEK293 Transcription Factor Acetylated by p300; inactivation [82]
NF-κB (p50) C62 Transcription factor Decreased DNA binding [83]
P53 C124 C141 C182 Transcription factor Enhanced DNA binding [38]
Yap1 C303 C598 Transcription factor Activation [84]
hBCATm C315 Transferase Reduced enzyme activity [85]
MST C247 Transferase Reduced enzyme activity [86]
MTAP C136 C223 HepG2 Transferase Reduced enzyme activity [87]
PRMT1 C101 C208 Transferase Reduced enzyme activity [88]

ATM.

Ataxia telangiectasia (AT) is an autosomal recessive genetic disorder characterized by neurodegeneration with progressive ataxia, variable immunodeficiency, premature aging and an increased disposition to cancer. Clinically, A-T patients showed defective antioxidative capacity [89]. ROS oxidize the ataxia telangiectasia mutated (ATM) in its active disulfide-crosslinked dimer form, which activates the DNA repair pathway. ATM-lacking cells failed to activate the DNA repair pathway and were hypersensitive to DNA damage under oxidative stress. Studies with ATM oxidation indicate ATM to be a redox sensor in the cells [24]; [90]; [91]. ATM is abundant in NSPCs and is downregulated during differentiation. ATM-lacking NSPCs cannot differentiate into neurons or astrocytes and have shown abnormal proliferation and genomic instability [92].

PI3K/AKT/mTOR signaling.

PI3K/AKT signaling regulates ROS-dependent induction of neurogenesis. Also, promotion of the mammalian target of rapamycin (mTOR) pathway by ROS alters NSPC differentiation and migration, dendrite development, and neuronal maturation. Disruption of the mTOR pathway results in a host of neurodevelopmental disorders [93].

Evidence indicates the importance of ROS and PI3K/AKT signaling in NSPC differentiation and neuronal maturation. It also suggests that an enhanced oxidative cellular status and increased PI3K/AKT signaling in NSPCs lead to premature neurogenesis and prevention of excessive proliferation [94]. However, it is important to note that a different study found that ROS-mediated enhancements in the self-renewal and neurogenesis of NSPCs are dependent on PI3K/AKT signaling. ROS activates the PI3K/AKT/mTOR pathway via oxidative inactivation of the PTEN protein, a negative regulator of PI3K signaling. PTEN counteracts the kinase function of PI3K, subsequently inhibiting AKT activation. This PI3K-dependent activation of AKT is crucial for the growth and proliferation of adult hippocampal stem/progenitor cells [95]. Also, H2O2 deactivates PTEN, which enhances AKT signaling and maintains physiological signaling and proliferation of adult hippocampal stem/progenitor cells [96]. Thus, high ROS status of NSPCs seem to be required in order to maintain adequate levels of PI3K signaling and in turn maintain self-renewal and neurogenesis [14].

Wnt.

Elevated ROS levels enhance activation of Wnt/β-catenin signaling, which regulates the efficiency of neural differentiation. Evidence indicates that ROS production precedes Wnt/β-catenin activation, and that mitochondrial ROS metabolism is involved in regulating early events of Wnt/β-catenin signaling transduction. Delaying the onset of Wnt/β-catenin activation will lead to impaired neuronal differentiation [97]. The activation of Wnt/β-catenin signaling can mediate neuroprotection and improve neurological function during oxidative stress via Nrf2/PI3K/AKT-Wnt/Fzd-1/β-catenin cooperation and as part of a feedback loop regulating cellular homeostasis [98].

Type-I Interferon response.

The type-I interferon response is an innate immune response. An increase in type-I interferon signaling is found to be correlated with increased oxidative stress, as well as aging in the brain [99]; [47]. ROS signaling activates FOXO, which triggers the activation of the type-I interferon pathway. This increased type-I interferon response under oxidative stress suppresses the proliferation of NSPCs and reduces neuronal differentiation [47]. Type-I interferon response also activates cellular autophagy [100] and may serve as a protective response against oxidative stress.

2.2. Redox responsive transcription factors

A primary defense mechanism to oxidative stress is the antioxidant response, which combats noxious effects of oxidative insults [98]. Antioxidants rescue cells from oxidative stress and preserve redox balance by promoting defenses to neutralize ROS and regulate transcription [101]. Anti-oxidative stress responsive transcription factors and defensive gene expression program, therefore, are critical for maintaining NSPC reserves and their function. PR domain containing 16 (PRDM16), a histone-lysine N-methyltransferase, is required for NSPC maintenance and regulation of neural progenitor behavior [102]. PRDM16 depletion results in an elevation of oxidative stress levels and drives the expression of oxidative stress-responsive genes in cortical radial glial progenitors (RPGs). Removal of PRDM16 is also found to alter neocortical organization and expand RPG proliferation in the embryonic stage (E13.5) [103]. In addition to PRDM16-mediated fitness keeping constitutive transcription, below are several transcription factors acutely responsive to redox stress.

NRF2.

Nuclear factor erythroid 2-related factor 2 (Nrf2) is the master regulator of the cellular defense against oxidative stress [98]. Nrf2 activity is controlled by Kelch-like erythroid cell-derived protein with CNC homology (ECH)-associated protein 1 (Keap1) which serves as a cysteine-based intracellular sensor for oxidative stress [104]; [105]. Nrf2 regulates antioxidant gene expression through binding to the antioxidant-responsive element (ARE) (TGA[C/T]NNGC) and modulates mitochondrial biogenesis and function. By improving mitochondrial function, Nrf2 is able to diminish intracellular ROS overproduction [106].

Nrf2 is crucial for NSPC function. Nrf2 knockout mice showed impaired NSPC stemness and neuronal differentiation [107]. Aging downregulated the Nrf2-Hmox adaptive response and thus impaired neuronal differentiation of NSPCs in Parkinson’s disease (PD) mouse model [108]. During aging, a diminishing Nrf2 expression compromises the regenerative potential of NSPCs and results in a specific temporal pattern of decline in neurogenesis [109]. Overexpression of Nrf2 in ‘old’ NSPCs rendered them similar to ‘young’ NSPCs, showing increased survival and regneration. This implies that Nrf2 is a significant influencer in NSPC survival and function during aging [110].

HIF-1α.

Hypoxia leads to an upregulation and stabilization in hypoxia inducible factor-1α (HIF-1α) levels [111]. This is due to a paradoxical increase of oxidative stress under hypoxia. Electrons leak from the mitochondrial electron transport chain generating ROS under low oxygen tension and increasing HIF-1α [112]. NSPCs are exposed to a hypoxic environment during embryonic development. Thus, hypoxia signaling plays an important role in regulating NSPC maintenance and differentiation. Under hypoxic conditions, HIF-1α promotes signaling pathways that promote NSPC self-renewal and inhibits pathways that promote differentiation [113]. HIF-1α is also crucial to maintaining NSPCs as well as the vascularity of the niche [114]. Similarly, genetic inactivation of HIF-1α reduced adult hippocampal NSPCs and impaired neurogenesis [115].

TP53.

Loss of function of the tumor suppressor gene p53 results in elevated ROS, leading to early neurogenesis [116]. Restoration in function of p53 and anti-oxidative treatments partially reverse this phenotype, suggesting that p53-mediated regulation of physiological ROS levels may be required for the appropriate timing of neurogenesis. p53 has also been identified to play a role in neuronal fate via mediation of ROS levels [117]. The mitochondrial translocation of p53 is associated with a decrease in mitochondrial ROS levels and a shift in differentiation toward neurons, rather than astrocytes.

NFκB.

Nuclear factor-κB (NFκB) signaling is a key regulator for multiple niche factors that are involved in regulating different stages or phases of neurogenesis. The activation of NFκB is important in regulating self-renewal and early differentiation, as well as apoptosis of NSPCs [118]. NFκB is also a redox-sensitive transcription factor activated by ROS [119]. NFκB plays a protective role under oxidative stress by suppressing ROS accumulation and activating antioxidant targets [120]. Conversely, depending on phase and context, ROS can have bimodal effects and can either activate or suppress NFκB signaling. Oxidative stress activates the NFκB pathway in the early phase and suppresses NFκB activation in the late phase [120].

FOXO.

Forkhead box O (FOXO) transcription factors have protective effects against aging-progressive axonal degeneration through the suppression of mTORC1-induced proteotoxic stress to restore autophagy capacity and reduce ROS [121]. As a critical defense mechanism against oxidative stress FOXOs emerged as a major determinant of neural cell fate [122].

FOXOs are important for the maintenance of NSPC homeostasis and have protective effects against ROS [15]. FOXO1 regulates neurogenesis and interacts with the Notch pathway to control NSPC fate [123]. FOXO3 regulates glucose or carbon metabolism in a manner that supports redox balance and the neurogenic potential of NSPCs [124]. Under oxidative stress, FOXO3 promotes upregulation of glycine-N-methyltransferase (GNMT), a methyltransferase for glycine, which leads to s-adenosylmethionine (SAM) depletion, activation of type I interferon response, and suppression of differentiation, sequentially [47]. These FOXO-mediated oxidative stress responses protect NSPCs from premature activation and maintain their quiescence.

Notably, direct protein modification by cysteine sulfenylation was proposed to be an oxidative stress sensing mechanism of FOXO3 [47]. Cysteine sulfenylation adjacent to inhibitory AKT phosphorylation site of FOXO3 circumvents PI3K-AKT-mediated inactivation signal under oxidative stress (Figure 2). This mechanism is also highly conserved in a number of redox signaling cascades as discussed above.

Figure 2. Integration of redox sensors and effectors.

Figure 2.

Receptor tyrosine kinases (RTK) produces ROS along with regulating PI3K/PTEN/AKT. AKT phosphorylates and inhibits nuclear translocation and activation of FOXO. Metabolic stress-induced ROS activates LKB/AMPK, which, in turn, activates autophagy and inhibits mTORC1 activity. Oxidative stress-activated SIRT and AMPK increase mitochondria biogenesis through PGC1α. ROS stabilize Nrf2 or HIF1α by dissociation of Keap1 and VHL, respectively. Cysteine oxidation among other modifications plays an important role in the response.

2.3. Redox stress-induced epigenetic alterations

Chromatin remodeling is an energy-demanding cellular process, and therefore requires balance with cellular metabolism and availability of metabolites. Several metabolic pathways produce intermediates that are involved in modifying the epigenetic landscape, and subsequently regulating NSPC fate. Oxidative stress is demonstrated to cause remodeling of chromatin and epigenetic reprogramming [125].

One-carbon metabolism drives epigenetic changes via two pathways: the folate pathway, which is involved in DNA synthesis, and the methionine pathway, which is involved in methylation reactions [126]. This metabolic pathway is involved in regulating NSPC self-renewal and differentiation. A recent study showed chromatin serves as methyl group sinks when phospholipid methylation is impaired [127]. Excess s-adenosylmethionine (SAM) is consumed by histone methylation reactions promoting trimethylation of histone H3 on Lys4 (H3K4me3), H3K36me3 and H3K79me3. These histone modifications could contribute to the altered gene expression, but they were not directly correlated with stable changes in transcription. Instead, histone methylation reaction facilitates SAM to s-adenosylhomocysteine (SAH) conversion, which then feeds into the transsulfuration reaction to boost anti-oxidant glutathione synthesis. Whether altered histone modification contributes to gene expression changes leading to NSPC fate change has not been reported.

The Eph:ephrin cell-cell communicating signaling regulates one-carbon metabolism to control NSPC differentiation. Ephrin B1 stimulation of NSPCs alters the methylation state of H3K4 by suppressing dihydrofolate reductase expression, a key enzyme for cellular proliferation. Eph forward signaling decreases H3K4 methylation on progenitor genes (Sox2, Nes) to promote differentiation in NSPCs [128]. Interestingly, this epigenetic reprogramming is long-lasting and inherited that NSPCs are locked in a differentiation-prone state. Given that dihydrofolate reductase engages NADPH as a cofactor, its action is likely to be sensitive to intracellular oxidative stress. H3K4me3 peak breadth has been linked to the cellular identity and transcriptional consistency in NSPCs [129]. How elevated redox stress may affect the H3K4me3 landscape and fate of NSPCs and their fate remains to be defined.

SAM, the principal methyl donor in cellular methylation reactions, is exhausted under oxidative stress [125]. SAM metabolism is required for the preservation of proliferative signaling in NSPCs. Oxidative stress is shown to suppress the neurogenic potential of NSPCs by activating type-I interferon response through reduction of intracellular SAM availability [47]. Depletion of SAM during oxidative stress response resulted in decreased nuclear lamin methylation which compromised nuclear envelope integrity. Nuclear lamina integrity is required for the proper 3D organization of nuclear chromatin [130]. Although not directly, redox stress is likely to have a profound role in both epigenetic and genomic reorganization in NSPCs.

The glutamine metabolism is essential for NSPC growth and long-term maintenance. It is a precursor for glutamate, a key building block of glutathione and therefore protects NSPCs from oxidative stress [124]. Glutamine also feeds into α-ketoglutarate, which is a key cofactor for histone and DNA demethylases. It is required for the removal of repressive histone modifications and DNA methylation in NSPCs to promote multipotency [131]. The metabolic reprogramming from aerobic glycolysis to the TCA cycle is associated with neuronal differentiation of NSPCs. Several TCA intermediates, including α-ketoglutarate, NADH, and FADH2, are involved in the epigenetic regulation of transcription [132].

Lipid metabolism is involved with promoting histone acetylation through acetyl-CoA and expression of growth-related genes to drive cellular growth [133]. This metabolic pathway plays a significant role in the interaction between adult NSPCs and their niche, and promotes NSPC proliferation and neurogenesis [132]; [134]. Altered histone acetylation and associated gene expression changes under oxidative stress have been reported [125]. Previously, the role of acetyl-CoA generated by acetyl-CoA synthetase 2 (ACSS2) in hippocampal neuronal differentiation has been demonstrated [135]. ACSS2 is localized to the nucleus and functions as a chromatin-bound transcriptional coactivator that stimulates histone acetylation and gene expression relevant to the plasticity of neurons. Similarly, cellular acetyl-CoA metabolism and neuronal differentiation connection has been reported. TP53-inducible glycolysis and apoptosis regulator (TIGAR) is an endogenous inhibitor of glycolysis and is upregulated during neuronal differentiation [136]. Depletion of TIGAR reduced acetyl-CoA and H3K9 acetylation at the promoters of neurogenic genes resulting in decreased neuronal differentiation. Acetate, a precursor of acetyl-CoA, restored H3K9 acetylation and NSPC differentiation of TIGAR-depleted cells. Given that TIGAR plays a role in oxidative stress-induced metabolic reprogramming [137], it is likely to regulate NSPC differentiation through a histone acetylation-based epigenetic mechanism.

Oxidative stress has been found to alter metabolites and disrupt the epigenetic state of the cell [138]. Changes in the redox state was shown to regulate the fate of NSPCs via histone deacetylase SIRT1. Under oxidative stress conditions, SIRT1 is upregulated through increased NAD+ to NADH ratio [139]. Oxidative stress-activated SIRT1 suppresses adult hippocampal NSPC self-renewal [140] and inhibits the pro-neuronal MASH1 transcription factor expression, diverting the fate of NSPCs from neurogenesis towards the astroglial lineage [141].

2.4. Redox stress-activated autophagy

Recent studies have begun to look at the role of autophagy in the regulation of neurogenesis in the adult brain. The available data indicate the autophagy pathway regulates processes including maintenance and activation of quiescent NSPCs [142]. FOXO transcription factors play a critical role in maintaining physiological autophagic flux to regulate neurogenesis [143]. Autophagy is vital for cellular homeostasis in removing protein aggregates and dysfunctional organelles for physiological neuronal development.

Evidence demonstrates a vital role for autophagy in neuronal differentiation. It acts as a response-survival mechanism to reduce oxidative stress and regulate synaptogenesis that is associated with this process [144]. The downregulation of pro-autophagic proteins Ambra1 and Beclin 1 in adult NSPCs results in a decrease in cell proliferation and enhanced sensitivity to DNA damage-induced cell death [145]. The findings from this study suggest that Ambra1- and Beclin 1-mediated autophagy is involved in adult neurogenesis by controlling the survival of NSPCs.

Increasing evidence suggest the crosstalk between ROS and autophagy may play a determinant role in the regulation of neuronal homeostasis. Under conditions of oxidative stress, mitochondrial ROS induce autophagy [146]. This activation of autophagy is key in the cellular response to oxidative stress in order to dispose of defective components and prevent further damage to cells. Excessive ROS production not only impairs cellular macromolecules, but can also cause damage to antioxidant enzymes and nonenzymatic oxidants [147]. The autophagic process is thus critical in maintaining homeostasis by serving as a buffer system to control ROS levels in cells and inhibit inappropriate activation of ROS in response to neuronal damage [148]

2.5. Metabolic flexibility against oxidative stress

The coordination of metabolic adaptation with cellular signaling and epigenetic regulation supports divergent NSPC cell fate [149]. Distinct metabolic processes and their involvement in the regulation of NSPC fate have been studied to provide insight into the regenerative potential of the brain [5]. For example, subventricular zone NSPCs mostly rely on fatty acid oxidation to produce energy and support their neurogenic activity [150]. Similarly, hippocampal NSPCs may operate on beta-oxidation and glycolysis as the major metabolic activity as inferred from single cell transcriptome analysis [151]. NSPCs also rely on the pentose phosphate pathway (PPP) and thus are sensitive to its perturbation [124]. Oxidative PPP forms reduced nicotinamide adenine dinucleotide phosphate (NADPH), which is important for the suppression of cellular oxidative stress [152]. Oxidative stress redirects glucose into the PPP by a feedback regulatory loop and generates NADPH, suppressing an elevation of ROS [153]; [154]. In addition, glutamine flux regulates mTOR activity, translation, and autophagy to coordinate cellular growth and proliferation [155]. Glutamine metabolism also contributes to the control of redox balance by increasing the NADPH/NADP(+) ratio to suppress oxidative stress in NSPCs [124]; [156].

Mitochondrial dynamics is central to metabolic adaptation under oxidative stress. As a central organelle generating intracellular ROS, mitochondrial function and morphology are closely associated with NSPC fate [116]. The plasticity of the mitochondria enables it to undergo morphological and functional changes in response to cellular demands. This mitochondrial plasticity plays a crucial role in integrating signaling cues to regulate stem cell fates [157]. Changes in mitochondrial dynamics modulate NSPC fate decisions by driving a physiological ROS-mediated process that suppresses self-renewal and promotes differentiation [158]. Dysfunctional mitochondria due to mitochondrial DNA mutations impairs oxidative phosphorylation and accumulation of ROS, resulting in oxidative stress and altered redox signaling pathway activity. Unlike mitochondria-derived physiological ROS signaling, high levels of ROS produced from dysfunctional mitochondria impair adult neurogenesis [159]. Conversely, redox signaling through cysteine modifications regulate mitochondrial function and metabolism [160].

Studies reveal changes in mitochondrial morphology as NSPCs undergo neuronal differentiations Mitochondrial morphology changes regulate mitochondrial function of metabolism and ROS generation. Mitochondrial dynamics acts as an upstream regulator of physiological ROS to control NSPC self-renewal and fate decisions [158]. There is a demonstrated increase in vulnerability of mitochondrial mass and DNA to oxidative damage during NSPC differentiation. Maintaining mitochondrial DNA integrity is vital for the proper maturation of the mitochondria during neural differentiation [161]. The accumulation of mitochondrial damage from oxidative stress would result in NSPCs failing to properly differentiate into neurons.

3. Conclusion

The above body of evidence supports that cellular redox states regulate the balance between NSPC quiescence maintenance and activation in adult mammalian brains. The interplay of key redox sensors and effectors is depicted in Figure 2. In general, increased levels of intracellular ROS were linked to proliferation and differentiation of NSPCs [162]. A more recent study, however, revealed that quiescent NSPCs of the adult hippocampus unexpectedly show the highest levels of ROS [13]. This finding adds more complexity to the link between redox regulation and NSPC fate decision. While many studies addressed the causality of redox stress in NSPC fate changes the majority of observations are based on perturbative and correlative conditions. Further studies using real tine tracing of NSPC cell fate with the biosensor-based quantitation should aid better appreciation of the role of dynamic redox balance. The interplay between NSPC microenvironment and cellular autonomy also needs to be better appreciated. With a recent advent of experimental platforms including cerebral organoids and assembloids, induced vascularization and single cell genomics, it is possible to directly address how redox potential may control NSPC fate without perturbing heterotypic interaction with other cell types. In a recent translational study, a personalized cell-therapy strategy of implanting autologous induced pluripotent stem cells into a patient with PD demonstrated clinical improvement coinciding with reinnervation of the putamen by projections from dopaminergic neurons [163]. This successful proof of principle study further speaks for the importance of steering NSPC cell fate in vivo. A better understanding of underlying redox-based molecular mechanisms may contribute to the development of methods to optimally stimulate neuronal differentiation of NSPCs in patients with neurodegeneration and brain damages.

Highlights.

  • NSPC is vulnerable to oxidative stresses due to the surrounding brain with high oxygen consumption.

  • Redox-stress sensing and coping mechanisms balance NSPC self-renewal and differentiation.

  • Direct cysteine modifications of signaling intermediates and transcription factors result in metabolic reprogramming, epigenetic alterations towards adaptive responses under increased redox potential.

Acknowledgement

We apologize to authors whose work has not been cited due to space limitations and to limitations of our knowledge. This work was supported by National Institutes of Health Grant AG048284 (to J.P.).

Abbreviations

aaRS

Alanyl-tRNA synthetase

ACSS2

Acyl coenzyme A synthetase-2

AKT

Protein kinase B

AMPK

5’adenosine monophosphate-activated protein kinase

ASK1

Apoptosis signal-regulating kinase 1

ATM

Ataxia telangiectasia mutated

Bcl-2

B-cell lymphoma 2

DLDH

Dihydrolipoamide Dehydrogenase

EGFR

Epidermal growth factor receptor

ERK

Extracellular signal-regulated kinase

FADH2

Flavin adenine dinucleotide

FGFR1

Fibroblast Growth Factor Receptor 1

FOXJ1

Forkhead box protein J1

FOXO

Forkhead box protein O

GAPDH

Glyceraldehyde 3-phosphate dehydrogenase

GNMT

Glycine N-methyltransferase

hBCATm

Human branched chain aminotransferase

HIF1

Hypoxia inducible factor I

hSESN2

Human sestrin-2

IKK

Inhibitor of nuclear factor-κB kinase

IQGAP

Ras Guanosine-5’-triphosphatase-activating-like protein

JNK

c-Jun N-terminal kinases

Keap1

Kelch-like erythroid cell-derived protein with CNC homology-associated protein 1

MASH1

mammalian achaete scute homolog-1

MEKK1

Mitogen-activated protein kinase kinase kinase-1

MGL

Monoacylglycerol lipase

MsrA

Methionine sulfoxide reductase A

MST

Macrophage-stimulating protein

MTAP

S-methyl-5’-thioadenosine phosphorylase

mTORC1

Mammalian target of rapamycin complex 1

NADPH

Nicotinamide adenine dinucleotide phosphate

NF-kB

Nuclear factor-kB

NOX

NADPH oxidase

Nrf2

Nuclear factor erythroid 2–related factor 2

NSPCs

Neural stem/progenitor cells

P38 MAPK

p38 mitogen-activated protein kinase

P56lck

p56 lymphocyte-specific protein kinase

PI3K

Phosphoinositide 3-Kinase

PKA

Protein kinase A

PKG1a

Protein kinase G type- 1alpha

PPP

Pentose phosphate pathway

PRDM16

PRDF1-RIZ (PR) domain containing 16

PRMT1

Protein arginine N-methyltransferase-1

Prx-1

Peroxiredoxin-1

PTEN

Phosphatase and tensin homolog

PTP

Protein tyrosine phosphatase

ROS

Reactive oxygen species

SAM

S-adenosylmethionine

SHP-1/2

Src homology 2 domain tyrosine phosphaoases-1/2

SIRT1

Sirtuin-1

STING

Stimulator of interferon genes

TCA

Tricarboxylic acid cycle

TIGAR

TP53 Induced Glycolysis Regulatory Phosphatase

TRPA

Tryptophan synthase, alpha subunit

USP1

Ubiquitin Specific Peptidase 1

VEGFR

Vascular endothelial growth factor

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • [1].Gage FH, Temple S, Neural stem cells: generating and regenerating the brain, Neuron 80(3) (2013) 588–601. [DOI] [PubMed] [Google Scholar]
  • [2].Ma DK, Bonaguidi MA, Ming GL, Song H, Adult neural stem cells in the mammalian central nervous system, Cell Res 19(6) (2009) 672–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Huang TT, Zou Y, Corniola R, Oxidative stress and adult neurogenesis--effects of radiation and superoxide dismutase deficiency, Semin Cell Dev Biol 23(7) (2012) 738–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Borsini A, Zunszain PA, Thuret S, Pariante CM, The role of inflammatory cytokines as key modulators of neurogenesis, Trends Neurosci 38(3) (2015) 145–57. [DOI] [PubMed] [Google Scholar]
  • [5].Kim DY, Rhee I, Paik J, Metabolic circuits in neural stem cells, Cell Mol Life Sci 71(21) (2014) 4221–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Chesnokova V, Pechnick RN, Wawrowsky K, Chronic peripheral inflammation, hippocampal neurogenesis, and behavior, Brain Behav Immun 58 (2016) 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Lupo G, Gioia R, Nisi PS, Biagioni S, Cacci E, Molecular Mechanisms of Neurogenic Aging in the Adult Mouse Subventricular Zone, J Exp Neurosci 13 (2019) 1179069519829040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Mattson MP, Arumugam TV, Hallmarks of Brain Aging: Adaptive and Pathological Modification by Metabolic States, Cell Metab 27(6) (2018) 1176–1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Uttara B, Singh AV, Zamboni P, Mahajan RT, Oxidative stress and neurodegenerative diseases: a review of upstream and downstream antioxidant therapeutic options, Curr Neuropharmacol 7(1) (2009) 65–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Ye ZW, Zhang J, Townsend DM, Tew KD, Oxidative stress, redox regulation and diseases of cellular differentiation, Biochim Biophys Acta 1850(8) (2015) 1607–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Woo HA, Yim SH, Shin DH, Kang D, Yu DY, Rhee SG, Inactivation of peroxiredoxin I by phosphorylation allows localized H(2)O(2) accumulation for cell signaling, Cell 140(4) (2010) 517–28. [DOI] [PubMed] [Google Scholar]
  • [12].Sundaresan M, Yu ZX, Ferrans VJ, Irani K, Finkel T, Requirement for generation of H2O2 for platelet-derived growth factor signal transduction, Science 270(5234) (1995) 296–9. [DOI] [PubMed] [Google Scholar]
  • [13].Adusumilli VS, Walker TL, Overall RW, Klatt GM, Zeidan SA, Zocher S, Kirova DG, Ntitsias K, Fischer TJ, Sykes AM, Reinhardt S, Dahl A, Mansfeld J, Rünker AE, Kempermann G, ROS Dynamics Delineate Functional States of Hippocampal Neural Stem Cells and Link to Their Activity-Dependent Exit from Quiescence, Cell Stem Cell (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Le Belle JE, Orozco NM, Paucar AA, Saxe JP, Mottahedeh J, Pyle AD, Wu H, Kornblum HI, Proliferative neural stem cells have high endogenous ROS levels that regulate self-renewal and neurogenesis in a PI3K/Akt-dependant manner, Cell Stem Cell 8(1) (2011) 59–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Paik JH, Ding Z, Narurkar R, Ramkissoon S, Muller F, Kamoun WS, Chae SS, Zheng H, Ying H, Mahoney J, Hiller D, Jiang S, Protopopov A, Wong WH, Chin L, Ligon KL, DePinho RA, FoxOs cooperatively regulate diverse pathways governing neural stem cell homeostasis, Cell Stem Cell 5(5) (2009) 540–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Renault VM, Rafalski VA, Morgan AA, Salih DA, Brett JO, Webb AE, Villeda SA, Thekkat PU, Guillerey C, Denko NC, Palmer TD, Butte AJ, Brunet A, FoxO3 regulates neural stem cell homeostasis, Cell Stem Cell 5(5) (2009) 527–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Li J, Johnson D, Calkins M, Wright L, Svendsen C, Johnson J, Stabilization of Nrf2 by tBHQ confers protection against oxidative stress-induced cell death in human neural stem cells, Toxicol Sci 83(2) (2005) 313–28. [DOI] [PubMed] [Google Scholar]
  • [18].Bigarella CL, Liang R, Ghaffari S, Stem cells and the impact of ROS signaling, Development 141(22) (2014) 4206–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Adane B, Ye H, Khan N, Pei S, Minhajuddin M, Stevens BM, Jones CL, D’Alessandro A, Reisz JA, Zaberezhnyy V, Gasparetto M, Ho TC, Kelly KK, Myers JR, Ashton JM, Siegenthaler J, Kume T, Campbell EL, Pollyea DA, Becker MW, Jordan CT, The Hematopoietic Oxidase NOX2 Regulates Self-Renewal of Leukemic Stem Cells, Cell Rep 27(1) (2019) 238–254 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Nayernia Z, Colaianna M, Robledinos-Anton N, Gutzwiller E, Sloan-Bena F, Stathaki E, Hibaoui Y, Cuadrado A, Hescheler J, Stasia MJ, Saric T, Jaquet V, Krause KH, Decreased neural precursor cell pool in NADPH oxidase 2-deficiency: From mouse brain to neural differentiation of patient derived iPSC, Redox Biol 13 (2017) 82–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Yoshikawa Y, Ago T, Kuroda J, Wakisaka Y, Tachibana M, Komori M, Shibahara T, Nakashima H, Nakashima K, Kitazono T, Nox4 Promotes Neural Stem/Precursor Cell Proliferation and Neurogenesis in the Hippocampus and Restores Memory Function Following Trimethyltin-Induced Injury, Neuroscience 398 (2019) 193–205. [DOI] [PubMed] [Google Scholar]
  • [22].Holmstrom KM, Finkel T, Cellular mechanisms and physiological consequences of redox-dependent signalling, Nat Rev Mol Cell Biol 15(6) (2014) 411–21. [DOI] [PubMed] [Google Scholar]
  • [23].Zmijewski JW, Banerjee S, Bae H, Friggeri A, Lazarowski ER, Abraham E, Exposure to hydrogen peroxide induces oxidation and activation of AMP-activated protein kinase, J Biol Chem 285(43) (2010) 33154–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Guo Z, Kozlov S, Lavin MF, Person MD, Paull TT, ATM activation by oxidative stress, Science 330(6003) (2010) 517–21. [DOI] [PubMed] [Google Scholar]
  • [25].Matsuzawa A, Saegusa K, Noguchi T, Sadamitsu C, Nishitoh H, Nagai S, Koyasu S, Matsumoto K, Takeda K, Ichijo H, ROS-dependent activation of the TRAF6-ASK1-p38 pathway is selectively required for TLR4-mediated innate immunity, Nat Immunol 6(6) (2005) 587–92. [DOI] [PubMed] [Google Scholar]
  • [26].Lei H, Kazlauskas A, Growth factors outside of the platelet-derived growth factor (PDGF) family employ reactive oxygen species/Src family kinases to activate PDGF receptor alpha and thereby promote proliferation and survival of cells, J Biol Chem 284(10) (2009) 6329–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Korn SH, Wouters EF, Vos N, Janssen-Heininger YM, Cytokine-induced activation of nuclear factor-kappa B is inhibited by hydrogen peroxide through oxidative inactivation of IkappaB kinase, J Biol Chem 276(38) (2001) 35693–700. [DOI] [PubMed] [Google Scholar]
  • [28].Wetzel MA, Scholle J, Teschke K, Artificial structures in sediment-dominated estuaries and their possible influences on the ecosystem, Mar Environ Res 99 (2014) 125–35. [DOI] [PubMed] [Google Scholar]
  • [29].Shen HM, Liu ZG, JNK signaling pathway is a key modulator in cell death mediated by reactive oxygen and nitrogen species, Free Radic Biol Med 40(6) (2006) 928–39. [DOI] [PubMed] [Google Scholar]
  • [30].Sato A, Okada M, Shibuya K, Watanabe E, Seino S, Narita Y, Shibui S, Kayama T, Kitanaka C, Pivotal role for ROS activation of p38 MAPK in the control of differentiation and tumor-initiating capacity of glioma-initiating cells, Stem Cell Res 12(1) (2014) 119–31. [DOI] [PubMed] [Google Scholar]
  • [31].Yoshida S, Hong S, Suzuki T, Nada S, Mannan AM, Wang J, Okada M, Guan KL, Inoki K, Redox regulates mammalian target of rapamycin complex 1 (mTORC1) activity by modulating the TSC1/TSC2-Rheb GTPase pathway, J Biol Chem 286(37) (2011) 32651–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Niwa K, Inanami O, Yamamori T, Ohta T, Hamasu T, Kuwabara M, Redox regulation of PI3K/Akt and p53 in bovine aortic endothelial cells exposed to hydrogen peroxide, Antioxid Redox Signal 5(6) (2003) 713–22. [DOI] [PubMed] [Google Scholar]
  • [33].Koundouros N, Poulogiannis G, Phosphoinositide 3-Kinase/Akt Signaling and Redox Metabolism in Cancer, Front Oncol 8 (2018) 160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Oeing CU, Nakamura T, Pan S, Mishra S, Dunkerly-Eyring BL, Kokkonen-Simon KM, Lin BL, Chen A, Zhu G, Bedja D, Lee DI, Kass DA, Ranek MJ, PKG1α Cysteine-42 Redox State Controls mTORC1 Activation in Pathological Cardiac Hypertrophy, Circ Res 127(4) (2020) 522–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Heppner DE, Dustin CM, Liao C, Hristova M, Veith C, Little AC, Ahlers BA, White SL, Deng B, Lam YW, Li J, van der Vliet A, Direct cysteine sulfenylation drives activation of the Src kinase, Nat Commun 9(1) (2018) 4522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Klotz LO, Sánchez-Ramos C, Prieto-Arroyo I, Urbánek P, Steinbrenner H, Monsalve M, Redox regulation of FoxO transcription factors, Redox Biol 6 (2015) 51–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Movafagh S, Crook S, Vo K, Regulation of hypoxia-inducible factor-1a by reactive oxygen species: new developments in an old debate, J Cell Biochem 116(5) (2015) 696–703. [DOI] [PubMed] [Google Scholar]
  • [38].Liu B, Chen Y, St Clair DK, ROS and p53: a versatile partnership, Free Radic Biol Med 44(8) (2008) 1529–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Gambhir L, Checker R, Sharma D, Thoh M, Patil A, Degani M, Gota V, Sandur SK, Thiol dependent NF-κB suppression and inhibition of T-cell mediated adaptive immune responses by a naturally occurring steroidal lactone Withaferin A, Toxicol Appl Pharmacol 289(2) (2015) 297–312. [DOI] [PubMed] [Google Scholar]
  • [40].Liu J, Yoshida Y, Yamashita U, DNA-binding activity of NF-kappaB and phosphorylation of p65 are induced by N-acetylcysteine through phosphatidylinositol (PI) 3-kinase, Mol Immunol 45(15) (2008) 3984–9. [DOI] [PubMed] [Google Scholar]
  • [41].Jamaluddin M, Wang S, Boldogh I, Tian B, Brasier AR, TNF-alpha-induced NF-kappaB/RelA Ser(276) phosphorylation and enhanceosome formation is mediated by an ROS-dependent PKAc pathway, Cell Signal 19(7) (2007) 1419–33. [DOI] [PubMed] [Google Scholar]
  • [42].Itoh K, Wakabayashi N, Katoh Y, Ishii T, Igarashi K, Engel JD, Yamamoto M, Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain, Genes Dev 13(1) (1999) 76–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Nguyen T, Nioi P, Pickett CB, The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress, J Biol Chem 284(20) (2009) 13291–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Denu JM, Tanner KG, Specific and reversible inactivation of protein tyrosine phosphatases by hydrogen peroxide: evidence for a sulfenic acid intermediate and implications for redox regulation, Biochemistry 37(16) (1998) 5633–42. [DOI] [PubMed] [Google Scholar]
  • [45].Lee SR, Kwon KS, Kim SR, Rhee SG, Reversible inactivation of protein-tyrosine phosphatase 1B in A431 cells stimulated with epidermal growth factor, J Biol Chem 273(25) (1998) 15366–72. [DOI] [PubMed] [Google Scholar]
  • [46].Kwon J, Lee SR, Yang KS, Ahn Y, Kim YJ, Stadtman ER, Rhee SG, Reversible oxidation and inactivation of the tumor suppressor PTEN in cells stimulated with peptide growth factors, Proc Natl Acad Sci U S A 101(47) (2004) 16419–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Hwang I, Uchida H, Dai Z, Li F, Sanchez T, Locasale JW, Cantley LC, Zheng H, Paik J, Cellular stress signaling activates type-I IFN response through FOXO3-regulated lamin posttranslational modification, Nat Commun 12(1) (2021) 640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Byrne DP, Shrestha S, Galler M, Cao M, Daly LA, Campbell AE, Eyers CE, Veal EA, Kannan N, Eyers PA, Aurora A regulation by reversible cysteine oxidation reveals evolutionarily conserved redox control of Ser/Thr protein kinase activity, Sci Signal 13(639) (2020). [DOI] [PubMed] [Google Scholar]
  • [49].Luanpitpong S, Chanvorachote P, Stehlik C, Tse W, Callery PS, Wang L, Rojanasakul Y, Regulation of apoptosis by Bcl-2 cysteine oxidation in human lung epithelial cells, Mol Biol Cell 24(6) (2013) 858–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Kato I, Maita H, Takahashi-Niki K, Saito Y, Noguchi N, Iguchi-Ariga SM, Ariga H, Oxidized DJ-1 inhibits p53 by sequestering p53 from promoters in a DNA-binding affinity-dependent manner, Mol Cell Biol 33(2) (2013) 340–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Kinumi T, Kimata J, Taira T, Ariga H, Niki E, Cysteine-106 of DJ-1 is the most sensitive cysteine residue to hydrogen peroxide-mediated oxidation in vivo in human umbilical vein endothelial cells, Biochem Biophys Res Commun 317(3) (2004) 722–8. [DOI] [PubMed] [Google Scholar]
  • [52].Kim H, An S, Ro SH, Teixeira F, Park GJ, Kim C, Cho CS, Kim JS, Jakob U, Lee JH, Cho US, Janus-faced Sestrin2 controls ROS and mTOR signalling through two separate functional domains, Nat Commun 6 (2015) 10025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Tao L, Lemoff A, Wang G, Zarek C, Lowe A, Yan N, Reese TA, Reactive oxygen species oxidize STING and suppress interferon production, Elife 9 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Cedervall T, Berggård T, Borek V, Thulin E, Linse S, Akerfeldt KS, Redox sensitive cysteine residues in calbindin D28k are structurally and functionally important, Biochemistry 44(2) (2005) 684–93. [DOI] [PubMed] [Google Scholar]
  • [55].Ranaghan MJ, Kumar RP, Chakrabarti KS, Buosi V, Kern D, Oprian DD, A highly conserved cysteine of neuronal calcium-sensing proteins controls cooperative binding of Ca2+ to recoverin, J Biol Chem 288(50) (2013) 36160–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Yan LJ, Sumien N, Thangthaeng N, Forster MJ, Reversible inactivation of dihydrolipoamide dehydrogenase by mitochondrial hydrogen peroxide, Free Radic Res 47(2) (2013) 123–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Maller C, Schröder E, Eaton P, Glyceraldehyde 3-phosphate dehydrogenase is unlikely to mediate hydrogen peroxide signaling: studies with a novel anti-dimedone sulfenic acid antibody, Antioxid Redox Signal 14(1) (2011) 49–60. [DOI] [PubMed] [Google Scholar]
  • [58].Kulathu Y, Garcia FJ, Mevissen TE, Busch M, Arnaudo N, Carroll KS, Barford D, Komander D, Regulation of A20 and other OTU deubiquitinases by reversible oxidation, Nat Commun 4 (2013) 1569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Cotto-Rios XM, Békés M, Chapman J, Ueberheide B, Huang TT, Deubiquitinases as a signaling target of oxidative stress, Cell Rep 2(6) (2012) 1475–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Dotsey EY, Jung KM, Basit A, Wei D, Daglian J, Vacondio F, Armirotti A, Mor M, Piomelli D, Peroxide-Dependent MGL Sulfenylation Regulates 2-AG-Mediated Endocannabinoid Signaling in Brain Neurons, Chem Biol 22(5) (2015) 619–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Legler PM, Kumaran D, Swaminathan S, Studier FW, Millard CB, Structural characterization and reversal of the natural organophosphate resistance of a D-type esterase, Saccharomyces cerevisiae S-formylglutathione hydrolase, Biochemistry 47(36) (2008) 9592–601. [DOI] [PubMed] [Google Scholar]
  • [62].Wani R, Qian J, Yin L, Bechtold E, King SB, Poole LB, Paek E, Tsang AW, Furdui CM, Isoform-specific regulation of Akt by PDGF-induced reactive oxygen species, Proc Natl Acad Sci U S A 108(26) (2011) 10550–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Nadeau PJ, Charette SJ, Landry J, REDOX reaction at ASK1-Cys250 is essential for activation of JNK and induction of apoptosis, Mol Biol Cell 20(16) (2009) 3628–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Nelson KJ, Bolduc JA, Wu H, Collins JA, Burke EA, Reisz JA, Klomsiri C, Wood ST, Yammani RR, Poole LB, Furdui CM, Loeser RF, H, J Biol Chem 293(42) (2018) 16376–16389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Cross JV, Templeton DJ, Oxidative stress inhibits MEKK1 by site-specific glutathionylation in the ATP-binding domain, Biochem J 381(Pt 3) (2004) 675–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Nakamura K, Hori T, Sato N, Sugie K, Kawakami T, Yodoi J, Redox regulation of a src family protein tyrosine kinase p56lck in T cells, Oncogene 8(11) (1993) 3133–9. [PubMed] [Google Scholar]
  • [67].Innes BT, Sowole MA, Gyenis L, Dubinsky M, Konermann L, Litchfield DW, Brandl CJ, Shilton BH, Peroxide-mediated oxidation and inhibition of the peptidyl-prolyl isomerase Pin1, Biochim Biophys Acta 1852(5) (2015) 905–12. [DOI] [PubMed] [Google Scholar]
  • [68].Chen CH, Li W, Sultana R, You MH, Kondo A, Shahpasand K, Kim BM, Luo ML, Nechama M, Lin YM, Yao Y, Lee TH, Zhou XZ, Swomley AM, Butterfield DA, Zhang Y, Lu KP, Pin1 cysteine-113 oxidation inhibits its catalytic activity and cellular function in Alzheimer’s disease, Neurobiol Dis 76 (2015) 13–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Kemble DJ, Sun G, Direct and specific inactivation of protein tyrosine kinases in the Src and FGFR families by reversible cysteine oxidation, Proc Natl Acad Sci U S A 106(13) (2009) 5070–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Kaiserova K, Srivastava S, Hoetker JD, Awe SO, Tang XL, Cai J, Bhatnagar A, Redox activation of aldose reductase in the ischemic heart, J Biol Chem 281(22) (2006) 15110–20. [DOI] [PubMed] [Google Scholar]
  • [71].Lowther WT, Brot N, Weissbach H, Honek JF, Matthews BW, Thiol-disulfide exchange is involved in the catalytic mechanism of peptide methionine sulfoxide reductase, Proc Natl Acad Sci U S A 97(12) (2000) 6463–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Rhee SG, Woo HA, Kang D, The Role of Peroxiredoxins in the Transduction of H, Antioxid Redox Signal 28(7) (2018) 537–557. [DOI] [PubMed] [Google Scholar]
  • [73].Lee SR, Yang KS, Kwon J, Lee C, Jeong W, Rhee SG, Reversible inactivation of the tumor suppressor PTEN by H2O2, J Biol Chem 277(23) (2002) 20336–42. [DOI] [PubMed] [Google Scholar]
  • [74].He M, Fan J, Jiang R, Tang WX, Wang ZW, Expression of DNMTs and genomic DNA methylation in gastric signet ring cell carcinoma, Mol Med Rep 8(3) (2013) 942–8. [DOI] [PubMed] [Google Scholar]
  • [75].Barrett WC, DeGnore JP, König S, Fales HM, Keng YF, Zhang ZY, Yim MB, Chock PB, Regulation of PTP1B via glutathionylation of the active site cysteine 215, Biochemistry 38(20) (1999) 6699–705. [DOI] [PubMed] [Google Scholar]
  • [76].van Montfort RL, Congreve M, Tisi D, Carr R, Jhoti H, Oxidation state of the active-site cysteine in protein tyrosine phosphatase 1B, Nature 423(6941) (2003) 773–7. [DOI] [PubMed] [Google Scholar]
  • [77].Crump KE, Juneau DG, Poole LB, Haas KM, Grayson JM, The reversible formation of cysteine sulfenic acid promotes B-cell activation and proliferation, Eur J Immunol 42(8) (2012) 2152–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Chen CY, Willard D, Rudolph J, Redox regulation of SH2-domain-containing protein tyrosine phosphatases by two backdoor cysteines, Biochemistry 48(6) (2009) 1399–409. [DOI] [PubMed] [Google Scholar]
  • [79].Godat E, Hervé-Grvépinet V, Veillard F, Lecaille F, Belghazi M, Brömme D, Lalmanach G, Regulation of cathepsin K activity by hydrogen peroxide, Biol Chem 389(8) (2008) 1123–6. [DOI] [PubMed] [Google Scholar]
  • [80].Paulsen CE, Truong TH, Garcia FJ, Homann A, Gupta V, Leonard SE, Carroll KS, Peroxide-dependent sulfenylation of the EGFR catalytic site enhances kinase activity, Nat Chem Biol 8(1) (2011) 57–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Xanthoudakis S, Curran T, Identification and characterization of Ref-1, a nuclear protein that facilitates AP-1 DNA-binding activity, EMBO J 11(2) (1992) 653–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Dansen TB, Smits LM, van Triest MH, de Keizer PL, van Leenen D, Koerkamp MG, Szypowska A, Meppelink A, Brenkman AB, Yodoi J, Holstege FC, Burgering BM, Redox-sensitive cysteines bridge p300/CBP-mediated acetylation and FoxO4 activity, Nat Chem Biol 5(9) (2009) 664–72. [DOI] [PubMed] [Google Scholar]
  • [83].Pineda-Molina E, Klatt P, Vázquez J, Marina A, García de Lacoba M, Pérez-Sala D, Lamas S, Glutathionylation of the p50 subunit of NF-kappaB: a mechanism for redox-induced inhibition of DNA binding, Biochemistry 40(47) (2001) 14134–42. [DOI] [PubMed] [Google Scholar]
  • [84].Delaunay A, Pflieger D, Barrault MB, Vinh J, Toledano MB, A thiol peroxidase is an H2O2 receptor and redox-transducer in gene activation, Cell 111(4) (2002) 471–81. [DOI] [PubMed] [Google Scholar]
  • [85].Conway ME, Poole LB, Hutson SM, Roles for cysteine residues in the regulatory CXXC motif of human mitochondrial branched chain aminotransferase enzyme, Biochemistry 43(23) (2004) 7356–64. [DOI] [PubMed] [Google Scholar]
  • [86].Nagahara N, Katayama A, Post-translational regulation of mercaptopyruvate sulfurtransferase via a low redox potential cysteine-sulfenate in the maintenance of redox homeostasis, J Biol Chem 280(41) (2005) 34569–76. [DOI] [PubMed] [Google Scholar]
  • [87].Fernández-Irigoyen J, Santamaría M, Sánchez-Quiles V, Latasa MU, Santamaría E, Muñoz J, Sánchez Del Pino MM, Valero ML, Prieto J, Avila MA, Corrales FJ, Redox regulation of methylthioadenosine phosphorylase in liver cells: molecular mechanism and functional implications, Biochem J 411(2) (2008) 457–65. [DOI] [PubMed] [Google Scholar]
  • [88].Morales Y, Nitzel DV, Price OM, Gui S, Li J, Qu J, Hevel JM, Redox Control of Protein Arginine Methyltransferase 1 (PRMT1) Activity, J Biol Chem 290(24) (2015) 14915–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Reichenbach J, Schubert R, Schwan C, Müller K, Böhles HJ, Zielen S, Anti-oxidative capacity in patients with ataxia telangiectasia, Clin Exp Immunol 117(3) (1999) 535–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Krüger A, Ralser M, ATM is a redox sensor linking genome stability and carbon metabolism, Sci Signal 4(167) (2011) pe17. [DOI] [PubMed] [Google Scholar]
  • [91].Guo Z, Deshpande R, Paull TT, ATM activation in the presence of oxidative stress, Cell Cycle 9(24) (2010) 4805–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Allen DM, van Praag H, Ray J, Weaver Z, Winrow CJ, Carter TA, Braquet R, Harrington E, Ried T, Brown KD, Gage FH, Barlow C, Ataxia telangiectasia mutated is essential during adult neurogenesis, Genes Dev 15(5) (2001) 554–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].LiCausi F, Hartman NW, Role of mTOR Complexes in Neurogenesis, Int J Mol Sci 19(5) (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Forsberg K, Wuttke A, Quadrato G, Chumakov PM, Wizenmann A, Di Giovanni S, The tumor suppressor p53 fine-tunes reactive oxygen species levels and neurogenesis via PI3 kinase signaling, J Neurosci 33(36) (2013) 14318–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Peltier J, O’Neill A, Schaffer DV, PI3K/Akt and CREB regulate adult neural hippocampal progenitor proliferation and differentiation, Dev Neurobiol 67(10) (2007) 1348–61. [DOI] [PubMed] [Google Scholar]
  • [96].Dickinson BC, Peltier J, Stone D, Schaffer DV, Chang CJ, Nox2 redox signaling maintains essential cell populations in the brain, Nat Chem Biol 7(2) (2011) 106–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Rharass T, Lemcke H, Lantow M, Kuznetsov SA, Weiss DG, Panáková D, Ca2+-mediated mitochondrial reactive oxygen species metabolism augments Wnt/β-catenin pathway activation to facilitate cell differentiation, J Biol Chem 289(40) (2014) 27937–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Marchetti B, Nrf2/Wnt resilience orchestrates rejuvenation of glia-neuron dialogue in Parkinson’s disease, Redox Biol 36 (2020) 101664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Baruch K, Deczkowska A, David E, Castellano JM, Miller O, Kertser A, Berkutzki T, Barnett-Itzhaki Z, Bezalel D, Wyss-Coray T, Amit I, Schwartz M, Aging. Aging-induced type I interferon response at the choroid plexus negatively affects brain function, Science 346(6205) (2014) 89–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Schmeisser H, Bekisz J, Zoon KC, New function of type I IFN: induction of autophagy, J Interferon Cytokine Res 34(2) (2014) 71–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Lee KH, Cha M, Lee BH, Neuroprotective Effect of Antioxidants in the Brain, Int J Mol Sci 21(19) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Chuikov S, Levi BP, Smith ML, Morrison SJ, Prdm16 promotes stem cell maintenance in multiple tissues, partly by regulating oxidative stress, Nat Cell Biol 12(10) (2010) 999–1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Chui A, Zhang Q, Dai Q, Shi SH, Oxidative stress regulates progenitor behavior and cortical neurogenesis, Development 147(5) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Wakabayashi N, Dinkova-Kostova AT, Holtzclaw WD, Kang MI, Kobayashi A, Yamamoto M, Kensler TW, Talalay P, Protection against electrophile and oxidant stress by induction of the phase 2 response: fate of cysteines of the Keap1 sensor modified by inducers, Proc Natl Acad Sci U S A 101(7) (2004) 2040–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Suzuki T, Muramatsu A, Saito R, Iso T, Shibata T, Kuwata K, Kawaguchi SI, Iwawaki T, Adachi S, Suda H, Morita M, Uchida K, Baird L, Yamamoto M, Molecular Mechanism of Cellular Oxidative Stress Sensing by Keap1, Cell Rep 28(3) (2019) 746–758.e4. [DOI] [PubMed] [Google Scholar]
  • [106].Brandes MS, Gray NE, NRF2 as a Therapeutic Target in Neurodegenerative Diseases, ASN Neuro 12 (2020) 1759091419899782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Robledinos-Antón N, Rojo AI, Ferreiro E, Núñez Á, Krause KH, Jaquet V, Cuadrado A, Transcription factor NRF2 controls the fate of neural stem cells in the subgranular zone of the hippocampus, Redox Biol 13 (2017) 393–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].L’Episcopo F, Tirolo C, Testa N, Caniglia S, Morale MC, Impagnatiello F, Pluchino S, Marchetti B, Aging-induced Nrf2-ARE pathway disruption in the subventricular zone drives neurogenic impairment in parkinsonian mice via PI3K-Wnt/β-catenin dysregulation, J Neurosci 33(4) (2013) 1462–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Ray S, Corenblum MJ, Anandhan A, Reed A, Ortiz FO, Zhang DD, Barnes CA, Madhavan L, A Role for Nrf2 Expression in Defining the Aging of Hippocampal Neural Stem Cells, Cell Transplant 27(4) (2018) 589–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Corenblum MJ, Ray S, Remley QW, Long M, Harder B, Zhang DD, Barnes CA, Madhavan L, Reduced Nrf2 expression mediates the decline in neural stem cell function during a critical middle-age period, Aging Cell 15(4) (2016) 725–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Wang GL, Jiang BH, Rue EA, Semenza GL, Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension, Proc Natl Acad Sci U S A 92(12) (1995) 5510–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Guzy RD, Hoyos B, Robin E, Chen H, Liu L, Mansfield KD, Simon MC, Hammerling U, Schumacker PT, Mitochondrial complex III is required for hypoxia-induced ROS production and cellular oxygen sensing, Cell Metab 1(6) (2005) 401–8. [DOI] [PubMed] [Google Scholar]
  • [113].Panchision DM, The role of oxygen in regulating neural stem cells in development and disease, J Cell Physiol 220(3) (2009) 562–8. [DOI] [PubMed] [Google Scholar]
  • [114].Li L, Candelario KM, Thomas K, Wang R, Wright K, Messier A, Cunningham LA, Hypoxia inducible factor-1α (HIF-1α) is required for neural stem cell maintenance and vascular stability in the adult mouse SVZ, J Neurosci 34(50) (2014) 16713–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Carrica L, Li L, Newville J, Kenton J, Gustus K, Brigman J, Cunningham LA, Genetic inactivation of hypoxia inducible factor 1-alpha (HIF-1α) in adult hippocampal progenitors impairs neurogenesis and pattern discrimination learning, Neurobiol Learn Mem 157 (2019) 79–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Beckervordersandforth R, Mitochondrial Metabolism-Mediated Regulation of Adult Neurogenesis, Brain Plast 3(1) (2017) 73–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Xavier JM, Morgado AL, Solá S, Rodrigues CM, Mitochondrial translocation of p53 modulates neuronal fate by preventing differentiation-induced mitochondrial stress, Antioxid Redox Signal 21(7) (2014) 1009–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Zhang Y, Hu W, NFκB signaling regulates embryonic and adult neurogenesis, Front Biol (Beijing) 7(4) (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Nakajima S, Kitamura M, Bidirectional regulation of NF-κB by reactive oxygen species: a role of unfolded protein response, Free Radic Biol Med 65 (2013) 162–174. [DOI] [PubMed] [Google Scholar]
  • [120].Lingappan K, NF-κB in Oxidative Stress, Curr Opin Toxicol 7 (2018) 81–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Hwang I, Oh H, Santo E, Kim DY, Chen JW, Bronson RT, Locasale JW, Na Y, Lee J, Reed S, Toth M, Yu WH, Muller FL, Paik J, FOXO protects against age-progressive axonal degeneration, Aging Cell 17(1) (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Santo EE, Paik J, FOXO in Neural Cells and Diseases of the Nervous System, Curr Top Dev Biol 127 (2018) 105–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Kim DY, Hwang I, Muller FL, Paik JH, Functional regulation of FoxO1 in neural stem cell differentiation, Cell Death Differ 22(12) (2015) 2034–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Yeo H, Lyssiotis CA, Zhang Y, Ying H, Asara JM, Cantley LC, Paik JH, FoxO3 coordinates metabolic pathways to maintain redox balance in neural stem cells, EMBO J 32(19) (2013) 2589–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Kreuz S, Fischle W, Oxidative stress signaling to chromatin in health and disease, Epigenomics 8(6) (2016) 843–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Ducker GS, Rabinowitz JD, One-Carbon Metabolism in Health and Disease, Cell Metab 25(1) (2017) 27–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Ye C, Sutter BM, Wang Y, Kuang Z, Tu BP, A Metabolic Function for Phospholipid and Histone Methylation, Mol Cell 66(2) (2017) 180–193.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Fawal MA, Jungas T, Kischel A, Audouard C, Iacovoni JS, Davy A, Cross Talk between One-Carbon Metabolism, Eph Signaling, and Histone Methylation Promotes Neural Stem Cell Differentiation, Cell Rep 23(10) (2018) 2864–2873.e7. [DOI] [PubMed] [Google Scholar]
  • [129].Benayoun BA, Pollina EA, Ucar D, Mahmoudi S, Karra K, Wong ED, Devarajan K, Daugherty AC, Kundaje AB, Mancini E, Hitz BC, Gupta R, Rando TA, Baker JC, Snyder MP, Cherry JM, Brunet A, H3K4me3 breadth is linked to cell identity and transcriptional consistency, Cell 158(3) (2014) 673–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Ulianov SV, Doronin SA, Khrameeva EE, Kos PI, Luzhin AV, Starikov SS, Galitsyna AA, Nenasheva VV, Ilyin AA, Flyamer IM, Mikhaleva EA, Logacheva MD, Gelfand MS, Chertovich AV, Gavrilov AA, Razin SV, Shevelyov YY, Nuclear lamina integrity is required for proper spatial organization of chromatin in Drosophila, Nat Commun 10(1) (2019) 1176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Carey BW, Finley LW, Cross JR, Allis CD, Thompson CB, Intracellular α-ketoglutarate maintains the pluripotency of embryonic stem cells, Nature 518(7539) (2015) 413–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Fawal MA, Davy A, Impact of Metabolic Pathways and Epigenetics on Neural Stem Cells, Epigenet Insights 11 (2018) 2516865718820946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Pietrocola F, Galluzzi L, Bravo-San Pedro JM, Madeo F, Kroemer G, Acetyl coenzyme A: a central metabolite and second messenger, Cell Metab 21(6) (2015) 805–21. [DOI] [PubMed] [Google Scholar]
  • [134].Hamilton LK, Dufresne M, Joppé SE, Petryszyn S, Aumont A, Calon F, Barnabé-Heider F, Furtos A, Parent M, Chaurand P, Fernandes KJ, Aberrant Lipid Metabolism in the Forebrain Niche Suppresses Adult Neural Stem Cell Proliferation in an Animal Model of Alzheimer’s Disease, Cell Stem Cell 17(4) (2015) 397–411. [DOI] [PubMed] [Google Scholar]
  • [135].Mews P, Donahue G, Drake AM, Luczak V, Abel T, Berger SL, Acetyl-CoA synthetase regulates histone acetylation and hippocampal memory, Nature 546(7658) (2017) 381–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Zhou W, Zhao T, Du J, Ji G, Li X, Ji S, Tian W, Wang X, Hao A, TIGAR promotes neural stem cell differentiation through acetyl-CoA-mediated histone acetylation, Cell Death Dis 10(3) (2019) 198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Bensaad K, Tsuruta A, Selak MA, Vidal MN, Nakano K, Bartrons R, Gottlieb E, Vousden KH, TIGAR, a p53-inducible regulator of glycolysis and apoptosis, Cell 126(1) (2006) 107–20. [DOI] [PubMed] [Google Scholar]
  • [138].Guillaumet-Adkins A, Yañez Y, Peris-Diaz MD, Calabria I, Palanca-Ballester C, Sandoval J, Epigenetics and Oxidative Stress in Aging, Oxid Med Cell Longev 2017 (2017) 9175806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Salminen A, Kaarniranta K, Kauppinen A, Crosstalk between Oxidative Stress and SIRT1: Impact on the Aging Process, Int J Mol Sci 14(2) (2013) 3834–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Ma CY, Yao MJ, Zhai QW, Jiao JW, Yuan XB, Poo MM, SIRT1 suppresses self-renewal of adult hippocampal neural stem cells, Development 141(24) (2014) 4697–709. [DOI] [PubMed] [Google Scholar]
  • [141].Prozorovski T, Schulze-Topphoff U, Glumm R, Baumgart J, Schröter F, Ninnemann O, Siegert E, Bendix I, Brüstle O, Nitsch R, Zipp F, Aktas O, Sirt1 contributes critically to the redox-dependent fate of neural progenitors, Nat Cell Biol 10(4) (2008) 385–94. [DOI] [PubMed] [Google Scholar]
  • [142].Casares-Crespo L, Calatayud-Baselga I, García-Corzo L, Mira H, On the Role of Basal Autophagy in Adult Neural Stem Cells and Neurogenesis, Front Cell Neurosci 12 (2018) 339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Schäffner I, Minakaki G, Khan MA, Balta EA, Schlötzer-Schrehardt U, Schwarz TJ, Beckervordersandforth R, Winner B, Webb AE, DePinho RA, Paik J, Wurst W, Klucken J, Lie DC, FoxO Function Is Essential for Maintenance of Autophagic Flux and Neuronal Morphogenesis in Adult Neurogenesis, Neuron 99(6) (2018) 1188–1203.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [144].Morgado AL, Xavier JM, Dionísio PA, Ribeiro MF, Dias RB, Sebastião AM, Solá S, Rodrigues CM, MicroRNA-34a Modulates Neural Stem Cell Differentiation by Regulating Expression of Synaptic and Autophagic Proteins, Mol Neurobiol 51(3) (2015) 1168–83. [DOI] [PubMed] [Google Scholar]
  • [145].Yazdankhah M, Farioli-Vecchioli S, Tonchev AB, Stoykova A, Cecconi F, The autophagy regulators Ambra1 and Beclin 1 are required for adult neurogenesis in the brain subventricular zone, Cell Death Dis 5 (2014) e1403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Filomeni G, De Zio D, Cecconi F, Oxidative stress and autophagy: the clash between damage and metabolic needs, Cell Death & Differentiation 22(3) (2015) 377–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [147].Hekimi S, Lapointe J, Wen Y, Taking a “good” look at free radicals in the aging process, Trends in Cell Biology 21(10) (2011) 569–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Fang C, Gu L, Smerin D, Mao S, Xiong X, The Interrelation between Reactive Oxygen Species and Autophagy in Neurological Disorders, Oxid Med Cell Longev 2017 (2017) 8495160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Folmes CD, Dzeja PP, Nelson TJ, Terzic A, Metabolic plasticity in stem cell homeostasis and differentiation, Cell Stem Cell 11(5) (2012) 596–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Stoll EA, Makin R, Sweet IR, Trevelyan AJ, Miwa S, Horner PJ, Turnbull DM, Neural Stem Cells in the Adult Subventricular Zone Oxidize Fatty Acids to Produce Energy and Support Neurogenic Activity, Stem Cells 33(7) (2015) 2306–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Shin J, Berg DA, Zhu Y, Shin JY, Song J, Bonaguidi MA, Enikolopov G, Nauen DW, Christian KM, Ming GL, Song H, Single-Cell RNA-Seq with Waterfall Reveals Molecular Cascades underlying Adult Neurogenesis, Cell Stem Cell 17(3) (2015) 360–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Doiron B, Cuif MH, Chen R, Kahn A, Transcriptional glucose signaling through the glucose response element is mediated by the pentose phosphate pathway, J Biol Chem 271(10) (1996) 5321–4. [DOI] [PubMed] [Google Scholar]
  • [153].Anastasiou D, Poulogiannis G, Asara JM, Boxer MB, Jiang JK, Shen M, Bellinger G, Sasaki AT, Locasale JW, Auld DS, Thomas CJ, Vander Heiden MG, Cantley LC, Inhibition of pyruvate kinase M2 by reactive oxygen species contributes to cellular antioxidant responses, Science 334(6060) (2011) 1278–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Grüning NM, Rinnerthaler M, Bluemlein K, Mülleder M, Wamelink MM, Lehrach H, Jakobs C, Breitenbach M, Ralser M, Pyruvate kinase triggers a metabolic feedback loop that controls redox metabolism in respiring cells, Cell Metab 14(3) (2011) 415–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Nicklin P, Bergman P, Zhang B, Triantafellow E, Wang H, Nyfeler B, Yang H, Hild M, Kung C, Wilson C, Myer VE, MacKeigan JP, Porter JA, Wang YK, Cantley LC, Finan PM, Murphy LO, Bidirectional transport of amino acids regulates mTOR and autophagy, Cell 136(3) (2009) 521–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, Perera RM, Ferrone CR, Mullarky E, Shyh-Chang N, Kang Y, Fleming JB, Bardeesy N, Asara JM, Haigis MC, DePinho RA, Cantley LC, Kimmelman AC, Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway, Nature 496(7443) (2013) 101–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Bahat A, Gross A, Mitochondrial plasticity in cell fate regulation, J Biol Chem 294(38) (2019) 13852–13863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Khacho M, Clark A, Svoboda DS, Azzi J, MacLaurin JG, Meghaizel C, Sesaki H, Lagace DC, Germain M, Harper ME, Park DS, Slack RS, Mitochondrial Dynamics Impacts Stem Cell Identity and Fate Decisions by Regulating a Nuclear Transcriptional Program, Cell Stem Cell 19(2) (2016) 232–247. [DOI] [PubMed] [Google Scholar]
  • [159].Khacho M, Clark A, Svoboda DS, MacLaurin JG, Lagace DC, Park DS, Slack RS, Mitochondrial dysfunction underlies cognitive defects as a result of neural stem cell depletion and impaired neurogenesis, Hum Mol Genet 26(17) (2017) 3327–3341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Mailloux RJ, Jin X, Willmore WG, Redox regulation of mitochondrial function with emphasis on cysteine oxidation reactions, Redox Biol 2 (2014) 123–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Khacho M, Slack RS, Mitochondrial dynamics in the regulation of neurogenesis: From development to the adult brain, Dev Dyn 247(1) (2018) 47–53. [DOI] [PubMed] [Google Scholar]
  • [162].Walton NM, Shin R, Tajinda K, Heusner CL, Kogan JH, Miyake S, Chen Q, Tamura K, Matsumoto M, Adult neurogenesis transiently generates oxidative stress, PLoS One 7(4) (2012) e35264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Schweitzer JS, Song B, Herrington TM, Park T-Y, Lee N, Ko S, Jeon J, Cha Y, Kim K, Li Q, Henchcliffe C, Kaplitt M, Neff C, Rapalino O, Seo H, Lee I-H, Kim J, Kim T, Petsko GA, Ritz J, Cohen BM, Kong S-W, Leblanc P, Carter BS, Kim K-S, Personalized iPSC-Derived Dopamine Progenitor Cells for Parkinson’s Disease, New England Journal of Medicine 382(20) (2020) 1926–1932. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES