Next Article in Journal
Somatic Copy Number Alterations in Colorectal Cancer Lead to a Differentially Expressed ceRNA Network (ceRNet)
Next Article in Special Issue
Exploring Myelin Dynamics in Demyelinating Disorders at the Molecular Level
Previous Article in Journal
miRNA Expression Profiling in Human Breast Cancer Diagnostics and Therapy
Previous Article in Special Issue
Therapeutic Plasma Exchange and Multiple Sclerosis Dysregulations: Focus on the Removal of Pathogenic Circulatory Factors and Altering Nerve Growth Factor and Sphingosine-1-Phosphate Plasma Levels
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Roles of Caloric Restriction Mimetics in Central Nervous System Demyelination and Remyelination

by
Despoina Kaffe
1,
Stefanos Ioannis Kaplanis
2,3 and
Domna Karagogeos
2,3,*
1
Department of Biology, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece
2
Department of Basic Science, School of Medicine, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece
3
Institute of Molecular Biology & Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), Vassilika Vouton, 70013 Heraklion, Greece
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2023, 45(12), 9526-9548; https://doi.org/10.3390/cimb45120596
Submission received: 2 October 2023 / Revised: 16 November 2023 / Accepted: 22 November 2023 / Published: 27 November 2023

Abstract

:
The dysfunction of myelinating glial cells, the oligodendrocytes, within the central nervous system (CNS) can result in the disruption of myelin, the lipid-rich multi-layered membrane structure that surrounds most vertebrate axons. This leads to axonal degeneration and motor/cognitive impairments. In response to demyelination in the CNS, the formation of new myelin sheaths occurs through the homeostatic process of remyelination, facilitated by the differentiation of newly formed oligodendrocytes. Apart from oligodendrocytes, the two other main glial cell types of the CNS, microglia and astrocytes, play a pivotal role in remyelination. Following a demyelination insult, microglia can phagocytose myelin debris, thus permitting remyelination, while the developing neuroinflammation in the demyelinated region triggers the activation of astrocytes. Modulating the profile of glial cells can enhance the likelihood of successful remyelination. In this context, recent studies have implicated autophagy as a pivotal pathway in glial cells, playing a significant role in both their maturation and the maintenance of myelin. In this Review, we examine the role of substances capable of modulating the autophagic machinery within the myelinating glial cells of the CNS. Such substances, called caloric restriction mimetics, have been shown to decelerate the aging process by mitigating age-related ailments, with their mechanisms of action intricately linked to the induction of autophagic processes.

Graphical Abstract

1. Introduction

Myelin is a lipid-rich multi-layered membrane structure that surrounds most vertebrate axons. It is characterized by a high lipid-to-protein ratio, containing 75–80% lipids (by dry weight) and 25–30% proteins [1]. A hallmark of myelinated fibers is saltatory conduction, a mechanism that enables the rapid and efficient propagation of action potentials along the axonal length. Beyond its insulating function, myelin also plays an active role in providing metabolic support to axons [2]. In the central nervous system (CNS), myelin is produced by oligodendrocytes (OLs), while in the peripheral nervous system (PNS), it is synthesized by Schwann cells (SCs) [3].
The dysfunction of myelinating glial cells within the CNS can result in the disruption of myelin, a phenomenon that can subsequently lead to axonal demyelination and contribute to eventual axonal degeneration [4]. In response to demyelination in the CNS, the formation of new myelin sheaths occurs through the homeostatic process of remyelination, facilitated by the differentiation of newly formed OLs [5]. It is important to note, however, that remyelination is often inadequate for fully replicating the original myelin ultrastructure [5].
Apart from OLs, microglia and astrocytes, the two other main glial cell types of the CNS, play a pivotal role in remyelination [6,7,8]. Microglia constitute the resident macrophages within the CNS and display dynamic diversity [9,10]. Following a demyelination insult, they can phagocytose myelin debris, which is vital for recruiting and differentiating oligodendrocyte progenitor cells (OPCs), and they secrete growth factors and chemotactic substances. They also alter the extracellular matrix to support OPCs, aiding remyelination [11,12,13,14]. The developing neuroinflammation in the demyelinated region triggers the activation of astrocytes, the third glial population involved in remyelination, in a process known as reactive astrogliosis [15]. Activated astrocytes play a dual role, sometimes favoring or hindering remyelination based on their specific phenotype [16,17]. Astrocytes can directly impact remyelination because they recruit microglia to the lesion site and, thus, modulate the removal of myelin debris, which is essential for the resolution of the inflammatory response and, ultimately, remyelination [18]. Additionally, they modulate the extracellular matrix, affecting OPC proliferation and differentiation [19]. Modulating the expression profile of these glial cell types can enhance the likelihood of successful CNS remyelination.
In addition, recently, studies have implicated autophagy as a pivotal pathway in glial cells, playing a significant role in both their maturation and the maintenance of myelin. A study by Bankston and colleagues reveals that autophagy deficiency hinders OL differentiation both in vitro and in vivo. Specifically, their findings indicate that inhibiting autophagy alters the ultrastructure of myelin, thus underscoring the critical role of autophagy in OLs for proper myelination. Additionally, the observed enrichment of autolysosomes in OL processes suggests a specialized role of autophagy in these cellular processes. Notably, this research group also reports an increase in autophagic flux during oligodendrocyte differentiation in vitro [20]. Two recent consecutive studies have highlighted the essential role of autophagy in OLs not only in myelination but also in maintaining myelin throughout the lifespan of mice. Aber et al. demonstrate that OLs orchestrate the autophagic machinery to turnover myelin sheaths during adulthood, as autophagy deficiency leads to increased myelin deposition, a phenomenon that intensifies over time [21]. Furthermore, Ktena et al. corroborate the hypothesis that autophagy plays a pivotal role in myelin maintenance. The inhibition of autophagy, both genetically and pharmacologically, results in defects in OL maturation in vitro. The ablation of the core autophagic gene atg5 in OLs in vivo in 2.5 month-old mice, following the completion of myelination, leads to an excess of both PLP protein and mRNA levels at the age of 6 months, also implicating the autophagic machinery in PLP mRNA degradation. Moreover, conditional knockout mice in which autophagy is ablated in OLs exhibit myelin decompaction with subsequent axonal degeneration and behavioral deficits at the age of 6 months [22]. Collectively, these recent findings regarding autophagy and its pivotal role in CNS myelination and myelin maintenance have made autophagy an attractive therapeutic target for repairing myelin insults and/or abnormalities.
In the context of this Review, we focused on substances capable of modulating the autophagic machinery within the myelinating glial cells of the CNS. This modulation is achieved through a process akin to caloric restriction (CR), and the substances are named caloric restriction mimetics (CRMs) [23,24]. Dietary or caloric restriction is defined as the deliberate reduction of food consumption while maintaining proper nutrition, irrespective of the selective restriction of specific food groups. After nearly a century of extensive investigation across various model organisms, including Saccharomyces cerevisiae, Drosophila melanogaster, rodents, and non-human primates, and the analysis of human epidemiological data, CR is presently widely acknowledged for its capacity to enhance the longevity of organisms and decelerate the aging process [25,26]. Furthermore, it is recognized for its ability to mitigate age-related ailments, with its mechanisms of action being intricately linked to the induction of autophagic processes [27].
In the specific context of the CNS myelin, the advantageous effects of CR have been demonstrated. Piccio and colleagues provided compelling evidence illustrating the efficacy of a chronic CR regimen in enhancing the clinical outcomes of both relapsing–remitting and chronic experimental autoimmune encephalomyelitis (EAE) models [23]. These improvements in clinical outcomes were further validated through the observation of reduced severity in CNS pathology among the mice subjected to CR. Furthermore, the beneficial impact of CR on myelin recovery has also been observed in the cuprizone (CPZ) model of demyelination. Studies have shown that CR fosters the remyelination process by significantly increasing the survival rates of OLs. Additionally, it leads to a decrease in both astrogliosis and microgliosis within the corpus callosum (cc) of mice with CPZ-induced demyelination [24]. Nevertheless, owing to the systemic and extensive impacts of CR, unraveling the specific signaling pathways and the exact mechanisms underpinning its favorable effects mediated via autophagy can prove to be a complex endeavor. Over the course of decades of research, several hypotheses have arisen, among which the predominant one suggests that CR primarily acts to preserve cellular homeostasis and overall health [28].
The depletion of nutrients leads to a reduction in intracellular acetyl coenzyme A (AcCoA) levels, concurrent with the deacetylation of cellular proteins. Within this conceptual framework, there are three potential approaches to replicate these effects: (i) decrease cytosolic AcCoA levels by disrupting its biosynthesis; (ii) inhibit acetyltransferases, enzymes responsible for transferring acetyl groups from AcCoA to various molecules; or (iii) promote the activity of deacetylases, which facilitate the removal of acetyl groups from leucine residues [27,29,30]. The impact of CR can be replicated through the use of specific pharmacological agents referred to as CRMs. These agents, including metformin, nicotinamide adenine dinucleotide (NAD+) precursors, and resveratrol, are non-toxic natural compounds, which exhibit the capability to modulate the autophagic flux by triggering pathways similar to those activated during nutrient deprivation [31]. For the reasons mentioned above, we will focus this Review on the roles of metformin, NAD+ precursors, and resveratrol, mainly in demyelinating diseases.

2. Metformin

Metformin is a derivative of the natural guanidines present in the plant Galega officinalis and is widely used as a drug for type II diabetes, primarily operating through the inhibition of hepatic gluconeogenesis [32,33,34]. Beyond its established role in managing type II diabetes, metformin administration seems to exert beneficial effects on diseases, including cancer [35,36], cardiovascular disease [37], and obesity [38], as well as on neurodegeneration [39] and aging [40]. However, the precise underlying mechanisms responsible for these diverse therapeutic benefits remain to be elucidated [41].
Metformin inhibits mitochondrial complex I [42], a crucial component of the electron transport chain, thereby leading to decreased cellular ATP/ADP and ATP/AMP ratios and, thus, adenosine 5′-monophosphate-activated protein kinase (AMPK) activation [43,44]. Importantly, metformin-mediated AMPK activation exerts regulatory effects on cell energy metabolism and the autophagic cascade by reducing the activity of EP300 acetyl-transferase [45] and simultaneously enhancing the activity of sirtuin 1 (SIRT1) protein deacetylase [46]. Furthermore, a recent study has highlighted an additional mode of AMPK activation by metformin, which directly acts on the lysosomal vacuolar-type ATPase (v-ATPase), promoting the formation of the v-ATPase-regulator-AXIN/liver kinase B1 (LKB1)-AMPK complex in the lysosome, ultimately leading to AMPK activation. Interestingly, when the v-ATPase-regulator complex is engaged by AXIN, it inactivates mammalian target of rapamycin complex 1 (mTORC1), demonstrating that metformin’s effects extend beyond AMPK activation, also encompassing mTORC1 inactivation [47]. The activation of AMPK together with the inactivation of mTORC1, the two major energy and nutrient sensors of the cell, induce the activation of the autophagic pathway [48].
It has long been reported that metformin exerts neuroprotective effects on several neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and Huntington’s disease (HD). Chronic metformin administration was found to ameliorate synaptic malfunctions and cognitive impairment in the amyloid precursor protein (APP)swe/presenilin-1(PS1)DE9 (APP/PS1) mouse model of AD via the inhibition of cyclin-dependent kinase 5 (Cdk5) activity [49]. In the same mouse model of early-onset AD, metformin promoted the phagocytosis of pathological amyloid-β (Aβ) and tau proteins by microglia via the enhancement of the autophagic pathway, thus reducing the abundance of Aβ deposits and severity of neuritic plaque (NP) tau-pathology [50]. Metformin was also found to exert neuroprotective effects on dopaminergic neurodegeneration and alpha-synuclein aggregation in Caenorhabditis elegans models of PD [51], while it alleviated motor and neuropsychiatric manifestations in the zQ175 mouse model of HD [52].
Even though most of the current evidence suggests a beneficial effect of metformin on the prevention of AD in humans, its efficacy seems to be controversial. Recently, an observational study has indicated that metformin was associated with slower cognitive decline and reduced risk of dementia in patients with type II diabetes [53]. Furthermore, a randomized, double-blinded, placebo-controlled crossover pilot study demonstrated that metformin is safe and well tolerated by individuals, while being able to penetrate the blood–brain barrier [54]. Interestingly, metformin improved the executive function and tended to ameliorate memory, learning, and attention [54]. However, results from a prospective trial revealed that metformin impaired cognitive performance and that this effect was, at least in part, mediated by metformin-induced vitamin B12 deficiency [55]. This controversy could be attributed to different sample sizes, statistical methods, and drug administration, suggesting that more clinical trials need to be conducted [40].
In line with the multitude of evidence indicating metformin’s favorable results in neurodegenerative conditions, many recent studies have diligently scrutinized the potential therapeutic implications of metformin in the context of multiple sclerosis (MS) [56]. MS, a complex and heterogeneous neurodegenerative disorder affecting the CNS, is primarily characterized by profound demyelination, inflammation, and reactive gliosis [57]. Metformin treatment was shown to protect against intense demyelination in the cc of the CPZ-induced demyelination mouse model, when administered with the copper chelator CPZ, by attenuating reactive microgliosis and astrogliosis in the cc (Figure 1 and Figure 2) [58,59].
Largani et al. attributed the beneficial effects of metformin on myelin maintenance and reduced gliosis to its ability to reduce oxidative stress and upregulate antioxidant enzymes. There have been reports suggesting that reactive oxygen species (ROS) can regulate the expression of pro-inflammatory genes in microglia [60] and stimulate astrocytes to secrete inflammatory cytokines [61]. Abdi and his colleagues showed that metformin reduced levels of pro-inflammatory microglia markers through suppressing nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κΒ) in the CPZ model of MS, an effect that was accompanied by the delayed initiation of gliosis. Moreover, metformin administration was shown to decrease inducible nitric oxide synthase (iNOS) mRNA levels in EAE mice (Figure 1) [62] as well as to protect myelin and promote an anti-inflammatory microglial phenotype that promotes the clearance of myelin debris in a rat spinal cord injury model (Figure 1) [63]. In this case, the effects of metformin were mediated by the induction of autophagy through the activation of AMPK and the inhibition of mTORC1 [63]. These results indicate that metformin can act in favor of a less inflammatory environment under demyelinating conditions, thus enabling the physiological process of remyelination to take over demyelination. In particular, metformin administration during the recovery period significantly promoted the recruitment of intermediate and premature OPCs to the lesion site in favor of the remyelination process in a CPZ-induced demyelination mouse model [64]. In this case, accelerated myelin recovery upon metformin treatment was mediated by AMPK activation and m-TORC inactivation in mature OLs (Figure 3), indicating a possible implication of the autophagic machinery in the recovery process.
Despite the positive impacts of metformin on myelin protection and recovery through the inhibition of mTOR, it has been demonstrated that mTOR signaling regulates the developmental myelination of the CNS [65]. In particular, the ablation of raptor, the defining subunit of mTORC1, in OLs results in impaired OPC differentiation and delayed initiation of myelination in the spinal cords of mutant mice, which are also characterized by the formation of thinner myelin sheaths [65]. Interestingly, mTORC1 signaling driven by phosphoinositide 3-kinase (PI3K)/Akt, rather than ERK1/2, regulates the differentiation of progenitors, whereas both pathways converge at the level of mTORC1 to modulate myelin growth during active myelination [66]. Nevertheless, it has been shown that the prevention of the expression of tuberous sclerosis complex 1 (TSC1), a suppressor of mTOR signaling, resulted unexpectedly in hypomyelination during development [67]. On the other hand, the loss of TSC1 in adult OPCs enhanced remyelination and increased myelin thickness following lysolecithin (LPC)-induced focal demyelination [68], indicating that the deficiency in mTOR suppressors may exert either beneficial or detrimental effects on the differentially regulated processes of developmental myelination and remyelination. Therefore, more research is warranted to elucidate the interactions between AMPK and mTOR upon metformin administration.
Regarding AMPK, it is a cellular energy regulator found in many types of brain cells, including neurons, astrocytes, and microglia, as well as OLs [69]. Metformin treatment accelerates the differentiation of OLs in an AMPK-dependent manner, also requiring active glycolysis and/or oxidative phosphorylation to mediate OL differentiation [70]. The same study demonstrates the potential of metformin to improve myelin recovery from CPZ-induced demyelination by promoting OL differentiation in vivo [70]. Metformin-mediated AMPK activation seems to protect OLs against cytokine toxicity and oxidative stress rescuing their loss in the spinal cords of EAE rats, thus attenuating the clinical impairments of the disease and restoring the CNS integrity [71]. These immunomodulatory activities of AMPK signaling are concomitant with the stimulation of neurotrophic factor production in astrocytes within the CNS, which subsequently provides a myelinogenic environment for OLs (Figure 1) [71]. Furthermore, metformin treatment leads to increased synthesis of neurotrophic factors, like nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and ciliary neurotrophic factor (CNTF), while it also induces the expression of mature oligodendrocyte markers and the activation of AMPK in the CPZ-induced demyelination mouse model [72]. Because neurotrophic factors are reported to enhance OPC survival, migration, proliferation, differentiation, and maturation [73], it is suggested that metformin enhances the secretion of these factors during the recovery phase after demyelination, thus affecting the migration and differentiation of oligodendrocyte transcription factor 2 (Olig2)+ cells in favor of remyelination, effects that are mediated by AMPK activation [72].
Apart from its AMPK-mediated effects on remyelination, metformin was also found to act via the phosphorylation of the histone acetyltransferase CREB-binding protein (CBP), ultimately promoting OPC recruitment and differentiation to the lesion site in an LPC-induced focal demyelination mouse model. In particular, in vitro experiments confirmed that CBP Ser436 phosphorylation is required for metformin to promote the differentiation of OPCs into mature OLs. However, it is not responsible for metformin-induced OPC proliferation, an effect that was connected with the ability of metformin to block autophagy at early stages (Figure 3) [74].
When the remyelination process is delayed or fails, demyelinated axons are susceptible to irreversible degeneration, which can eventually lead to neuronal death [75]. The deceleration of remyelination that occurs in aging is marked by the deficient recruitment of OPCs to the site of the injury, coupled with the delayed progression in their differentiation into mature OLs [76]. Reversing the age-related intrinsic deficiencies of OPCs that are associated with their inability to respond to pro-differentiation factors was able to enhance OPC differentiation and remyelination in aged animals [77]. In this study, metformin was found to improve the mitochondrial function of aged OPCs by modulating the AMPK pathway and to restore the CNS remyelination capacity in aged rats, following ethidium bromide-induced focal demyelination in the cerebellar white matter (Figure 4). The authors postulated that metformin’s impact on remyelination could also be attributed to its capacity to enhance DNA repair and induce autophagy, both of which are established effects associated with metformin [78,79].
Regarding its functional behavioral effects, metformin treatment improved motor impairment and reduced anxiety in the CPZ-induced demyelination mouse model [64], while it also improved the social interaction of juvenile mice in an LPC-induced focal demyelination model [74]. These results render metformin a promising remyelinating agent that could treat neural deficits and impaired social behavior, a common symptom of white-matter-demyelinating diseases.

3. NAD+ Precursors

Nicotinic acid (NA), commonly referred to as niacin or vitamin B3, along with nicotinamide (NAM), its amide derivative, nicotinamide riboside (NR), and nicotinamide mononucleotide (NMN), serve as precursors for NAD+ (Figure 5). These compounds are available in various dietary products of both animal and plant origin, and they exhibit CRM-like properties [80,81]. NA is converted to NAD+ via the Preiss–Handler pathway, while NAM and NR enter the NAD+ salvage pathway, playing a pivotal role in the maintenance of cellular NAD+ levels [82]. NMN is synthesized from NAM by nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting NAD+ biosynthetic enzyme in mammals, as well as from NR by nicotinamide riboside kinases (NRKs), effectively bypassing the need for NAMPT (Figure 5) [83]. Accumulating evidence suggests that NAD+ intermediates not only prolong healthspan and/or lifespan [84,85,86], compensating for reduced NAD+ levels during aging, but also seem to be an effective intervention for various age-associated diseases, including cardiovascular diseases [87,88], cancer [89,90], and neurodegenerative disorders [91].
NAD+ plays a dual and pivotal role in cellular responses, serving as an essential coenzyme for enzymes facilitating oxidation–reduction reactions and as a co-substrate for NAD+-consuming enzymes. These enzymes compete for bioavailable NAD+ and belong into three classes: the cyclic ADP-ribose (cADPR) synthases, such as CD38; the poly (ADP-ribose) polymerase (PARP) protein family; and the sirtuin family of deacetylases (Figure 5) [92,93].
Specifically, CD38 plays important roles in many physiological processes, including glucose homeostasis, inflammation, and neuroprotection. Its deletion and the subsequent elevation in NAD+ levels protect against high-fat diet (HFD)-induced obesity [94], inflammatory reactions of microglia and astrocytes, and ROS, while it improves CPZ-induced demyelination and neurodegeneration [95,96]. PARP proteins mediate ADP-ribosylation and act as DNA-damage sensors [97]. It has been reported that the upregulation of PARP induces OL death, whereas its inhibition reduces CPZ-induced demyelination by suppressing p38 mitogen-activated protein kinases (p-38-MAPK) and JNK activation and increasing the activation of the PI3K/Akt pathway [98].
Among the three classes of NAD+-consuming enzymes, sirtuins are the most well studied. Upon increased NAD+ levels, SIRT1 is activated, leading to the deacetylation of critical proteins of the autophagic pathway, including autophagy-related gene 5 (Atg5), Atg7, and microtubule-associated protein 1 light chain 3 (LC3), thus inducing autophagy, as well as to the deacetylation of transcription factors, like NF-κΒ, thus regulating inflammatory signaling [99,100]. Additionally, the administration of NA has been shown to inhibit vascular inflammation in vivo along with the suppression of the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome in vascular endothelial cells in vitro via SIRT1 upregulation [101,102]. Apart from SIRT1, NAD+ supplementation has also been found to mediate the activation of sirtuin 2 (SIRT2). Specifically, NAD+ administration increased intracellular ATP levels via the activation of SIRT2, which regulates Akt phosphorylation in BV2 microglial cells [103], while NR treatment alleviated cisplatin-induced peripheral neuropathy in a SIRT2-dependent manner [104].
NAD+ supplementation was demonstrated to exert neuroprotective effects on various neurodegenerative diseases, including AD, PD, and HD. NR treatment reduced neuroinflammation in an APP/PS1 mouse model of AD, promoting the protective, phagocyting phenotype of microglia, while it also improved cognitive and synaptic functions [105]. A very recent study has highlighted NMN as a regulator of the gut microbiota, which exerted positive effects on AD [106]. Furthermore, NAM administration significantly protected against neuronal loss and attenuated motor dysfunction, oxidative stress, and neuroinflammation in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD [107], while it also provided neuroprotection in the 3-nitropropionic acid-induced animal model of HD [108].
Apart from these pre-clinical studies, multiple clinical trials are currently being conducted to evaluate the safety and the effects of NAD+ precursors on neurological disorders. In particular, NR has been demonstrated to be orally bio-available without serious adverse effects [109,110]. A recent randomized, placebo-controlled, crossover trial of oral NR supplementation has indicated increased levels of NAD+ in plasma extracellular vesicles enriched for neuronal origin (NEVs). Increased NAD+ levels were accompanied by decreased levels of Aβ42 and of the activated kinases pJNK and pERK1/2, which are implicated in AD [111]. Furthermore, in a randomized, placebo-controlled, phase I clinical trial, oral NR administration increased cerebral NAD levels in individuals with PD, an effect that was associated with the downregulation of inflammatory cytokines and general clinical improvement [112]. Regarding NAM, a new clinical trial (NCT03061474) is investigating whether NAM can reduce the phosphorylation of the tau protein found in cerebrospinal fluid (CSF) in people with mild cognitive impairment or mild AD [113].
In the same context of neurodegenerative conditions, NAD+ precursors have been shown to exert beneficial effects on demyelinating, neuroinflammatory diseases, like MS. NAD+ treatment was shown to alleviate demyelination and neuroinflammation in both the spinal cord [114] and the optic nerve [115] of a murine EAE model. Its administration exhibited a marked reduction in the activation of microglia and astrocytes, as well as in the expression of pro-inflammatory cytokines, while it facilitated the expression of anti-inflammatory cytokines, thereby fostering a less inflammatory milieu in vivo, which could support the remyelination process (Figure 1 and Figure 2) [114,115]. Notably, successful remyelination hinges upon the activities of OPCs. In response to myelin damage, these cells proliferate and migrate to lesion sites, where they mature to myelin-forming cells [116]. Guo et al. demonstrated that NAD+ supplementation effectively mitigates apoptosis among OLs and concurrently facilitates the recruitment and proliferation of OPCs in the optic nerve of mice with EAE. These beneficial effects of NAD+ in optic neuritis were orchestrated through the activation of the SIRT1-signaling pathway (Figure 3) [115]. Additionally, Wang et al. attributed the beneficial outcomes of NAD+ administration to the induction of autophagy because its inhibition abolished the protective effects of NAD+ [114]. Given the established role of SIRT1 deacetylase as an autophagy inducer [100], both studies converge on the critical role of the autophagic pathway in ameliorating EAE symptoms after NAD+ treatment.
In addition to NAD+ supplementation, numerous pre-clinical investigations have explored NAD+ precursors as potential therapeutic strategies for alleviating symptoms associated with MS. A recent study from our laboratory has revealed that NAM treatment resulted in a substantial augmentation in myelin production at the lesion site in the cc of an LPC-induced focal demyelination mouse model, concurrently with a reduction in microgliosis and astrogliosis (Figure 1 and Figure 2). Importantly, NAM treatment did not exert a direct influence on oligodendrocyte lineage cells, thereby suggesting that it accelerated the overall myelin production under demyelinating conditions by mitigating both microgliosis and astrogliosis [117]. Furthermore, the same study indicated that NAM directly affected microglial and astrocyte polarization toward their anti-inflammatory phenotypes, thus fostering a beneficial, less inflammatory microenvironment for remyelination. In addition to NAM, NR pre-treatment attenuated inflammatory responses, glial activation, and subsequent neurodegeneration in the brain of a lipopolysaccharide (LPS)-injected mouse model (Figure 1 and Figure 2) [118]. Despite these promising findings, the precise molecular mechanisms underpinning the NAD+-mediated regulation of glial activity remain elusive. Kaplanis et al. suggested that the shift in astrocytes toward their anti-inflammatory phenotype arises, at least in part, from the induction of autophagy, as observed in primary astrocyte cultures following NAM treatment (Figure 1) [117]. Notably, a recent study has identified a correlation between the induction of autophagy and the suppression of the inflammatory phenotype in astrocytes [119]. Regarding microglia, it appears that NAD+-dependent deacetylase SIRT2 inhibits pro-inflammatory responses through the deacetylation of NF-κΒ [120], indicating the participation of different NAD+-dependent pathways in glial phenotype commitment.
Among all seven sirtuins, SIRT2 is the most abundantly expressed in the brain, primarily residing in the cytoplasm of mature OLs but also present in neurons, astrocytes, and microglia [121,122]. Ma et al. further demonstrated that SIRT2 is predominantly expressed in the nuclei of postnatal OPCs during myelin development and changes its expression pattern in mature OLs, where it is found in the cytoplasm. Following a demyelination injury induced by LPC, SIRT2 is re-expressed in the majority of OPCs, primarily localizing within the OPC nuclear compartment in young adult mice. However, this re-expression and nuclear localization of SIRT2 declines with aging. Interestingly, β-NMN supplementation rescues SIRT2 nuclear localization in aged mice and affects the myelin status. In particular, it delays myelin aging under normal aging conditions and influences myelin compaction and thickness after a focal demyelinating LPC-induced lesion, thus enhancing remyelination by promoting the differentiation of OPCs (Figure 4) [123].
Enhanced remyelination was also observed in middle-aged animals upon niacin (NA) treatment in an LPC-induced focal demyelination mouse model [124]. Notably, the aging process is associated with delayed microglial recruitment to the lesion site and deficient phagocytosis, contributing to the establishment of an inhibitory microenvironment [125,126]. Rawji et al. demonstrated that NA administration enhanced the phagocytic activity of microglia in middle-aged animals, thus promoting the clearance of myelin debris from the lesion site and the recruitment of OPCs in favor of the remyelination process (Figure 4) [124]. Finally, NAM treatment promoted the maturation of OPCs and enhanced remyelination after stroke. NAM-treated animals had increased motor, sensory, and cognitive functions, and this functional remyelination was mediated by the BDNF/tropomyosin receptor kinase B (TrkB) pathway (Figure 3) [127]. Because BDNF is reported to enhance myelination via a direct effect on OLs [128], it is plausible that BDNF mediates the maturation of OPCs during remyelination.
These investigations highlight the favorable impacts of NAD+ and its precursors in age-related diseases, particularly in neurodegenerative and demyelinating diseases, like MS, rendering them promising therapeutic agents. Nevertheless, it is evident that distinct NAD+ precursors manifest their beneficial effects by targeting diverse molecules/pathways in various cell types of the CNS and within different mouse models of MS. Hence, it becomes imperative to discern the optimal precursor based on the considerations of absorption, kinetics, and specific MS symptoms. Finally, given that the majority of research efforts have centered on SIRT1 activation upon NAD+ treatment, it is equally important to delve into the roles of the other two classes of NAD+-depleting enzymes under demyelinating conditions.

4. Resveratrol

Resveratrol (3,4′,5-trihydroxystilbene) is a natural polyphenol that acts as a phytoalexin and is found in a wide variety of foods, including blueberries and peanuts, as well as grapes and products derived from them, like red wine [129,130]. Ever since resveratrol’s potent anticancer properties were highlighted by Jang in 1997 [131], both experimental and epidemiological studies have been conducted to elucidate its diverse bioactivities and, consequentially, its health advantages. Interestingly, resveratrol has a positive impact on a wide spectrum of diseases, including heart diseases [132], diabetes [133], cancer [134], obesity [135], and neurodegenerative diseases [136,137], while it also exerts beneficial effects on aging [138,139].
Mechanistically, resveratrol is mainly associated with the activation of the NAD+-dependent deacetylase SIRT1. Once activated, SIRT1 can deacetylate the core proteins of the autophagic pathway, like Atg5 and Atg7, leading to the induction of this pathway [100,140]. Furthermore, the induction of SIRT1 activation by resveratrol necessitates its phosphorylation by LKB1 in multiple cell lines, subsequently resulting in the deacetylation of the peroxisome proliferator-activated receptor-gamma coactivator-1-alpha (PGC-1a) transcriptional co-activator, which regulates mitochondrial biogenesis and respiration [141,142]. Beyond SIRT1, resveratrol also engages AMPK as a target [143]. Its activation upon resveratrol treatment was found to rescue Aβ-mediated neurotoxicity in human neural stem cells (hNSCs) [144] as well as oxygen and glucose deprivation in human SH-SY5Y neuroblastoma cells [145]. Because AMPK has been shown to activate SIRT1 through an indirect increase in cellular NAD+ levels [146], there is clearly a dynamic interaction between the two pathways. Finally, it is worth mentioning that resveratrol exhibits anti-inflammatory properties by suppressing the production of ROS and downregulating NF-κΒ [147].
Resveratrol has been documented for its neuroprotective potential in various CNS disorders, notably AD and PD. In particular, resveratrol treatment has been shown to mitigate neuroinflammation and reduce Aβ accumulation in the brains of 3×Tg-AD mice [148]. Furthermore, it inhibited tau aggregation and cytotoxicity in vitro, and it reduced the levels of phosphorylated tau, neuroinflammation, and synapse loss in the brain of a PS19 mouse model of AD, thus rescuing the cognitive deficits [149]. Resveratrol treatment was also able to ameliorate motor and cognitive impairments in an A53T α-synuclein mouse model of PD by diminishing the levels of α-synuclein aggregates and reducing microgliosis, astrocytosis, and oxidative-stress levels within the brain [150].
Meanwhile, the effects of resveratrol on neurological disorders are evaluated through clinical trials. One of the first studies evaluating the effects of resveratrol on individuals with mild to moderate AD was conducted by Turner et al. in 2015. This randomized, double-blinded, placebo-controlled phase II study indicated that resveratrol is safe and well tolerated by patients, while it could also penetrate the blood–brain barrier because it was detectable in the CSF. However, neuroprotective benefits could not be detected in this study, while the longer AD duration, measured in years from the diagnosis, in the placebo-treated group should be taken into consideration [151]. In the next step, the same research group analyzed samples of CSF and plasma from a subset of AD subjects with CSF Aβ 42 concentrations of <600 ng/mL. In this subset analysis, resveratrol decreased the levels of metalloproteinase (MMP) 9 in the CSF, suggesting increased maintenance of the blood–brain barrier and reduced infiltration of immune cells, while it also regulated neuroinflammation, induced adaptive immunity, and mitigated progressive cognitive decline [152].
The established capacity of resveratrol to mitigate inflammation and attenuate gliosis in the context of neurodegenerative pathologies endows it with substantial therapeutic potential for addressing demyelinating diseases, like MS. MS is predominantly characterized by pronounced inflammatory responses orchestrated by microglia, which can participate in mechanisms of tissue repair and injury depending on their activation state [14,153]. Traditionally, microglia have been categorized into pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes; however, this categorization appears to be simplistic. The use of new technologies, including single-cell RNA sequencing, has led to the identification of intermediate subpopulations that display a combination of pro- and anti-inflammatory markers, suggesting that microglial activation is a dynamic process [154,155,156]. A crucial requirement for the achievement of successful remyelination in MS is a switch toward the M2 activation state [157].
Resveratrol was shown to not only suppress microglial polarization toward the M1 phenotype but also promote the M2 phenotype of LPS-stimulated BV2 microglial cells in vitro and of microglial cells in vivo, in a model of systemic LPS administration that leads to brain inflammation (Figure 1) [158]. The neuroprotective role of resveratrol in microglial polarization was mainly attributed to PGC-1a activation, which can not only halt M1 polarization by suppressing NF-κB phosphorylation and the expression of inflammatory cytokines, like tumor necrosis factor alpha (TNF-α), but also interact with transcription factor signal transducer and activator of transcription 3 (STAT3) and STAT6, promoting the expression of the anti-inflammatory M2 markers arginase 1 (Arg1) and IL-10 [158]. A recent study has also indicated that resveratrol can promote the M2 microglial phenotype and reduce the degree of neuroinflammation after cerebral ischemia by inhibiting miR-155, a molecule linked to inflammatory processes and to the promotion of M1 polarization [159]. Interestingly, resveratrol-loaded macrophage exosomes, which addressed the low solubility of resveratrol, alleviated inflammation and symptom severity in EAE mice by targeting microglia [160]. These results indicate that resveratrol promotes the M2 microglial phenotype by mitigating inflammation, which is the main impediment of the remyelination process.
Like microglia, astrocytes can exert both detrimental and beneficial effects on remyelination depending on their neurotoxic (A1) or their neuroprotective (A2) phenotype, respectively [161]. Resveratrol treatment inhibited the expression of LPS-induced pro-inflammatory cytokines in both primary murine microglia and astrocytes, while it also reduced the expression of iNOS and the production of nitric oxide (NO) in these glial cell types (Figure 1 and Figure 2) [162]. It is well established that in response to CNS demyelination, astrocytes become activated, proliferate, and form the glial scar, which impedes the remyelination process and is STAT3 dependent [163]. Resveratrol was found to attenuate reactive astrocyte proliferation and activation by downregulating STAT3 signaling in primary rat astrocyte cultures (Figure 2) [164]. These in vitro results suggest that resveratrol could be a promising agent for facilitating remyelination in vivo by regulating the glial scar and establishing a less inflammatory microenvironment.
Reduced inflammation following the administration of resveratrol was further observed in a mouse model of CPZ-induced demyelination. In this context, resveratrol also reduced lipid peroxidation and countered the negative impact of CPZ on the mitochondrial respiratory chain, as assayed by increased cytochrome oxidase activity and ATP levels [165]. Recent studies have established a connection between oxidative stress and the compromised differentiation capacity of OLs, consequently contributing to the process of demyelination [166]. Thus, resveratrol’s effect on the alleviation of oxidative stress could be correlated with the myelin status recovery. Indeed, Ghaiad et al. showed that resveratrol increased myelin basic protein (MBP) expression levels and the stain intensity of Luxol fast blue (LFB), while it also improved balance and motor coordination that were impaired in CPZ-intoxicated mice. These biochemical, histological, and behavioral results indicate that resveratrol reversed CPZ-induced demyelination and enhanced the remyelination process (Figure 3) [165].
Similar effects of resveratrol on balance and motor coordination as well as on enhanced myelin repair in CPZ-treated mice were reported by Samy et al. in 2023. However, the significant improvement in behavioral tests was incomplete compared to control animals, whereas despite the increased number of myelinated axons in the cc, not all the repaired myelin was compacted, and resveratrol failed to upregulate MBP expression levels. These conflicting effects of resveratrol were attributed to different disease-induction and treatment protocols [167]. Interestingly, Samy et al. correlated the positive effects of resveratrol with the induction of autophagy, which is the main result of CR and was interrupted at a late stage in CPZ-treated mice. The induction of the autophagic flux and the successful autophagic degradation upon resveratrol administration involved the activation of the SIRT1/forkhead box protein O1 (FOXO1) pathway [167]. However, the cell-autonomous effect of resveratrol regarding the induction of autophagy and its beneficial effects on remyelination are yet to be determined.
Previous studies have highlighted the advantageous impact of resveratrol on myelination within the PNS. Using an in vitro system comprising a dorsal root ganglion (DRG)/SC co-culture, researchers discerned that resveratrol enhanced myelination, an effect that was mediated, at least in part, by SIRT1 activation in SCs, which serve as the myelinating cells of the PNS [168]. Furthermore, resveratrol induced autophagy in SCs, leading to myelin sheath degeneration in the early stages of nerve injury and, thus, promoting recovery from sciatic nerve crush injury [169]. It is important to acknowledge that myelin clearance represents a critical phase in the regeneration process following peripheral nerve injury [170].
In the context of the CNS, resveratrol mediates protective effects on OLs by preventing LPS-mediated cytotoxicity and reducing the abundance of ROS [171], while it also promotes the survival, migration, proliferation, and differentiation of OPCs in a rat model of ischemic cerebral injury (Figure 3) [172]. Furthermore, resveratrol has demonstrated neuroprotective effects on a chronic EAE mouse model because its administration attenuated the neuronal loss of retinal ganglion cells (RGCs) [173]. Correspondingly, recent research has corroborated resveratrol’s neuroprotective effects, which were attributed to its capacity to promote autophagic activity in a mouse spinal cord injury model [174]. Although the existing evidence pertaining to the roles of resveratrol in myelination and regeneration in the CNS is limited, it is plausible that these processes are regulated, similarly to PNS, by the induction of the autophagic pathway in OLs, which are the myelinating cells of the CNS.
Recent research has emphasized the significance of oligodendroglial autophagy in OL maturation and the maintenance of CNS myelin [21,22]. The activation of the autophagy inducer SIRT1 has been shown to mediate the proliferation and regeneration of OPCs in the white matter of neonatal mice under hypoxic conditions [175]. During adulthood, on the other hand, the genetic ablation of SIRT1 increased the pool of OPCs after focal demyelination, promoting the remyelination process and, thus, indicating the temporally restricted role of SIRT1 in glial regeneration following brain injury [176]. Furthermore, recent findings have suggested that SIRT1 is upregulated in OPCs in EAE and likely plays a role in remyelination [177]. Given that resveratrol is predominantly associated with SIRT1 activation, these pieces of evidence underscore the importance for investigating the cell-autonomous effects of resveratrol on OLs under demyelinating conditions and its potent role as a therapeutic agent for MS.

5. Conclusions

CRMs have gained significant attention within the scientific community, emerging as promising agents capable of emulating numerous effects that are typically induced by CR. Notably, many CRM candidates can induce autophagy, prolong lifespan and/or healthspan, and mitigate the symptoms of age-related diseases, all without the subjective discomfort associated with CR. In addition, CRMs have been shown to exert beneficial effects on demyelinating neuroinflammatory diseases, like MS, by modulating the profile of glial cells, ultimately facilitating the remyelination process. In particular, apart from targeting the migration, proliferation, and differentiation of OLs, CRMs affect microglia and astrocytes by promoting their protective phenotypes, thereby establishing a less inflammatory microenvironment that supports remyelination.
This translational research on CRMs has now progressed to the clinical phase because there is an unmet need to verify their favorable effects through clinical trials. Presently, there are ongoing clinical trials investigating the effects of metformin on endogenous neural progenitor cells in children or young adults with MS (NCT04121468) [178], as well as the safety of metformin for the treatment of progressive MS (NCT05349474) [179]. There is also a clinical trial (NCT05740722), currently recruiting patients, that aims to evaluate the safety and efficacy of NR in the treatment of patients with progressive MS [180]. Despite the large number of in vitro and in vivo studies using animal models of MS, clinical evidence for the protective role of polyphenols in MS patients is restricted, encompassing only a few compounds, like curcumin [181]. However, taking into consideration the beneficial effects of resveratrol on mouse models of MS, as well as its established safety and its ability to modulate neuroinflammation in patients with AD, clinical trials need to be conducted to evaluate the potential of resveratrol to mitigate the symptoms of patients with MS. It is important, though, that aspects such as bioavailability, cellular uptake, systemic distribution, and organ-specific effects are settled. A recent study has used resveratrol nanoparticles to address the poor water solubility and bioavailability of resveratrol in an EAE mouse model. The results have suggested that the nanoparticles increased the bioavailability of the resveratrol and exerted neuroprotective effects by reducing the loss of retinal ganglion cells [182]. Moreover, it is possible that the combination of mechanistically different CRMs will have synergistic effects, thereby maximizing their positive impact.
Additional research also needs to be undertaken to elucidate the influences of distinct CRMs on the myelin sheaths of the elderly. As the human brain ages, the capacity of OPCs to differentiate in mature myelinating OLs significantly declines [183,184]. Notably, the transition from relapsing–remitting to progressive MS takes place at around the same age in MS patients, indicating that it is mostly age rather that disease-duration dependent [185]. Apart from aging, both the disease duration and anatomic sites of lesions affect the remyelination potential of MS patients. It is suggested that remyelination is a more frequent event at the beginning of the disease than in the chronic phase, when remyelination is scarce and predominantly confined to the peripheries of lesions [186]. Regarding the location, cortical lesions are more extensively remyelinated than white-matter ones [187]. Thus, it becomes evident that even though remyelination is highly efficient in animal models and is commonly observed in MS patients, it varies considerably between lesions and between individuals, and these facts should be taken into account during the design of clinical trials. Finally, neuropathological studies reveal that some lesions of MS patients lack OPCs [188], whereas other lesions are characterized by a great number of OPCs with an impaired differentiation capacity [189], indicating that proliferating or differentiating agents should be used according to the lesion status. Consequently, even though spontaneous remyelination exists in humans following a demyelination insult, many obstacles need to be overcome not only for the development of efficient MS treatments based on CRMs but also for the proper evaluation of remyelination. However, despite the imperative for validating CRMs as therapeutic approaches, the existing body of evidence corroborates the considerable potential of these autophagy inducers against MS and multiple age-related diseases.

Author Contributions

D.K. (Despoina Kaffe), writing—original draft preparation and visualization; S.I.K., writing—review and editing; D.K. (Domna Karagogeos), conceptualization, supervision and editing; D.K. (Despoina Kaffe), S.I.K. and D.K. (Domna Karagogeos), funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Hellenic Foundation for Research and Innovation (HFRI) under grant agreement No. 1676, the Hellenic Academy of Neuroimmunology (HELANI), and the Fondation Sante (The Sidney Altman Scholarship Program). De.K was supported by an Onassis Foundation Scholarship (G ZS 004-1/2022-2023).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank all the members of the Karagogeos laboratory for helpful and thought-provoking conversations.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Williams, K.A.; Deber, C.M. The structure and function of central nervous system myelin. Crit. Rev. Clin. Lab. Sci. 1993, 30, 29–64. [Google Scholar] [CrossRef] [PubMed]
  2. Stadelmann, C.; Timmler, S.; Barrantes-Freer, A.; Simons, M. Myelin in the Central Nervous System: Structure, Function, and Pathology. Physiol. Rev. 2019, 99, 1381–1431. [Google Scholar] [CrossRef] [PubMed]
  3. Poliak, S.; Peles, E. The local differentiation of myelinated axons at nodes of Ranvier. Nat. Rev. Neurosci. 2003, 4, 968–980. [Google Scholar] [CrossRef] [PubMed]
  4. Chen, J.F.; Wang, F.; Huang, N.X.; Xiao, L.; Mei, F. Oligodendrocytes and myelin: Active players in neurodegenerative brains? Dev. Neurobiol. 2022, 82, 160–174. [Google Scholar] [CrossRef] [PubMed]
  5. Neely, S.A.; Williamson, J.M.; Klingseisen, A.; Zoupi, L.; Early, J.J.; Williams, A.; Lyons, D.A. New oligodendrocytes exhibit more abundant and accurate myelin regeneration than those that survive demyelination. Nat. Neurosci. 2022, 25, 415–420. [Google Scholar] [CrossRef] [PubMed]
  6. Baaklini, C.S.; Rawji, K.S.; Duncan, G.J.; Ho, M.F.S.; Plemel, J.R. Central Nervous System Remyelination: Roles of Glia and Innate Immune Cells. Front. Mol. Neurosci. 2019, 12, 225. [Google Scholar] [CrossRef]
  7. Djannatian, M.; Radha, S.; Weikert, U.; Safaiyan, S.; Wrede, C.; Deichsel, C.; Kislinger, G.; Rhomberg, A.; Ruhwedel, T.; Campbell, D.S.; et al. Myelination generates aberrant ultrastructure that is resolved by microglia. J. Cell Biol. 2023, 222, e202204010. [Google Scholar] [CrossRef]
  8. Nave, K.A.; Werner, H.B. Myelination of the nervous system: Mechanisms and functions. Annu. Rev. Cell Dev. Biol. 2014, 30, 503–533. [Google Scholar] [CrossRef]
  9. Yamasaki, R.; Lu, H.; Butovsky, O.; Ohno, N.; Rietsch, A.M.; Cialic, R.; Wu, P.M.; Doykan, C.E.; Lin, J.; Cotleur, A.C.; et al. Differential roles of microglia and monocytes in the inflamed central nervous system. J. Exp. Med. 2014, 211, 1533–1549. [Google Scholar] [CrossRef]
  10. Lloyd, A.F.; Miron, V.E. The pro-remyelination properties of microglia in the central nervous system. Nat. Rev. Neurol. 2019, 15, 447–458. [Google Scholar] [CrossRef]
  11. Miron, V.E. Microglia-driven regulation of oligodendrocyte lineage cells, myelination, and remyelination. J. Leukoc. Biol. 2017, 101, 1103–1108. [Google Scholar] [CrossRef] [PubMed]
  12. Lampron, A.; Larochelle, A.; Laflamme, N.; Préfontaine, P.; Plante, M.M.; Sánchez, M.G.; Yong, V.W.; Stys, P.K.; Tremblay, M.; Rivest, S. Inefficient clearance of myelin debris by microglia impairs remyelinating processes. J. Exp. Med. 2015, 212, 481–495. [Google Scholar] [CrossRef] [PubMed]
  13. Ghorbani, S.; Yong, V.W. The extracellular matrix as modifier of neuroinflammation and remyelination in multiple sclerosis. Brain A J. Neurol. 2021, 144, 1958–1973. [Google Scholar] [CrossRef] [PubMed]
  14. Kalafatakis, I.; Karagogeos, D. Oligodendrocytes and Microglia: Key Players in Myelin Development, Damage and Repair. Biomolecules 2021, 11, 1058. [Google Scholar] [CrossRef]
  15. Zamanian, J.L.; Xu, L.; Foo, L.C.; Nouri, N.; Zhou, L.; Giffard, R.G.; Barres, B.A. Genomic analysis of reactive astrogliosis. J. Neurosci. Off. J. Soc. Neurosci. 2012, 32, 6391–6410. [Google Scholar] [CrossRef]
  16. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef]
  17. Liddelow, S.A.; Barres, B.A. Reactive Astrocytes: Production, Function, and Therapeutic Potential. Immunity 2017, 46, 957–967. [Google Scholar] [CrossRef]
  18. Skripuletz, T.; Hackstette, D.; Bauer, K.; Gudi, V.; Pul, R.; Voss, E.; Berger, K.; Kipp, M.; Baumgärtner, W.; Stangel, M. Astrocytes regulate myelin clearance through recruitment of microglia during cuprizone-induced demyelination. Brain A J. Neurol. 2013, 136, 147–167. [Google Scholar] [CrossRef]
  19. Pu, A.; Stephenson, E.L.; Yong, V.W. The extracellular matrix: Focus on oligodendrocyte biology and targeting CSPGs for remyelination therapies. Glia 2018, 66, 1809–1825. [Google Scholar] [CrossRef]
  20. Bankston, A.N.; Forston, M.D.; Howard, R.M.; Andres, K.R.; Smith, A.E.; Ohri, S.S.; Bates, M.L.; Bunge, M.B.; Whittemore, S.R. Autophagy is essential for oligodendrocyte differentiation, survival, and proper myelination. Glia 2019, 67, 1745–1759. [Google Scholar] [CrossRef]
  21. Aber, E.R.; Griffey, C.J.; Davies, T.; Li, A.M.; Yang, Y.J.; Croce, K.R.; Goldman, J.E.; Grutzendler, J.; Canman, J.C.; Yamamoto, A. Oligodendroglial macroautophagy is essential for myelin sheath turnover to prevent neurodegeneration and death. Cell Rep. 2022, 41, 111480. [Google Scholar] [CrossRef]
  22. Ktena, N.; Kaplanis, S.I.; Kolotuev, I.; Georgilis, A.; Kallergi, E.; Stavroulaki, V.; Nikoletopoulou, V.; Savvaki, M.; Karagogeos, D. Autophagic degradation of CNS myelin maintains axon integrity. Cell Stress 2022, 6, 93–107. [Google Scholar] [CrossRef]
  23. Piccio, L.; Stark, J.L.; Cross, A.H. Chronic calorie restriction attenuates experimental autoimmune encephalomyelitis. J. Leukoc. Biol. 2008, 84, 940–948. [Google Scholar] [CrossRef]
  24. Mojaverrostami, S.; Pasbakhsh, P.; Madadi, S.; Nekoonam, S.; Zarini, D.; Noori, L.; Shiri, E.; Salama, M.; Zibara, K.; Kashani, I.R. Calorie restriction promotes remyelination in a Cuprizone-Induced demyelination mouse model of multiple sclerosis. Metab. Brain Dis. 2020, 35, 1211–1224. [Google Scholar] [CrossRef]
  25. Masoro, E.J. Overview of caloric restriction and ageing. Mech. Ageing Dev. 2005, 126, 913–922. [Google Scholar] [CrossRef]
  26. Fontana, L.; Partridge, L.; Longo, V.D. Extending healthy life span—From yeast to humans. Science 2010, 328, 321–326. [Google Scholar] [CrossRef]
  27. Mariño, G.; Pietrocola, F.; Eisenberg, T.; Kong, Y.; Malik, S.A.; Andryushkova, A.; Schroeder, S.; Pendl, T.; Harger, A.; Niso-Santano, M.; et al. Regulation of autophagy by cytosolic acetyl-coenzyme A. Mol. Cell 2014, 53, 710–725. [Google Scholar] [CrossRef]
  28. Menzies, F.M.; Fleming, A.; Caricasole, A.; Bento, C.F.; Andrews, S.P.; Ashkenazi, A.; Füllgrabe, J.; Jackson, A.; Jimenez Sanchez, M.; Karabiyik, C.; et al. Autophagy and Neurodegeneration: Pathogenic Mechanisms and Therapeutic Opportunities. Neuron 2017, 93, 1015–1034. [Google Scholar] [CrossRef] [PubMed]
  29. Yang, S.; Moon, S.; Hur, S.C.; Jeong, S.M. Fatty acid oxidation regulates cellular senescence by modulating the autophagy-SIRT1 axis. BMB Rep. 2023, 5938. [Google Scholar] [CrossRef]
  30. Lv, S.; Zhang, Y.; Lin, Y.; Fang, W.; Wang, Y.; Li, Z.; Lin, A.; Dai, X.; Ye, Q.; Zhang, J.; et al. ApoE4 exacerbates the senescence of hippocampal neurons and spatial cognitive impairment by downregulating acetyl-CoA level. Aging Cell 2023, 22, e13932. [Google Scholar] [CrossRef] [PubMed]
  31. Ren, J.; Zhang, Y. Targeting Autophagy in Aging and Aging-Related Cardiovascular Diseases. Trends Pharmacol. Sci. 2018, 39, 1064–1076. [Google Scholar] [CrossRef]
  32. Bosi, E. Metformin--the gold standard in type 2 diabetes: What does the evidence tell us? Diabetes Obes. Metab. 2009, 11 (Suppl. S2), 3–8. [Google Scholar] [CrossRef]
  33. Madeo, F.; Carmona-Gutierrez, D.; Hofer, S.J.; Kroemer, G. Caloric Restriction Mimetics against Age-Associated Disease: Targets, Mechanisms, and Therapeutic Potential. Cell Metab. 2019, 29, 592–610. [Google Scholar] [CrossRef]
  34. LaMoia, T.E.; Shulman, G.I. Cellular and Molecular Mechanisms of Metformin Action. Endocr. Rev. 2021, 42, 77–96. [Google Scholar] [CrossRef]
  35. Wang, J.C.; Li, G.Y.; Wang, B.; Han, S.X.; Sun, X.; Jiang, Y.N.; Shen, Y.W.; Zhou, C.; Feng, J.; Lu, S.Y.; et al. Metformin inhibits metastatic breast cancer progression and improves chemosensitivity by inducing vessel normalization via PDGF-B downregulation. J. Exp. Clin. Cancer Res. 2019, 38, 235. [Google Scholar] [CrossRef]
  36. Sugiura, K.; Okabayashi, K.; Seishima, R.; Ishida, T.; Shigeta, K.; Tsuruta, M.; Hasegawa, H.; Kitagawa, Y. Metformin inhibits the development and metastasis of colorectal cancer. Med. Oncol. 2022, 39, 136. [Google Scholar] [CrossRef]
  37. Zhang, J.; Huang, L.; Shi, X.; Yang, L.; Hua, F.; Ma, J.; Zhu, W.; Liu, X.; Xuan, R.; Shen, Y.; et al. Metformin protects against myocardial ischemia-reperfusion injury and cell pyroptosis via AMPK/NLRP3 inflammasome pathway. Aging 2020, 12, 24270–24287. [Google Scholar] [CrossRef]
  38. Yerevanian, A.; Soukas, A.A. Metformin: Mechanisms in Human Obesity and Weight Loss. Curr. Obes. Rep. 2019, 8, 156–164. [Google Scholar] [CrossRef] [PubMed]
  39. Du, M.R.; Gao, Q.Y.; Liu, C.L.; Bai, L.Y.; Li, T.; Wei, F.L. Exploring the Pharmacological Potential of Metformin for Neurodegenerative Diseases. Front. Aging Neurosci. 2022, 14, 838173. [Google Scholar] [CrossRef] [PubMed]
  40. Chen, S.; Gan, D.; Lin, S.; Zhong, Y.; Chen, M.; Zou, X.; Shao, Z.; Xiao, G. Metformin in aging and aging-related diseases: Clinical applications and relevant mechanisms. Theranostics 2022, 12, 2722–2740. [Google Scholar] [CrossRef] [PubMed]
  41. Lv, Z.; Guo, Y. Metformin and Its Benefits for Various Diseases. Front. Endocrinol. 2020, 11, 191. [Google Scholar] [CrossRef] [PubMed]
  42. Owen, M.R.; Doran, E.; Halestrap, A.P. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem. J. 2000, 348 Pt 3, 607–614. [Google Scholar] [CrossRef] [PubMed]
  43. Zhou, G.; Myers, R.; Li, Y.; Chen, Y.; Shen, X.; Fenyk-Melody, J.; Wu, M.; Ventre, J.; Doebber, T.; Fujii, N.; et al. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Investig. 2001, 108, 1167–1174. [Google Scholar] [CrossRef]
  44. Duca, F.A.; Côté, C.D.; Rasmussen, B.A.; Zadeh-Tahmasebi, M.; Rutter, G.A.; Filippi, B.M.; Lam, T.K. Metformin activates a duodenal Ampk-dependent pathway to lower hepatic glucose production in rats. Nat. Med. 2015, 21, 506–511. [Google Scholar] [CrossRef] [PubMed]
  45. Lim, J.Y.; Oh, M.A.; Kim, W.H.; Sohn, H.Y.; Park, S.I. AMP-activated protein kinase inhibits TGF-β-induced fibrogenic responses of hepatic stellate cells by targeting transcriptional coactivator p300. J. Cell. Physiol. 2012, 227, 1081–1089. [Google Scholar] [CrossRef] [PubMed]
  46. Caton, P.W.; Nayuni, N.K.; Kieswich, J.; Khan, N.Q.; Yaqoob, M.M.; Corder, R. Metformin suppresses hepatic gluconeogenesis through induction of SIRT1 and GCN5. J. Endocrinol. 2010, 205, 97–106. [Google Scholar] [CrossRef]
  47. Zhang, C.S.; Li, M.; Ma, T.; Zong, Y.; Cui, J.; Feng, J.W.; Wu, Y.Q.; Lin, S.Y.; Lin, S.C. Metformin Activates AMPK through the Lysosomal Pathway. Cell Metab. 2016, 24, 521–522. [Google Scholar] [CrossRef]
  48. Li, Y.; Chen, Y. AMPK and Autophagy. Adv. Exp. Med. Biol. 2019, 1206, 85–108. [Google Scholar] [CrossRef]
  49. Wang, Y.; Zhao, J.; Guo, F.L.; Gao, X.; Xie, X.; Liu, S.; Yang, X.; Yang, X.; Zhang, L.; Ye, Y.; et al. Metformin Ameliorates Synaptic Defects in a Mouse Model of AD by Inhibiting Cdk5 Activity. Front. Cell. Neurosci. 2020, 14, 170. [Google Scholar] [CrossRef]
  50. Chen, Y.; Zhao, S.; Fan, Z.; Li, Z.; Zhu, Y.; Shen, T.; Li, K.; Yan, Y.; Tian, J.; Liu, Z.; et al. Metformin attenuates plaque-associated tau pathology and reduces amyloid-β burden in APP/PS1 mice. Alzheimers Res. Ther. 2021, 13, 40. [Google Scholar] [CrossRef]
  51. Saewanee, N.; Praputpittaya, T.; Malaiwong, N.; Chalorak, P.; Meemon, K. Neuroprotective effect of metformin on dopaminergic neurodegeneration and α-synuclein aggregation in C. elegans model of Parkinson’s disease. Neurosci. Res. 2019, 162, 13–21. [Google Scholar] [CrossRef]
  52. Sanchis, A.; García-Gimeno, M.A.; Cañada-Martínez, A.J.; Sequedo, M.D.; Millán, J.M.; Sanz, P.; Vázquez-Manrique, R.P. Metformin treatment reduces motor and neuropsychiatric phenotypes in the zQ175 mouse model of Huntington disease. Exp. Mol. Med. 2019, 51, 1–16. [Google Scholar] [CrossRef]
  53. Samaras, K.; Makkar, S.; Crawford, J.D.; Kochan, N.A.; Wen, W.; Draper, B.; Trollor, J.N.; Brodaty, H.; Sachdev, P.S. Metformin Use Is Associated With Slowed Cognitive Decline and Reduced Incident Dementia in Older Adults With Type 2 Diabetes: The Sydney Memory and Ageing Study. Diabetes Care 2020, 43, 2691–2701. [Google Scholar] [CrossRef] [PubMed]
  54. Koenig, A.M.; Mechanic-Hamilton, D.; Xie, S.X.; Combs, M.F.; Cappola, A.R.; Xie, L.; Detre, J.A.; Wolk, D.A.; Arnold, S.E. Effects of the Insulin Sensitizer Metformin in Alzheimer Disease: Pilot Data From a Randomized Placebo-controlled Crossover Study. Alzheimer Dis. Assoc. Disord. 2017, 31, 107–113. [Google Scholar] [CrossRef]
  55. Moore, E.M.; Mander, A.G.; Ames, D.; Kotowicz, M.A.; Carne, R.P.; Brodaty, H.; Woodward, M.; Boundy, K.; Ellis, K.A.; Bush, A.I.; et al. Increased risk of cognitive impairment in patients with diabetes is associated with metformin. Diabetes Care 2013, 36, 2981–2987. [Google Scholar] [CrossRef]
  56. Dziedzic, A.; Saluk-Bijak, J.; Miller, E.; Bijak, M. Metformin as a Potential Agent in the Treatment of Multiple Sclerosis. Int. J. Mol. Sci. 2020, 21, 5957. [Google Scholar] [CrossRef]
  57. Thompson, A.J.; Baranzini, S.E.; Geurts, J.; Hemmer, B.; Ciccarelli, O. Multiple sclerosis. Lancet 2018, 391, 1622–1636. [Google Scholar] [CrossRef]
  58. Largani, S.H.H.; Borhani-Haghighi, M.; Pasbakhsh, P.; Mahabadi, V.P.; Nekoonam, S.; Shiri, E.; Kashani, I.R.; Zendehdel, A. Oligoprotective effect of metformin through the AMPK-dependent on restoration of mitochondrial hemostasis in the cuprizone-induced multiple sclerosis model. J. Mol. Histol. 2019, 50, 263–271. [Google Scholar] [CrossRef]
  59. Abdi, M.; Pasbakhsh, P.; Shabani, M.; Nekoonam, S.; Sadeghi, A.; Fathi, F.; Abouzaripour, M.; Mohamed, W.; Zibara, K.; Kashani, I.R.; et al. Metformin Therapy Attenuates Pro-inflammatory Microglia by Inhibiting NF-κB in Cuprizone Demyelinating Mouse Model of Multiple Sclerosis. Neurotox. Res. 2021, 39, 1732–1746. [Google Scholar] [CrossRef] [PubMed]
  60. Ishihara, Y.; Itoh, K. Microglial inflammatory reactions regulated by oxidative stress. J. Clin. Biochem. Nutr. 2023, 72, 23–27. [Google Scholar] [CrossRef] [PubMed]
  61. Nahirnyj, A.; Livne-Bar, I.; Guo, X.; Sivak, J.M. ROS detoxification and proinflammatory cytokines are linked by p38 MAPK signaling in a model of mature astrocyte activation. PLoS ONE 2013, 8, e83049. [Google Scholar] [CrossRef] [PubMed]
  62. Nath, N.; Khan, M.; Paintlia, M.K.; Singh, I.; Hoda, M.N.; Giri, S. Metformin attenuated the autoimmune disease of the central nervous system in animal models of multiple sclerosis. J. Immunol. 2009, 182, 8005–8014. [Google Scholar] [CrossRef] [PubMed]
  63. Wu, Y.Q.; Xiong, J.; He, Z.L.; Yuan, Y.; Wang, B.N.; Xu, J.Y.; Wu, M.; Zhang, S.S.; Cai, S.F.; Zhao, J.X.; et al. Metformin promotes microglial cells to facilitate myelin debris clearance and accelerate nerve repairment after spinal cord injury. Acta Pharmacol. Sin. 2022, 43, 1360–1371. [Google Scholar] [CrossRef] [PubMed]
  64. Sanadgol, N.; Barati, M.; Houshmand, F.; Hassani, S.; Clarner, T.; Shahlaei, M.; Golab, F. Metformin accelerates myelin recovery and ameliorates behavioral deficits in the animal model of multiple sclerosis via adjustment of AMPK/Nrf2/mTOR signaling and maintenance of endogenous oligodendrogenesis during brain self-repairing period. Pharmacol. Rep. 2020, 72, 641–658. [Google Scholar] [CrossRef] [PubMed]
  65. Bercury, K.K.; Dai, J.; Sachs, H.H.; Ahrendsen, J.T.; Wood, T.L.; Macklin, W.B. Conditional ablation of raptor or rictor has differential impact on oligodendrocyte differentiation and CNS myelination. J. Neurosci. 2014, 34, 4466–4480. [Google Scholar] [CrossRef] [PubMed]
  66. Ishii, A.; Furusho, M.; Macklin, W.; Bansal, R. Independent and cooperative roles of the Mek/ERK1/2-MAPK and PI3K/Akt/mTOR pathways during developmental myelination and in adulthood. Glia 2019, 67, 1277–1295. [Google Scholar] [CrossRef]
  67. Carson, R.P.; Van Nielen, D.L.; Winzenburger, P.A.; Ess, K.C. Neuronal and glia abnormalities in Tsc1-deficient forebrain and partial rescue by rapamycin. Neurobiol. Dis. 2012, 45, 369–380. [Google Scholar] [CrossRef] [PubMed]
  68. McLane, L.E.; Bourne, J.N.; Evangelou, A.V.; Khandker, L.; Macklin, W.B.; Wood, T.L. Loss of Tuberous Sclerosis Complex1 in Adult Oligodendrocyte Progenitor Cells Enhances Axon Remyelination and Increases Myelin Thickness after a Focal Demyelination. J. Neurosci. 2017, 37, 7534–7546. [Google Scholar] [CrossRef]
  69. Manwani, B.; McCullough, L.D. Function of the master energy regulator adenosine monophosphate-activated protein kinase in stroke. J. Neurosci. Res. 2013, 91, 1018–1029. [Google Scholar] [CrossRef]
  70. Narine, M.; Azmi, M.A.; Umali, M.; Volz, A.; Colognato, H. The AMPK activator metformin improves recovery from demyelination by shifting oligodendrocyte bioenergetics and accelerating OPC differentiation. Front. Cell. Neurosci. 2023, 17, 1254303. [Google Scholar] [CrossRef]
  71. Paintlia, A.S.; Paintlia, M.K.; Mohan, S.; Singh, A.K.; Singh, I. AMP-activated protein kinase signaling protects oligodendrocytes that restore central nervous system functions in an experimental autoimmune encephalomyelitis model. Am. J. Pathol. 2013, 183, 526–541. [Google Scholar] [CrossRef]
  72. Houshmand, F.; Barati, M.; Golab, F.; Ramezani-Sefidar, S.; Tanbakooie, S.; Tabatabaei, M.; Amiri, M.; Sanadgol, N. Metformin-induced AMPK activation stimulates remyelination through induction of neurotrophic factors, downregulation of NogoA and recruitment of Olig2+ precursor cells in the cuprizone murine model of multiple sclerosis. Daru 2019, 27, 583–592. [Google Scholar] [CrossRef]
  73. Pöyhönen, S.; Er, S.; Domanskyi, A.; Airavaara, M. Effects of Neurotrophic Factors in Glial Cells in the Central Nervous System: Expression and Properties in Neurodegeneration and Injury. Front. Physiol. 2019, 10, 486. [Google Scholar] [CrossRef]
  74. Kosaraju, J.; Seegobin, M.; Gouveia, A.; Syal, C.; Sarma, S.N.; Lu, K.J.; Ilin, J.; He, L.; Wondisford, F.E.; Lagace, D.; et al. Metformin promotes CNS remyelination and improves social interaction following focal demyelination through CBP Ser436 phosphorylation. Exp. Neurol. 2020, 334, 113454. [Google Scholar] [CrossRef]
  75. Franklin, R.J.; Ffrench-Constant, C.; Edgar, J.M.; Smith, K.J. Neuroprotection and repair in multiple sclerosis. Nat. Rev. Neurol. 2012, 8, 624–634. [Google Scholar] [CrossRef]
  76. Sim, F.J.; Zhao, C.; Penderis, J.; Franklin, R.J. The age-related decrease in CNS remyelination efficiency is attributable to an impairment of both oligodendrocyte progenitor recruitment and differentiation. J. Neurosci. 2002, 22, 2451–2459. [Google Scholar] [CrossRef]
  77. Neumann, B.; Baror, R.; Zhao, C.; Segel, M.; Dietmann, S.; Rawji, K.S.; Foerster, S.; McClain, C.R.; Chalut, K.; van Wijngaarden, P.; et al. Metformin Restores CNS Remyelination Capacity by Rejuvenating Aged Stem Cells. Cell Stem Cell 2019, 25, 473–485.e8. [Google Scholar] [CrossRef]
  78. Najafi, M.; Cheki, M.; Rezapoor, S.; Geraily, G.; Motevaseli, E.; Carnovale, C.; Clementi, E.; Shirazi, A. Metformin: Prevention of genomic instability and cancer: A review. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2018, 827, 1–8. [Google Scholar] [CrossRef]
  79. Bharath, L.P.; Agrawal, M.; McCambridge, G.; Nicholas, D.A.; Hasturk, H.; Liu, J.; Jiang, K.; Liu, R.; Guo, Z.; Deeney, J.; et al. Metformin Enhances Autophagy and Normalizes Mitochondrial Function to Alleviate Aging-Associated Inflammation. Cell Metab. 2020, 32, 44–55.e6. [Google Scholar] [CrossRef]
  80. Bogan, K.L.; Brenner, C. Nicotinic acid, nicotinamide, and nicotinamide riboside: A molecular evaluation of NAD+ precursor vitamins in human nutrition. Annu. Rev. Nutr. 2008, 28, 115–130. [Google Scholar] [CrossRef]
  81. Hofer, S.J.; Davinelli, S.; Bergmann, M.; Scapagnini, G.; Madeo, F. Caloric Restriction Mimetics in Nutrition and Clinical Trials. Front. Nutr. 2021, 8, 717343. [Google Scholar] [CrossRef]
  82. Verdin, E. NAD+ in aging, metabolism, and neurodegeneration. Science 2015, 350, 1208–1213. [Google Scholar] [CrossRef]
  83. Yoshino, J.; Baur, J.A.; Imai, S.I. NAD+ Intermediates: The Biology and Therapeutic Potential of NMN and NR. Cell Metab. 2018, 27, 513–528. [Google Scholar] [CrossRef]
  84. Fang, E.F.; Kassahun, H.; Croteau, D.L.; Scheibye-Knudsen, M.; Marosi, K.; Lu, H.; Shamanna, R.A.; Kalyanasundaram, S.; Bollineni, R.C.; Wilson, M.A.; et al. NAD+ Replenishment Improves Lifespan and Healthspan in Ataxia Telangiectasia Models via Mitophagy and DNA Repair. Cell Metab. 2016, 24, 566–581. [Google Scholar] [CrossRef]
  85. Zhang, H.; Ryu, D.; Wu, Y.; Gariani, K.; Wang, X.; Luan, P.; D’Amico, D.; Ropelle, E.R.; Lutolf, M.P.; Aebersold, R.; et al. NAD+ repletion improves mitochondrial and stem cell function and enhances life span in mice. Science 2016, 352, 1436–1443. [Google Scholar] [CrossRef]
  86. Mitchell, S.J.; Bernier, M.; Aon, M.A.; Cortassa, S.; Kim, E.Y.; Fang, E.F.; Palacios, H.H.; Ali, A.; Navas-Enamorado, I.; Di Francesco, A.; et al. Nicotinamide Improves Aspects of Healthspan, but Not Lifespan, in Mice. Cell Metab. 2018, 27, 667–676.e4. [Google Scholar] [CrossRef]
  87. Abdellatif, M.; Trummer-Herbst, V.; Koser, F.; Durand, S.; Adão, R.; Vasques-Nóvoa, F.; Freundt, J.K.; Voglhuber, J.; Pricolo, M.R.; Kasa, M.; et al. Nicotinamide for the treatment of heart failure with preserved ejection fraction. Sci. Transl. Med. 2021, 13, eabd7064. [Google Scholar] [CrossRef]
  88. Wu, K.; Li, B.; Lin, Q.; Xu, W.; Zuo, W.; Li, J.; Liu, N.; Tu, T.; Zhang, B.; Xiao, Y.; et al. Nicotinamide mononucleotide attenuates isoproterenol-induced cardiac fibrosis by regulating oxidative stress and Smad3 acetylation. Life Sci. 2021, 274, 119299. [Google Scholar] [CrossRef]
  89. Scatozza, F.; Moschella, F.; D’Arcangelo, D.; Rossi, S.; Tabolacci, C.; Giampietri, C.; Proietti, E.; Facchiano, F.; Facchiano, A. Nicotinamide inhibits melanoma in vitro and in vivo. J. Exp. Clin. Cancer Res. 2020, 39, 211. [Google Scholar] [CrossRef]
  90. Jung, M.; Lee, K.M.; Im, Y.; Seok, S.H.; Chung, H.; Kim, D.Y.; Han, D.; Lee, C.H.; Hwang, E.H.; Park, S.Y.; et al. Nicotinamide (niacin) supplement increases lipid metabolism and ROS-induced energy disruption in triple-negative breast cancer: Potential for drug repositioning as an anti-tumor agent. Mol. Oncol. 2022, 16, 1795–1815. [Google Scholar] [CrossRef]
  91. Lautrup, S.; Sinclair, D.A.; Mattson, M.P.; Fang, E.F. NAD+ in Brain Aging and Neurodegenerative Disorders. Cell Metab. 2019, 30, 630–655. [Google Scholar] [CrossRef]
  92. Fang, E.F.; Lautrup, S.; Hou, Y.; Demarest, T.G.; Croteau, D.L.; Mattson, M.P.; Bohr, V.A. NAD+ in Aging: Molecular Mechanisms and Translational Implications. Trends Mol. Med. 2017, 23, 899–916. [Google Scholar] [CrossRef] [PubMed]
  93. Katsyuba, E.; Romani, M.; Hofer, D.; Auwerx, J. NAD+ homeostasis in health and disease. Nat. Metab. 2020, 2, 9–31. [Google Scholar] [CrossRef] [PubMed]
  94. Barbosa, M.T.; Soares, S.M.; Novak, C.M.; Sinclair, D.; Levine, J.A.; Aksoy, P.; Chini, E.N. The enzyme CD38 (a NAD glycohydrolase, EC 3.2.2.5) is necessary for the development of diet-induced obesity. FASEB J. 2007, 21, 3629–3639. [Google Scholar] [CrossRef] [PubMed]
  95. Langley, M.R.; Choi, C.I.; Peclat, T.R.; Guo, Y.; Simon, W.L.; Yoon, H.; Kleppe, L.; Lucchinetti, C.F.; Chini, C.C.S.; Chini, E.N.; et al. Critical Role of Astrocyte NAD+ Glycohydrolase in Myelin Injury and Regeneration. J. Neurosci. 2021, 41, 8644–8667. [Google Scholar] [CrossRef] [PubMed]
  96. Roboon, J.; Hattori, T.; Ishii, H.; Takarada-Iemata, M.; Le, T.M.; Shiraishi, Y.; Ozaki, N.; Yamamoto, Y.; Sugawara, A.; Okamoto, H.; et al. Deletion of CD38 Suppresses Glial Activation and Neuroinflammation in a Mouse Model of Demyelination. Front. Cell. Neurosci. 2019, 13, 258. [Google Scholar] [CrossRef] [PubMed]
  97. Rouleau, M.; Patel, A.; Hendzel, M.J.; Kaufmann, S.H.; Poirier, G.G. PARP inhibition: PARP1 and beyond. Nat. Rev. Cancer 2010, 10, 293–301. [Google Scholar] [CrossRef]
  98. Veto, S.; Acs, P.; Bauer, J.; Lassmann, H.; Berente, Z.; Setalo, G., Jr.; Borgulya, G.; Sumegi, B.; Komoly, S.; Gallyas, F., Jr.; et al. Inhibiting poly(ADP-ribose) polymerase: A potential therapy against oligodendrocyte death. Brain A J. Neurol. 2010, 133, 822–834. [Google Scholar] [CrossRef]
  99. Lee, I.H.; Cao, L.; Mostoslavsky, R.; Lombard, D.B.; Liu, J.; Bruns, N.E.; Tsokos, M.; Alt, F.W.; Finkel, T. A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Proc. Natl. Acad. Sci. USA 2008, 105, 3374–3379. [Google Scholar] [CrossRef]
  100. Kitada, M.; Ogura, Y.; Koya, D. Chapter 3—Role of Sirt1 as a Regulator of Autophagy. In Autophagy: Cancer, Other Pathologies, Inflammation, Immunity, Infection, and Aging; Hayat, M.A., Ed.; Academic Press: San Diego, CA, USA, 2016; pp. 89–100. [Google Scholar] [CrossRef]
  101. Li, Y.; Yang, G.; Yang, X.; He, Y.; Wang, W.; Zhang, J.; Li, T.; Zhang, W.; Lin, R. Nicotinic acid inhibits vascular inflammation via the SIRT1-dependent signaling pathway. J. Nutr. Biochem. 2015, 26, 1338–1347. [Google Scholar] [CrossRef]
  102. Li, Y.; Yang, G.; Yang, X.; Wang, W.; Zhang, J.; He, Y.; Zhang, W.; Jing, T.; Lin, R. Nicotinic acid inhibits NLRP3 inflammasome activation via SIRT1 in vascular endothelial cells. Int. Immunopharmacol. 2016, 40, 211–218. [Google Scholar] [CrossRef] [PubMed]
  103. Zhang, J.; Wang, C.; Ying, W. SIRT2 and Akt mediate NAD+-induced and NADH-induced increases in the intracellular ATP levels of BV2 microglia under basal conditions. Neuroreport 2018, 29, 65–70. [Google Scholar] [CrossRef] [PubMed]
  104. Acklin, S.; Sadhukhan, R.; Du, W.; Patra, M.; Cholia, R.; Xia, F. Nicotinamide riboside alleviates cisplatin-induced peripheral neuropathy via SIRT2 activation. Neurooncol. Adv. 2022, 4, vdac101. [Google Scholar] [CrossRef] [PubMed]
  105. Hou, Y.; Wei, Y.; Lautrup, S.; Yang, B.; Wang, Y.; Cordonnier, S.; Mattson, M.P.; Croteau, D.L.; Bohr, V.A. NAD(+) supplementation reduces neuroinflammation and cell senescence in a transgenic mouse model of Alzheimer’s disease via cGAS-STING. Proc. Natl. Acad. Sci. USA 2021, 118, e2011226118. [Google Scholar] [CrossRef] [PubMed]
  106. Zhao, X.; Kong, M.; Wang, Y.; Mao, Y.; Xu, H.; He, W.; He, Y.; Gu, J. Nicotinamide mononucleotide improves the Alzheimer’s disease by regulating intestinal microbiota. Biochem. Biophys. Res. Commun. 2023, 670, 27–35. [Google Scholar] [CrossRef] [PubMed]
  107. Rehman, I.U.; Khan, A.; Ahmad, R.; Choe, K.; Park, H.Y.; Lee, H.J.; Atiq, A.; Park, J.; Hahm, J.R.; Kim, M.O. Neuroprotective Effects of Nicotinamide against MPTP-Induced Parkinson’s Disease in Mice: Impact on Oxidative Stress, Neuroinflammation, Nrf2/HO-1 and TLR4 Signaling Pathways. Biomedicines 2022, 10, 2929. [Google Scholar] [CrossRef] [PubMed]
  108. Sidhu, A.; Diwan, V.; Kaur, H.; Bhateja, D.; Singh, C.K.; Sharma, S.; Padi, S.S.V. Nicotinamide reverses behavioral impairments and provides neuroprotection in 3-nitropropionic acid induced animal model ofHuntington’s disease: Implication of oxidative stress- poly(ADP- ribose) polymerase pathway. Metab. Brain Dis. 2018, 33, 1911–1921. [Google Scholar] [CrossRef]
  109. Trammell, S.A.; Schmidt, M.S.; Weidemann, B.J.; Redpath, P.; Jaksch, F.; Dellinger, R.W.; Li, Z.; Abel, E.D.; Migaud, M.E.; Brenner, C. Nicotinamide riboside is uniquely and orally bioavailable in mice and humans. Nat. Commun. 2016, 7, 12948. [Google Scholar] [CrossRef]
  110. Martens, C.R.; Denman, B.A.; Mazzo, M.R.; Armstrong, M.L.; Reisdorph, N.; McQueen, M.B.; Chonchol, M.; Seals, D.R. Chronic nicotinamide riboside supplementation is well-tolerated and elevates NAD+ in healthy middle-aged and older adults. Nat. Commun. 2018, 9, 1286. [Google Scholar] [CrossRef]
  111. Vreones, M.; Mustapic, M.; Moaddel, R.; Pucha, K.A.; Lovett, J.; Seals, D.R.; Kapogiannis, D.; Martens, C.R. Oral nicotinamide riboside raises NAD+ and lowers biomarkers of neurodegenerative pathology in plasma extracellular vesicles enriched for neuronal origin. Aging Cell 2023, 22, e13754. [Google Scholar] [CrossRef]
  112. Brakedal, B.; Dölle, C.; Riemer, F.; Ma, Y.; Nido, G.S.; Skeie, G.O.; Craven, A.R.; Schwarzlmüller, T.; Brekke, N.; Diab, J.; et al. The NADPARK study: A randomized phase I trial of nicotinamide riboside supplementation in Parkinson’s disease. Cell Metab. 2022, 34, 396–407.e6. [Google Scholar] [CrossRef] [PubMed]
  113. Nicotinamide as an Early Alzheimer’s Disease Treatment. Available online: https://clinicaltrials.gov/study/NCT03061474 (accessed on 8 November 2023).
  114. Wang, X.; Li, B.; Liu, L.; Zhang, L.; Ma, T.; Guo, L. Nicotinamide adenine dinucleotide treatment alleviates the symptoms of experimental autoimmune encephalomyelitis by activating autophagy and inhibiting the NLRP3 inflammasome. Int. Immunopharmacol. 2021, 90, 107092. [Google Scholar] [CrossRef] [PubMed]
  115. Guo, J.; Li, B.; Wang, J.; Guo, R.; Tian, Y.; Song, S.; Guo, L. Protective effect and mechanism of nicotinamide adenine dinucleotide against optic neuritis in mice with experimental autoimmune encephalomyelitis. Int. Immunopharmacol. 2021, 98, 107846. [Google Scholar] [CrossRef] [PubMed]
  116. Franklin, R.J.; Ffrench-Constant, C. Remyelination in the CNS: From biology to therapy. Nat. Rev. Neurosci. 2008, 9, 839–855. [Google Scholar] [CrossRef] [PubMed]
  117. Kaplanis, S.I.; Kaffe, D.; Ktena, N.; Lygeraki, A.; Kolliniati, O.; Savvaki, M.; Karagogeos, D. Nicotinamide enhances myelin production after demyelination through reduction of astrogliosis and microgliosis. Front. Cell. Neurosci. 2023, 17, 1201317. [Google Scholar] [CrossRef] [PubMed]
  118. Roboon, J.; Hattori, T.; Ishii, H.; Takarada-Iemata, M.; Nguyen, D.T.; Heer, C.D.; O’Meally, D.; Brenner, C.; Yamamoto, Y.; Okamoto, H.; et al. Inhibition of CD38 and supplementation of nicotinamide riboside ameliorate lipopolysaccharide-induced microglial and astrocytic neuroinflammation by increasing NAD+. J. Neurochem. 2021, 158, 311–327. [Google Scholar] [CrossRef] [PubMed]
  119. Hong, Y.; Liu, Y.; Zhang, G.; Wu, H.; Hou, Y. Progesterone suppresses Aβ(42)-induced neuroinflammation by enhancing autophagy in astrocytes. Int. Immunopharmacol. 2018, 54, 336–343. [Google Scholar] [CrossRef]
  120. Pais, T.F.; Szegő, É.M.; Marques, O.; Miller-Fleming, L.; Antas, P.; Guerreiro, P.; de Oliveira, R.M.; Kasapoglu, B.; Outeiro, T.F. The NAD-dependent deacetylase sirtuin 2 is a suppressor of microglial activation and brain inflammation. EMBO J. 2013, 32, 2603–2616. [Google Scholar] [CrossRef]
  121. Li, W.; Zhang, B.; Tang, J.; Cao, Q.; Wu, Y.; Wu, C.; Guo, J.; Ling, E.A.; Liang, F. Sirtuin 2, a mammalian homolog of yeast silent information regulator-2 longevity regulator, is an oligodendroglial protein that decelerates cell differentiation through deacetylating alpha-tubulin. J. Neurosci. 2007, 27, 2606–2616. [Google Scholar] [CrossRef]
  122. Jayasena, T.; Poljak, A.; Braidy, N.; Zhong, L.; Rowlands, B.; Muenchhoff, J.; Grant, R.; Smythe, G.; Teo, C.; Raftery, M.; et al. Application of Targeted Mass Spectrometry for the Quantification of Sirtuins in the Central Nervous System. Sci. Rep. 2016, 6, 35391. [Google Scholar] [CrossRef]
  123. Ma, X.R.; Zhu, X.; Xiao, Y.; Gu, H.M.; Zheng, S.S.; Li, L.; Wang, F.; Dong, Z.J.; Wang, D.X.; Wu, Y.; et al. Restoring nuclear entry of Sirtuin 2 in oligodendrocyte progenitor cells promotes remyelination during ageing. Nat. Commun. 2022, 13, 1225. [Google Scholar] [CrossRef] [PubMed]
  124. Rawji, K.S.; Young, A.M.H.; Ghosh, T.; Michaels, N.J.; Mirzaei, R.; Kappen, J.; Kolehmainen, K.L.; Alaeiilkhchi, N.; Lozinski, B.; Mishra, M.K.; et al. Niacin-mediated rejuvenation of macrophage/microglia enhances remyelination of the aging central nervous system. Acta Neuropathol. 2020, 139, 893–909. [Google Scholar] [CrossRef] [PubMed]
  125. Zhao, C.; Li, W.W.; Franklin, R.J. Differences in the early inflammatory responses to toxin-induced demyelination are associated with the age-related decline in CNS remyelination. Neurobiol. Aging 2006, 27, 1298–1307. [Google Scholar] [CrossRef] [PubMed]
  126. Natrajan, M.S.; de la Fuente, A.G.; Crawford, A.H.; Linehan, E.; Nuñez, V.; Johnson, K.R.; Wu, T.; Fitzgerald, D.C.; Ricote, M.; Bielekova, B.; et al. Retinoid X receptor activation reverses age-related deficiencies in myelin debris phagocytosis and remyelination. Brain A J. Neurol. 2015, 138, 3581–3597. [Google Scholar] [CrossRef] [PubMed]
  127. Wang, C.; Zhang, Y.; Ding, J.; Zhao, Z.; Qian, C.; Luan, Y.; Teng, G.J. Nicotinamide Administration Improves Remyelination after Stroke. Neural Plast. 2017, 2017, 7019803. [Google Scholar] [CrossRef] [PubMed]
  128. Xiao, J.; Wong, A.W.; Willingham, M.M.; van den Buuse, M.; Kilpatrick, T.J.; Murray, S.S. Brain-derived neurotrophic factor promotes central nervous system myelination via a direct effect upon oligodendrocytes. Neurosignals 2010, 18, 186–202. [Google Scholar] [CrossRef]
  129. Pirola, L.; Fröjdö, S. Resveratrol: One molecule, many targets. IUBMB Life 2008, 60, 323–332. [Google Scholar] [CrossRef]
  130. Meng, X.; Zhou, J.; Zhao, C.N.; Gan, R.Y.; Li, H.B. Health Benefits and Molecular Mechanisms of Resveratrol: A Narrative Review. Foods 2020, 9, 340. [Google Scholar] [CrossRef]
  131. Jang, M.; Cai, L.; Udeani, G.O.; Slowing, K.V.; Thomas, C.F.; Beecher, C.W.; Fong, H.H.; Farnsworth, N.R.; Kinghorn, A.D.; Mehta, R.G.; et al. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science 1997, 275, 218–220. [Google Scholar] [CrossRef]
  132. Li, T.; Tan, Y.; Ouyang, S.; He, J.; Liu, L. Resveratrol protects against myocardial ischemia-reperfusion injury via attenuating ferroptosis. Gene 2022, 808, 145968. [Google Scholar] [CrossRef]
  133. Mahjabeen, W.; Khan, D.A.; Mirza, S.A. Role of resveratrol supplementation in regulation of glucose hemostasis, inflammation and oxidative stress in patients with diabetes mellitus type 2: A randomized, placebo-controlled trial. Complement. Ther. Med. 2022, 66, 102819. [Google Scholar] [CrossRef] [PubMed]
  134. Sun, Y.; Zhou, Q.M.; Lu, Y.Y.; Zhang, H.; Chen, Q.L.; Zhao, M.; Su, S.B. Resveratrol Inhibits the Migration and Metastasis of MDA-MB-231 Human Breast Cancer by Reversing TGF-β1-Induced Epithelial-Mesenchymal Transition. Molecules 2019, 24, 1131. [Google Scholar] [CrossRef] [PubMed]
  135. Wang, P.; Gao, J.; Ke, W.; Wang, J.; Li, D.; Liu, R.; Jia, Y.; Wang, X.; Chen, X.; Chen, F.; et al. Resveratrol reduces obesity in high-fat diet-fed mice via modulating the composition and metabolic function of the gut microbiota. Free Radic. Biol. Med. 2020, 156, 83–98. [Google Scholar] [CrossRef] [PubMed]
  136. Yan, Y.; Yang, H.; Xie, Y.; Ding, Y.; Kong, D.; Yu, H. Research Progress on Alzheimer’s Disease and Resveratrol. Neurochem. Res. 2020, 45, 989–1006. [Google Scholar] [CrossRef] [PubMed]
  137. Dos Santos, M.G.; Schimith, L.E.; André-Miral, C.; Muccillo-Baisch, A.L.; Arbo, B.D.; Hort, M.A. Neuroprotective Effects of Resveratrol in In vivo and In vitro Experimental Models of Parkinson’s Disease: A Systematic Review. Neurotox. Res. 2022, 40, 319–345. [Google Scholar] [CrossRef] [PubMed]
  138. Ginés, C.; Cuesta, S.; Kireev, R.; García, C.; Rancan, L.; Paredes, S.D.; Vara, E.; Tresguerres, J.A.F. Protective effect of resveratrol against inflammation, oxidative stress and apoptosis in pancreas of aged SAMP8 mice. Exp. Gerontol. 2017, 90, 61–70. [Google Scholar] [CrossRef] [PubMed]
  139. Wang, N.; Luo, Z.; Jin, M.; Sheng, W.; Wang, H.T.; Long, X.; Wu, Y.; Hu, P.; Xu, H.; Zhang, X. Exploration of age-related mitochondrial dysfunction and the anti-aging effects of resveratrol in zebrafish retina. Aging 2019, 11, 3117–3137. [Google Scholar] [CrossRef]
  140. Morselli, E.; Maiuri, M.C.; Markaki, M.; Megalou, E.; Pasparaki, A.; Palikaras, K.; Criollo, A.; Galluzzi, L.; Malik, S.A.; Vitale, I.; et al. Caloric restriction and resveratrol promote longevity through the Sirtuin-1-dependent induction of autophagy. Cell Death Dis. 2010, 1, e10. [Google Scholar] [CrossRef]
  141. Price, N.L.; Gomes, A.P.; Ling, A.J.; Duarte, F.V.; Martin-Montalvo, A.; North, B.J.; Agarwal, B.; Ye, L.; Ramadori, G.; Teodoro, J.S.; et al. SIRT1 is required for AMPK activation and the beneficial effects of resveratrol on mitochondrial function. Cell Metab. 2012, 15, 675–690. [Google Scholar] [CrossRef]
  142. Huang, Y.; Lu, J.; Zhan, L.; Wang, M.; Shi, R.; Yuan, X.; Gao, X.; Liu, X.; Zang, J.; Liu, W.; et al. Resveratrol-induced Sirt1 phosphorylation by LKB1 mediates mitochondrial metabolism. J. Biol. Chem. 2021, 297, 100929. [Google Scholar] [CrossRef]
  143. Um, J.H.; Park, S.J.; Kang, H.; Yang, S.; Foretz, M.; McBurney, M.W.; Kim, M.K.; Viollet, B.; Chung, J.H. AMP-activated protein kinase-deficient mice are resistant to the metabolic effects of resveratrol. Diabetes 2010, 59, 554–563. [Google Scholar] [CrossRef] [PubMed]
  144. Chiang, M.C.; Nicol, C.J.; Cheng, Y.C. Resveratrol activation of AMPK-dependent pathways is neuroprotective in human neural stem cells against amyloid-beta-induced inflammation and oxidative stress. Neurochem. Int. 2018, 115, 1–10. [Google Scholar] [CrossRef] [PubMed]
  145. Lin, C.H.; Nicol, C.J.B.; Cheng, Y.C.; Yen, C.; Wang, Y.S.; Chiang, M.C. Neuroprotective effects of resveratrol against oxygen glucose deprivation induced mitochondrial dysfunction by activation of AMPK in SH-SY5Y cells with 3D gelatin scaffold. Brain Res. 2020, 1726, 146492. [Google Scholar] [CrossRef] [PubMed]
  146. Cantó, C.; Gerhart-Hines, Z.; Feige, J.N.; Lagouge, M.; Noriega, L.; Milne, J.C.; Elliott, P.J.; Puigserver, P.; Auwerx, J. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature 2009, 458, 1056–1060. [Google Scholar] [CrossRef] [PubMed]
  147. Meng, T.; Xiao, D.; Muhammed, A.; Deng, J.; Chen, L.; He, J. Anti-Inflammatory Action and Mechanisms of Resveratrol. Molecules 2021, 26, 229. [Google Scholar] [CrossRef]
  148. Broderick, T.L.; Rasool, S.; Li, R.; Zhang, Y.; Anderson, M.; Al-Nakkash, L.; Plochocki, J.H.; Geetha, T.; Babu, J.R. Neuroprotective Effects of Chronic Resveratrol Treatment and Exercise Training in the 3xTg-AD Mouse Model of Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 7337. [Google Scholar] [CrossRef] [PubMed]
  149. Sun, X.Y.; Dong, Q.X.; Zhu, J.; Sun, X.; Zhang, L.F.; Qiu, M.; Yu, X.L.; Liu, R.T. Resveratrol Rescues Tau-Induced Cognitive Deficits and Neuropathology in a Mouse Model of Tauopathy. Curr. Alzheimer Res. 2019, 16, 710–722. [Google Scholar] [CrossRef] [PubMed]
  150. Zhang, L.F.; Yu, X.L.; Ji, M.; Liu, S.Y.; Wu, X.L.; Wang, Y.J.; Liu, R.T. Resveratrol alleviates motor and cognitive deficits and neuropathology in the A53T α-synuclein mouse model of Parkinson’s disease. Food Funct. 2018, 9, 6414–6426. [Google Scholar] [CrossRef]
  151. Turner, R.S.; Thomas, R.G.; Craft, S.; van Dyck, C.H.; Mintzer, J.; Reynolds, B.A.; Brewer, J.B.; Rissman, R.A.; Raman, R.; Aisen, P.S. A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology 2015, 85, 1383–1391. [Google Scholar] [CrossRef]
  152. Moussa, C.; Hebron, M.; Huang, X.; Ahn, J.; Rissman, R.A.; Aisen, P.S.; Turner, R.S. Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer’s disease. J. Neuroinflamm. 2017, 14, 1. [Google Scholar] [CrossRef]
  153. Wang, J.; Wang, J.; Wang, J.; Yang, B.; Weng, Q.; He, Q. Targeting Microglia and Macrophages: A Potential Treatment Strategy for Multiple Sclerosis. Front. Pharmacol. 2019, 10, 286. [Google Scholar] [CrossRef] [PubMed]
  154. Jha, M.K.; Lee, W.H.; Suk, K. Functional polarization of neuroglia: Implications in neuroinflammation and neurological disorders. Biochem. Pharmacol. 2016, 103, 1–16. [Google Scholar] [CrossRef] [PubMed]
  155. Hammond, T.R.; Dufort, C.; Dissing-Olesen, L.; Giera, S.; Young, A.; Wysoker, A.; Walker, A.J.; Gergits, F.; Segel, M.; Nemesh, J.; et al. Single-Cell RNA Sequencing of Microglia throughout the Mouse Lifespan and in the Injured Brain Reveals Complex Cell-State Changes. Immunity 2019, 50, 253–271.e6. [Google Scholar] [CrossRef] [PubMed]
  156. Plemel, J.R.; Stratton, J.A.; Michaels, N.J.; Rawji, K.S.; Zhang, E.; Sinha, S.; Baaklini, C.S.; Dong, Y.; Ho, M.; Thorburn, K.; et al. Microglia response following acute demyelination is heterogeneous and limits infiltrating macrophage dispersion. Sci. Adv. 2020, 6, eaay6324. [Google Scholar] [CrossRef]
  157. Lombardi, M.; Parolisi, R.; Scaroni, F.; Bonfanti, E.; Gualerzi, A.; Gabrielli, M.; Kerlero de Rosbo, N.; Uccelli, A.; Giussani, P.; Viani, P.; et al. Detrimental and protective action of microglial extracellular vesicles on myelin lesions: Astrocyte involvement in remyelination failure. Acta Neuropathol. 2019, 138, 987–1012. [Google Scholar] [CrossRef] [PubMed]
  158. Yang, X.; Xu, S.; Qian, Y.; Xiao, Q. Resveratrol regulates microglia M1/M2 polarization via PGC-1α in conditions of neuroinflammatory injury. Brain Behav. Immun. 2017, 64, 162–172. [Google Scholar] [CrossRef]
  159. Ma, S.; Fan, L.; Li, J.; Zhang, B.; Yan, Z. Resveratrol promoted the M2 polarization of microglia and reduced neuroinflammation after cerebral ischemia by inhibiting miR-155. Int. J. Neurosci. 2020, 130, 817–825. [Google Scholar] [CrossRef]
  160. Zheng, X.; Sun, K.; Liu, Y.; Yin, X.; Zhu, H.; Yu, F.; Zhao, W. Resveratrol-loaded macrophage exosomes alleviate multiple sclerosis through targeting microglia. J. Control. Release 2023, 353, 675–684. [Google Scholar] [CrossRef]
  161. Quan, L.; Uyeda, A.; Muramatsu, R. Central nervous system regeneration: The roles of glial cells in the potential molecular mechanism underlying remyelination. Inflamm. Regen. 2022, 42, 7. [Google Scholar] [CrossRef]
  162. Lu, X.; Ma, L.; Ruan, L.; Kong, Y.; Mou, H.; Zhang, Z.; Wang, Z.; Wang, J.M.; Le, Y. Resveratrol differentially modulates inflammatory responses of microglia and astrocytes. J. Neuroinflamm. 2010, 7, 46. [Google Scholar] [CrossRef]
  163. Fan, Y.Y.; Huo, J. A1/A2 astrocytes in central nervous system injuries and diseases: Angels or devils? Neurochem. Int. 2021, 148, 105080. [Google Scholar] [CrossRef] [PubMed]
  164. Wu, M.; Wang, L.; Li, F.; Hu, R.; Ma, J.; Zhang, K.; Cheng, X. Resveratrol Downregulates STAT3 Expression and Astrocyte Activation in Primary Astrocyte Cultures of Rat. Neurochem. Res. 2020, 45, 455–464. [Google Scholar] [CrossRef] [PubMed]
  165. Ghaiad, H.R.; Nooh, M.M.; El-Sawalhi, M.M.; Shaheen, A.A. Resveratrol Promotes Remyelination in Cuprizone Model of Multiple Sclerosis: Biochemical and Histological Study. Mol. Neurobiol. 2017, 54, 3219–3229. [Google Scholar] [CrossRef] [PubMed]
  166. Spaas, J.; van Veggel, L.; Schepers, M.; Tiane, A.; van Horssen, J.; Wilson, D.M., 3rd; Moya, P.R.; Piccart, E.; Hellings, N.; Eijnde, B.O.; et al. Oxidative stress and impaired oligodendrocyte precursor cell differentiation in neurological disorders. Cell. Mol. Life Sci. 2021, 78, 4615–4637. [Google Scholar] [CrossRef] [PubMed]
  167. Samy, D.M.; Zaki, E.I.; Hassaan, P.S.; Abdelmonsif, D.A.; Mohamed, D.Y.; Saleh, S.R. Neurobehavioral, biochemical and histological assessment of the effects of resveratrol on cuprizone-induced demyelination in mice: Role of autophagy modulation. J. Physiol. Biochem. 2023, 79, 583–596. [Google Scholar] [CrossRef] [PubMed]
  168. Stettner, M.; Wolffram, K.; Mausberg, A.K.; Albrecht, P.; Derksen, A.; Methner, A.; Dehmel, T.; Hartung, H.P.; Dietrich, H.; Kieseier, B.C. Promoting myelination in an in vitro mouse model of the peripheral nervous system: The effect of wine ingredients. PLoS ONE 2013, 8, e66079. [Google Scholar] [CrossRef]
  169. Zhang, J.; Ren, J.; Liu, Y.; Huang, D.; Lu, L. Resveratrol regulates the recovery of rat sciatic nerve crush injury by promoting the autophagy of Schwann cells. Life Sci. 2020, 256, 117959. [Google Scholar] [CrossRef]
  170. Yuan, Y.; Wang, Y.; Wu, S.; Zhao, M.Y. Review: Myelin clearance is critical for regeneration after peripheral nerve injury. Front. Neurol. 2022, 13, 908148. [Google Scholar] [CrossRef]
  171. Rosa, P.M.; Martins, L.A.M.; Souza, D.O.; Quincozes-Santos, A. Glioprotective Effect of Resveratrol: An Emerging Therapeutic Role for Oligodendroglial Cells. Mol. Neurobiol. 2018, 55, 2967–2978. [Google Scholar] [CrossRef]
  172. Yu, P.; Wang, L.; Tang, F.; Guo, S.; Liao, H.; Fan, C.; Yang, Q. Resveratrol-mediated neurorestoration after cerebral ischemic injury—Sonic Hedgehog signaling pathway. Life Sci. 2021, 280, 119715. [Google Scholar] [CrossRef]
  173. Fonseca-Kelly, Z.; Nassrallah, M.; Uribe, J.; Khan, R.S.; Dine, K.; Dutt, M.; Shindler, K.S. Resveratrol neuroprotection in a chronic mouse model of multiple sclerosis. Front. Neurol. 2012, 3, 84. [Google Scholar] [CrossRef] [PubMed]
  174. Hu, J.; Han, H.; Cao, P.; Yu, W.; Yang, C.; Gao, Y.; Yuan, W. Resveratrol improves neuron protection and functional recovery through enhancement of autophagy after spinal cord injury in mice. Am. J. Transl. Res. 2017, 9, 4607–4616. [Google Scholar] [PubMed]
  175. Jablonska, B.; Gierdalski, M.; Chew, L.J.; Hawley, T.; Catron, M.; Lichauco, A.; Cabrera-Luque, J.; Yuen, T.; Rowitch, D.; Gallo, V. Sirt1 regulates glial progenitor proliferation and regeneration in white matter after neonatal brain injury. Nat. Commun. 2016, 7, 13866. [Google Scholar] [CrossRef] [PubMed]
  176. Rafalski, V.A.; Ho, P.P.; Brett, J.O.; Ucar, D.; Dugas, J.C.; Pollina, E.A.; Chow, L.M.; Ibrahim, A.; Baker, S.J.; Barres, B.A.; et al. Expansion of oligodendrocyte progenitor cells following SIRT1 inactivation in the adult brain. Nat. Cell Biol. 2013, 15, 614–624. [Google Scholar] [CrossRef] [PubMed]
  177. Prozorovski, T.; Ingwersen, J.; Lukas, D.; Göttle, P.; Koop, B.; Graf, J.; Schneider, R.; Franke, K.; Schumacher, S.; Britsch, S.; et al. Regulation of sirtuin expression in autoimmune neuroinflammation: Induction of SIRT1 in oligodendrocyte progenitor cells. Neurosci. Lett. 2019, 704, 116–125. [Google Scholar] [CrossRef] [PubMed]
  178. A Phase I Double Blind Study of Metformin Acting on Endogenous Neural Progenitor Cells in Children with Multiple Sclerosis. Available online: https://clinicaltrials.gov/study/NCT04121468 (accessed on 8 November 2023).
  179. Metformin Treatment in Progressive Multiple Sclerosis. Available online: https://www.clinicaltrials.gov/study/NCT05349474 (accessed on 8 November 2023).
  180. Nicotinamide Riboside Supplementation in Progressive Multiple Sclerosis. Available online: https://clinicaltrials.gov/study/NCT05740722 (accessed on 8 November 2023).
  181. La Rosa, G.; Lonardo, M.S.; Cacciapuoti, N.; Muscariello, E.; Guida, B.; Faraonio, R.; Santillo, M.; Damiano, S. Dietary Polyphenols, Microbiome, and Multiple Sclerosis: From Molecular Anti-Inflammatory and Neuroprotective Mechanisms to Clinical Evidence. Int. J. Mol. Sci. 2023, 24, 7247. [Google Scholar] [CrossRef]
  182. Shamsher, E.; Khan, R.S.; Davis, B.M.; Dine, K.; Luong, V.; Somavarapu, S.; Cordeiro, M.F.; Shindler, K.S. Nanoparticles Enhance Solubility and Neuroprotective Effects of Resveratrol in Demyelinating Disease. Neurotherapeutics 2023, 20, 1138–1153. [Google Scholar] [CrossRef]
  183. Ruckh, J.M.; Zhao, J.W.; Shadrach, J.L.; van Wijngaarden, P.; Rao, T.N.; Wagers, A.J.; Franklin, R.J. Rejuvenation of regeneration in the aging central nervous system. Cell Stem Cell 2012, 10, 96–103. [Google Scholar] [CrossRef]
  184. Shen, S.; Sandoval, J.; Swiss, V.A.; Li, J.; Dupree, J.; Franklin, R.J.; Casaccia-Bonnefil, P. Age-dependent epigenetic control of differentiation inhibitors is critical for remyelination efficiency. Nat. Neurosci. 2008, 11, 1024–1034. [Google Scholar] [CrossRef]
  185. Tutuncu, M.; Tang, J.; Zeid, N.A.; Kale, N.; Crusan, D.J.; Atkinson, E.J.; Siva, A.; Pittock, S.J.; Pirko, I.; Keegan, B.M.; et al. Onset of progressive phase is an age-dependent clinical milestone in multiple sclerosis. Mult. Scler. 2013, 19, 188–198. [Google Scholar] [CrossRef]
  186. Goldschmidt, T.; Antel, J.; König, F.B.; Brück, W.; Kuhlmann, T. Remyelination capacity of the MS brain decreases with disease chronicity. Neurology 2009, 72, 1914–1921. [Google Scholar] [CrossRef] [PubMed]
  187. Strijbis, E.M.M.; Kooi, E.J.; van der Valk, P.; Geurts, J.J.G. Cortical Remyelination Is Heterogeneous in Multiple Sclerosis. J. Neuropathol. Exp. Neurol. 2017, 76, 390–401. [Google Scholar] [CrossRef] [PubMed]
  188. Boyd, A.; Zhang, H.; Williams, A. Insufficient OPC migration into demyelinated lesions is a cause of poor remyelination in MS and mouse models. Acta Neuropathol. 2013, 125, 841–859. [Google Scholar] [CrossRef] [PubMed]
  189. Kuhlmann, T.; Miron, V.; Cui, Q.; Wegner, C.; Antel, J.; Brück, W. Differentiation block of oligodendroglial progenitor cells as a cause for remyelination failure in chronic multiple sclerosis. Brain A J. Neurol. 2008, 131, 1749–1758. [Google Scholar] [CrossRef]
Figure 1. The effects of different caloric restriction mimetics (CRMs) on microglia. Metformin treatment reduces oxidative stress, upregulates antioxidant enzymes, and downregulates NF-κB signaling in microglia, thus attenuating the production of pro-inflammatory cytokines, ultimately leading to reduced microgliosis. Moreover, a similar reduction in reactive microgliosis is observed following treatment with NR, NAD+, and NAM. NAD+ administration results in a decreased expression of pro-inflammatory cytokines and an increased expression of anti-inflammatory ones. In parallel, NAM facilitates the polarization of microglia toward their anti-inflammatory phenotype, an effect that is also evident in response to metformin and resveratrol treatment, which additionally reduce iNOS and NO levels. The cumulative impact of these cellular responses contributes to the establishment of a less inflammatory milieu that could support the remyelination process. iNOS: inducible nitric oxide synthase; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; NR: nicotinamide riboside; NAD+: nicotinamide adenine dinucleotide; NAM: nicotinamide; NO: nitric oxide. Created with BioRender.com (accessed on 21 November 2023).
Figure 1. The effects of different caloric restriction mimetics (CRMs) on microglia. Metformin treatment reduces oxidative stress, upregulates antioxidant enzymes, and downregulates NF-κB signaling in microglia, thus attenuating the production of pro-inflammatory cytokines, ultimately leading to reduced microgliosis. Moreover, a similar reduction in reactive microgliosis is observed following treatment with NR, NAD+, and NAM. NAD+ administration results in a decreased expression of pro-inflammatory cytokines and an increased expression of anti-inflammatory ones. In parallel, NAM facilitates the polarization of microglia toward their anti-inflammatory phenotype, an effect that is also evident in response to metformin and resveratrol treatment, which additionally reduce iNOS and NO levels. The cumulative impact of these cellular responses contributes to the establishment of a less inflammatory milieu that could support the remyelination process. iNOS: inducible nitric oxide synthase; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; NR: nicotinamide riboside; NAD+: nicotinamide adenine dinucleotide; NAM: nicotinamide; NO: nitric oxide. Created with BioRender.com (accessed on 21 November 2023).
Cimb 45 00596 g001
Figure 2. The effects of different CRMs on astrocytes. Resveratrol attenuates the expression of pro-inflammatory cytokines and inflammatory markers, such as iNOS and NO, while it also downregulates STAT3 signaling, leading to reduced proliferation and activation of reactive astrocytes. Given that STAT3 is implicated in the formation of glial scars in response to CNS demyelination, it is possible that resveratrol can reduce reactive astrogliosis in vivo. Furthermore, reduced reactive astrogliosis is observed upon NR, NAD+, NAM, and metformin treatments. NAD+ administration results in a decreased expression of pro-inflammatory cytokines and an increased expression of anti-inflammatory ones. In a similar way, NAM facilitates the polarization of astrocytes toward their anti-inflammatory phenotype, partially through the induction of autophagy. Metformin, in turn, acts through the activation of AMPK and induces the production of neurotrophic factors by astrocytes. This protective, less inflammatory microenvironment can support the process of remyelination in vivo after a demyelination insult. STAT3: signal transducer and activator of transcription 3; AMPK: adenosine 5′-monophosphate-activated protein kinase. Created with BioRender.com (accessed on 21 November 2023).
Figure 2. The effects of different CRMs on astrocytes. Resveratrol attenuates the expression of pro-inflammatory cytokines and inflammatory markers, such as iNOS and NO, while it also downregulates STAT3 signaling, leading to reduced proliferation and activation of reactive astrocytes. Given that STAT3 is implicated in the formation of glial scars in response to CNS demyelination, it is possible that resveratrol can reduce reactive astrogliosis in vivo. Furthermore, reduced reactive astrogliosis is observed upon NR, NAD+, NAM, and metformin treatments. NAD+ administration results in a decreased expression of pro-inflammatory cytokines and an increased expression of anti-inflammatory ones. In a similar way, NAM facilitates the polarization of astrocytes toward their anti-inflammatory phenotype, partially through the induction of autophagy. Metformin, in turn, acts through the activation of AMPK and induces the production of neurotrophic factors by astrocytes. This protective, less inflammatory microenvironment can support the process of remyelination in vivo after a demyelination insult. STAT3: signal transducer and activator of transcription 3; AMPK: adenosine 5′-monophosphate-activated protein kinase. Created with BioRender.com (accessed on 21 November 2023).
Cimb 45 00596 g002
Figure 3. The effects of different CRMs on oligodendrocyte lineage cells and the process of remyelination. Metformin administration promotes the recruitment of OPCs to the lesion site and attenuates demyelination through its ability to reduce oxidative stress and upregulate antioxidant enzymes. Furthermore, metformin promotes OPC proliferation via the blockage of autophagy and enhances OPC differentiation and maturation into myelinating OLs via CBP phosphorylation. The remyelination process is also enhanced by the modulation of AMPK/mTORC pathways in mature OLs. Apart from metformin, NAM and resveratrol can promote the remyelination process. NAM treatment promotes the maturation of OPCs via the BDNF/TrkB pathway. Resveratrol protects OLs by reducing the abundance of ROS while it also induces autophagy through the activation of SIRT1, a mechanism that likely supports remyelination. Finally, direct NAD+ supplementation facilitates the recruitment of OPCs and alleviates demyelination through the activation of the SIRT1 signaling pathway and, probably, through the induction of autophagy. OPCs: oligodendrocyte progenitor cells; OLs: oligodendrocytes; CBP: CREB-binding protein; mTORC: mammalian target of rapamycin complex; BDNF: brain-derived neurotrophic factor; TrkB: tropomyosin receptor kinase B; SIRT1: sirtuin 1. Created with BioRender.com (accessed on 21 November 2023).
Figure 3. The effects of different CRMs on oligodendrocyte lineage cells and the process of remyelination. Metformin administration promotes the recruitment of OPCs to the lesion site and attenuates demyelination through its ability to reduce oxidative stress and upregulate antioxidant enzymes. Furthermore, metformin promotes OPC proliferation via the blockage of autophagy and enhances OPC differentiation and maturation into myelinating OLs via CBP phosphorylation. The remyelination process is also enhanced by the modulation of AMPK/mTORC pathways in mature OLs. Apart from metformin, NAM and resveratrol can promote the remyelination process. NAM treatment promotes the maturation of OPCs via the BDNF/TrkB pathway. Resveratrol protects OLs by reducing the abundance of ROS while it also induces autophagy through the activation of SIRT1, a mechanism that likely supports remyelination. Finally, direct NAD+ supplementation facilitates the recruitment of OPCs and alleviates demyelination through the activation of the SIRT1 signaling pathway and, probably, through the induction of autophagy. OPCs: oligodendrocyte progenitor cells; OLs: oligodendrocytes; CBP: CREB-binding protein; mTORC: mammalian target of rapamycin complex; BDNF: brain-derived neurotrophic factor; TrkB: tropomyosin receptor kinase B; SIRT1: sirtuin 1. Created with BioRender.com (accessed on 21 November 2023).
Cimb 45 00596 g003
Figure 4. The effects of different CRMs on the aged central nervous system (CNS) after a demyelination insult. The administration of metformin improves the mitochondrial function of aged OPCs via the activation of AMPK, consequently facilitating the restoration of the CNS myelination, an effect that could be attributed to the induction of autophagy. Furthermore, β-NMN supplementation rescues SIRT2 nuclear localization in the OPCs of aged mice, thus enhancing remyelination by promoting the differentiation of aged OPCs. Finally, NA promotes remyelination by enhancing the phagocyting activity of middle-aged microglia. This process results in the removal of myelin debris from the lesion site and the recruitment and maturation of OPCs into myelinating OLs. β-NMN: β-nicotinamide mononucleotide; SIRT2: sirtuin 2; NA: nicotinic acid. Created with BioRender.com (accessed on 21 November 2023).
Figure 4. The effects of different CRMs on the aged central nervous system (CNS) after a demyelination insult. The administration of metformin improves the mitochondrial function of aged OPCs via the activation of AMPK, consequently facilitating the restoration of the CNS myelination, an effect that could be attributed to the induction of autophagy. Furthermore, β-NMN supplementation rescues SIRT2 nuclear localization in the OPCs of aged mice, thus enhancing remyelination by promoting the differentiation of aged OPCs. Finally, NA promotes remyelination by enhancing the phagocyting activity of middle-aged microglia. This process results in the removal of myelin debris from the lesion site and the recruitment and maturation of OPCs into myelinating OLs. β-NMN: β-nicotinamide mononucleotide; SIRT2: sirtuin 2; NA: nicotinic acid. Created with BioRender.com (accessed on 21 November 2023).
Cimb 45 00596 g004
Figure 5. NAD+ biosynthetic pathways. NA is converted to NAD+ via the Preiss–Handler pathway, whereas NAM and NR enter the salvage pathway to produce NAD+. The NAD+ salvage pathway recycles NAM that is generated as a byproduct of the activity of NAD+-consuming enzymes: CD38, PARP, and sirtuins. NMN is synthesized from NAM by NAMPT as well as from NR by NRKs. PARP: poly (ADP-ribose) polymerase; NMN: nicotinamide mononucleotide; NAMPT: nicotinamide phosphoribosyltransferase; NRKs: nicotinamide riboside kinases. Created with BioRender.com (accessed on 21 November 2023).
Figure 5. NAD+ biosynthetic pathways. NA is converted to NAD+ via the Preiss–Handler pathway, whereas NAM and NR enter the salvage pathway to produce NAD+. The NAD+ salvage pathway recycles NAM that is generated as a byproduct of the activity of NAD+-consuming enzymes: CD38, PARP, and sirtuins. NMN is synthesized from NAM by NAMPT as well as from NR by NRKs. PARP: poly (ADP-ribose) polymerase; NMN: nicotinamide mononucleotide; NAMPT: nicotinamide phosphoribosyltransferase; NRKs: nicotinamide riboside kinases. Created with BioRender.com (accessed on 21 November 2023).
Cimb 45 00596 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kaffe, D.; Kaplanis, S.I.; Karagogeos, D. The Roles of Caloric Restriction Mimetics in Central Nervous System Demyelination and Remyelination. Curr. Issues Mol. Biol. 2023, 45, 9526-9548. https://doi.org/10.3390/cimb45120596

AMA Style

Kaffe D, Kaplanis SI, Karagogeos D. The Roles of Caloric Restriction Mimetics in Central Nervous System Demyelination and Remyelination. Current Issues in Molecular Biology. 2023; 45(12):9526-9548. https://doi.org/10.3390/cimb45120596

Chicago/Turabian Style

Kaffe, Despoina, Stefanos Ioannis Kaplanis, and Domna Karagogeos. 2023. "The Roles of Caloric Restriction Mimetics in Central Nervous System Demyelination and Remyelination" Current Issues in Molecular Biology 45, no. 12: 9526-9548. https://doi.org/10.3390/cimb45120596

Article Metrics

Back to TopTop