Next Article in Journal
Vanillic Acid Suppresses HIF-1α Expression via Inhibition of mTOR/p70S6K/4E-BP1 and Raf/MEK/ERK Pathways in Human Colon Cancer HCT116 Cells
Next Article in Special Issue
Healthy Brain Aging Modifies Microglial Calcium Signaling In Vivo
Previous Article in Journal
Discovery of Food-Derived Dipeptidyl Peptidase IV Inhibitory Peptides: A Review
Previous Article in Special Issue
Hydrogen Indirectly Suppresses Increases in Hydrogen Peroxide in Cytoplasmic Hydroxyl Radical-Induced Cells and Suppresses Cellular Senescence
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dietary Restriction and Neuroinflammation: A Potential Mechanistic Link

Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2019, 20(3), 464; https://doi.org/10.3390/ijms20030464
Submission received: 18 January 2019 / Accepted: 19 January 2019 / Published: 22 January 2019
(This article belongs to the Special Issue Nutrition and Aging)

Abstract

:
Chronic neuroinflammation is a common feature of the aged brain, and its association with the major neurodegenerative changes involved in cognitive impairment and motor dysfunction is well established. One of the most potent antiaging interventions tested so far is dietary restriction (DR), which extends the lifespan in various organisms. Microglia and astrocytes are two major types of glial cells involved in the regulation of neuroinflammation. Accumulating evidence suggests that the age-related proinflammatory activation of astrocytes and microglia is attenuated under DR. However, the molecular mechanisms underlying DR-mediated regulation of neuroinflammation are not well understood. Here, we review the current understanding of the effects of DR on neuroinflammation and suggest an underlying mechanistic link between DR and neuroinflammation that may provide novel insights into the role of DR in aging and age-associated brain disorders.

Graphical Abstract

1. Introduction

Aging is a naturally occurring multifactorial process that leads to morbidity and mortality. In particular, brain aging manifests as a gradual decline in memory and cognitive, executive, and motor functions. It is now widely accepted that brain aging is accompanied by region-dependent morphological and functional alterations. These include anatomical atrophy, reduction in brain volume, synaptic deficits, decline in the capacity for neurogenesis, cytoskeletal abnormalities, mitochondrial dysfunction, an increase in reactive oxygen species (ROS) and oxidized proteins, a reduction in neurotrophic factors, impairment of the blood–brain barrier (BBB), and induction of chronic neuroinflammation (reviewed in [1,2]).
In experimental and clinical settings, dietary restriction (DR) is induced by reducing the caloric intake and/or intermittent fasting. Since McCay and colleagues first reported the effect of DR on lifespan in rats in 1935 [3], DR has been shown to be the most robust and reliable experimental intervention for extending longevity. Accumulating evidence suggests that DR extends lifespan in an evolutionary scale from worms to rodents [4]. Although the effects of DR on longevity and brain function in nonhuman primates and human beings are still controversial [5,6,7,8,9,10,11,12], a recent report from the CALERIE (Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy) trial noted that two-year-long DR in healthy, nonobese human subjects caused weight loss and reduction in energy expenditure and oxidative stress [13]. These findings indicate that the mechanism underlying DR-mediated antiaging may be evolutionally conserved from worms to mammals, possibly including humans.
A growing body of evidence demonstrates that DR exerts its beneficial effects on brain aging at multiple levels. Although there is some degree of discrepancy across studies, likely due to the difference in the model organisms and experimental design, DR appears to mitigate all of the morphological and functional alterations in the brain associated with aging (reviewed in [1,14,15,16,17]).
A major hallmark of aging is systemic, low-grade chronic inflammation throughout the body, termed inflamm-aging, a term coined by Claudio Franceschi in 2000 [18]. Notably, these inflammatory signs are similar to the ones associated with obesity and metabolic diseases [19,20], providing a possible glimpse into why DR exerts anti-inflammatory effects on aging-associated inflammation. As with other organs, chronic low-grade inflammation is a common feature of the aged brain [21]. Neuroinflammation is a host defense mechanism against harmful stimuli and damage in the brain. However, chronic inflammation can be deleterious in normal aging as well as in pathological aging related to neurodegenerative diseases. The central nervous system (CNS) is composed of heterogeneous cell types, including neurons, microglia, astrocytes, and oligodendrocytes. Although two major glial cell types, astrocytes and microglia, are known to be key players in inflammatory responses in the brain, it is now well recognized that all neural cells participate to some degree in the neuroinflammatory responses. Neuroinflammation often manifests as astrogliosis, microgliosis, and an increase in secreted inflammatory mediators, such as cytokines, chemokines, and complement proteins. Accumulating evidence from clinical and basic research suggests that neuroinflammation is tightly connected to the decline in brain function during aging [22]. In this article, we review the evidence that DR has an anti-neuroinflammatory effect and suggest an underlying mechanistic link between DR and neuroinflammation that may provide novel insights into the role of DR in aging and age-associated brain disorders.

2. Neuroinflammation in the Aged Brain

2.1. Evidence of Increased Neuroinflammation with Age

The brain was once considered to be an absolute immune-privileged organ isolated from the systemic immune system by the BBB. In fact, the brain is an immunologically active organ that communicates with the immune and endocrine systems. Moreover, circulating cytokines and chemokines can signal to the CNS, although the underlying mechanisms are still elusive [23,24]. Similar to the changes seen in the systemic immune system with aging, numerous studies have revealed that the inflammatory status of the brain increases during normal aging. The hallmarks of brain aging include impairment of DNA repair, accumulation of oxidative damage, and inflammatory activation of glial cells [21]. Consistent with this, induction of the genes associated with immune and inflammatory responses is observed in most human brain regions during normal aging, although the changes vary across brain regions [25,26,27]. Moreover, proinflammatory cytokines, such as interleukin-1 beta (IL-1β), tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6), are increased in multiple brain regions, including hippocampus and cortex, during normal aging [28,29,30,31,32,33,34]. Furthermore, some immune regulatory molecules and anti-inflammatory cytokines, including interleukin-10 (IL-10) and interleukin-4 (IL-4), were found to be reduced in aged rodent brains [35,36]. In addition, the aged brain is more vulnerable to peripheral systemic inflammatory stimuli [37]. Hypothalamic inflammation impairs the brain-governed energy control and glucose homeostasis, thereby promoting peripheral inflammation as well as neuroinflammation [14].
Another hallmark of neuroinflammation is gliosis. Gliosis is the focal proliferation and activation of glial cells (astrocytes, microglia, and oligodendrocytes) in the CNS in response to various insults. Overwhelming evidence shows that astrogliosis is increased in multiple brain regions during normal aging in both rodents and humans [38,39,40]. Activation of astrocytes is observed relatively early in adult brains during aging, as evidenced by increased expression of glial fibrillary acidic protein (GFAP) [40,41,42,43,44,45,46]. Signs of microgliosis and microglial activation, such as induction of major histocompatibility complex II (MHC II), scavenger receptor (CD86), and CD40, are also prominent in the aged brain [37,41,42,47,48,49,50]. Microglia isolated from the aged mouse brains were found to be more sensitive to inflammatory stimuli than those from the young brains [37].
Taken together, these studies demonstrate that even healthy aged brains have a significant increase in neuroinflammatory responses, as evidenced by increased gliosis, elevated levels of inflammatory cytokines, and decreased levels of anti-inflammatory molecules.

2.2. Microglia in the Aged Brain

Microglia are the innate immune cells in the CNS that play a pivotal role in maintaining microenvironment homeostasis, synaptic plasticity, and immune surveillance [51,52,53]. During embryogenesis, microglial progenitor cells originated from yolk sac-derived myeloid migrate into the CNS before the BBB construction is completed [54,55]. Once the BBB construction is completed, the peripheral hematopoietic system does not further contribute to the microglia pool of the CNS under normal condition, meaning that the adult microglia population is maintained by self-renewal in the brain [56,57]. In addition, microglia are long-lived cells with the median lifetime of 15 months in mouse neocortex, and only about 26% of microglia are replaced per year [58]. Réu and colleagues reported that microglia in the human brain is on average 4.2 years old, and the median renewal rate is 28% per year [59]. A long life span of microglia may make them more susceptible to the aging-related changes compared to other neural cells.
Microglia play a key role in neuroinflammation associated with aging. Microglia in the young adult brain, which are typically in a quiescent resting state, become activated in response to various types of insults. With aging, microglia develop an altered phenotype compatible with proinflammatory activation. The elevated inflammatory profile of microglia during aging is closely associated with a “primed” state. “Primed” microglia have a higher basal expression of inflammatory response genes, a lower threshold for inflammatory activation, and elevated reactivity following an immune challenge [60]. Many inflammatory markers, such as MHC II, complement receptor 3 (CD11b), Toll-like receptors (TLRs), CD86, CD11b, and CD11c, are upregulated in microglia of the aged brain [61,62,63]. Among TLRs, TLR2–4 are known to be important for the ATP-dependent secretion of IL-1β in primed microglia [64]. Besides molecular changes, morphological changes in microglia also occur in the aged brain from rodent to human. Microglia in the aged brain show a “de-ramified” morphology characterized by short processes and reduced dendritic branching, suggesting an activation of microglia with age [65,66,67,68,69].
Godbout and colleagues showed that peripheral injection of lipopolysaccharide (LPS), a powerful inducer of the inflammatory response, caused an exaggerated and prolonged neuroinflammatory response in the aged brain [29]. The elevated inflammatory responses in aged mice were associated with sustained behavioral deficits, such as reduced motor function and altered social behavior [29]. In addition, elevated levels of IL-1β after systemic injection of LPS were mainly derived from MHC II (a marker for primed microglia)-positive microglia in the aged mouse brain [37]. Consistent with this, systemic injection of Escherichia coli resulted in an exaggerated and prolonged upregulation of IL-1β in hippocampus of aged rats compared with young adults [70]. Aged rodents and older individuals showed enhanced neuroinflammation, increased neurodegeneration, and poorer recovery after traumatic brain injury (TBI) than their younger counterparts [71,72,73]. Aged mice experienced more severe neuronal damage upon TBI induction by controlled cortical impact that young mice [72]. Moreover, MHC II was strongly upregulated in microglia of the aged TBI brain [72]. Taken together, these reports indicate that primed microglia play an important role in enhancing neuroinflammatory responses to immune challenges in the aged brain.
The effect of aging on microglia gene expression was recently investigated through transcriptome analysis in microglia isolated from young and aged mouse brains [74]. Consistent with the characteristics of aged microglia, genes associated with the immune, phagosome, lysosome, oxidative phosphorylation, and antigen presentation signaling pathways were significantly affected by aging [74]. It is noteworthy that the transcriptional profile of aged microglia was clearly different from that of M1 macrophage, M2 macrophages, or acutely activated microglia [74]. A list of differentially expressed genes (DEG) between young and aged microglia associated with the immune, inflammatory responses, and antigen presentation signaling pathways is summarized in Table 1 [74].

2.3. Astrocytes in the Aged Brain

Astrocytes are the most abundant cell type in the mammalian brain. Astrocytes are essential for neuroprotection against excitotoxicity, ROS, insults, and extracellular overload of potassium ions [75]. They also have functions associated with synaptic plasticity and trophic support for neurons [75]. Similar to microglia, astrocytes display an elevated inflammatory profile with age, including morphological and molecular alterations. For example, astrocytes in young human subjects were found to have long and slender processes, whereas astrocytes in aged brains possessed short and stubby processes [76]. In addition, upregulation of GFAP and vimentin has been reported in astrocytes of aged brains [60]. Notably, increased expression of GFAP and vimentin is a typical signature of reactive astrocytes [77,78]. Thus, these findings indicate that astrocytes become reactive with age.
Upon immune challenge to the CNS, such as with an injury, activated astrocytes secrete various inflammatory mediators, such as chemokines, cytokines, and growth factors [79]. Astrocytes interact with microglia to regulate inflammatory responses in the brain. For instance, orosomucoid-2 (ORM2) derived from astrocytes effectively inhibited the proinflammatory activation of microglia via C-C chemokine ligand 4 (CCL4) during the late phase of neuroinflammation [80]. Recently, Liddelow and colleagues reported that activated microglia can induce the formation of A1 reactive astrocytes, a neurotoxic inflammatory astrocyte [81], by secreting cytokines, including IL-1α, TNFα, and C1q [77]. A subset of genes associated with reactive astrocytes was upregulated in the aged brain of wild-type mice, whereas their upregulation was significantly attenuated in mice lacking Il-1α, Tnfα, and C1q [82]. These data suggest that Il-1α, TNFα, and C1q are critical for activation of astrocytes in the aged brain.
Recently, two groups performed transcriptomic analyses in astrocytes isolated from multiple regions of young and aged mouse brains [82,83]. Both studies suggest that astrocytes have region-specific transcriptional identities and that their transcriptional changes with age are also region-dependent. Moreover, compared with young astrocytes, aged astrocytes show a stronger gene expression profile associated with reactive astrocytes [82,83]. A list of aging-induced DEG in astrocytes associated with immune responses, inflammatory responses, and antigen presentation signaling pathways is summarized in Table 2.

3. The Effects of Dietary Restriction on Neuroinflammation

3.1. The Effects of Dietary Restriction on Neuroinflammation in Normal Aging

The beneficial effects of DR on cognition and memory are under debate, with some studies reporting beneficial effects and others showing no benefits in the aging process [1,16,84,85,86,87,88,89,90,91,92,93]. However, there is agreement across studies that DR exerts anti-inflammatory effects against aging-driven neuroinflammation [15,17,94,95]. DR attenuated aging-driven increase in GFAP levels in multiple brain regions, including hypothalamus [96,97], hippocampus [97,98], corpus callosum [41], and cortex [97] in middle-aged rodents. Short-term DR in middle age in rhesus macaques also attenuated astrogliosis in hippocampus, suggesting conservation of the effects of DR on astrogliosis from rodents to nonhuman primates [5]. Downregulation of GFAP levels by DR is regulated at least in part at the transcriptional level [38,41]. Rozovsky and colleagues reported that neurite outgrowth was significantly reduced in cortical neurons cultured together with astrocytes derived from 24-month-old rats compared with those from 3-month-old rats [99]. Interestingly, knock-down of GFAP by RNAi diminished effects of aged astrocytes on neurite outgrowth, suggesting that the role of activated astrocytes in normal aging is not restricted to inflammatory regulation in the brain. Besides astrogliosis, DR also attenuated aging-driven microgliosis in corpus callosum, striatum, hippocampus, and hypothalamus in rodents [32,38,41,100]. DR downregulated circulating inflammatory mediators in the periphery [32,101,102]. In line with this, short-term DR in old age attenuated aging-associated induction of inflammatory cytokines, such as IL-1β in mouse hippocampus [28], TNF-α, and IL-6 in rat hypothalamus [32].
Morgan and colleagues showed that activation of astrocytes and microglia in rat brain was regulated by chronic DR from young to middle age in a region-specific manner [41]. Besides inflammation, DR also regulated aging-associated decrease in Sirtuin 1 (SIRT1) [103,104] and Brain-derived neurotrophic factor (BDNF) [105,106] in the brain in a region-specific manner, raising the possibility that different regulatory pathways may respond to DR or that the same regulatory pathways may respond to DR to different extents in different brain regions. Of note, gross effects of DR seen in the aged brain were not observed at young age [96,107,108], suggesting that molecular pathways regulated by DR are somehow inert in the brain at young age or that they respond to DR only when they are dysregulated over certain thresholds with advancing age.
Several studies have performed transcriptome analyses in dietary-restricted rodents during aging [109,110,111,112]. A number of genes associated with inflammation or the immune response showed changed expression levels under DR in aged rodent brains. When the DEGs for aged microglia were compared with the immune-related DEGs that were affected by DR, genes involved in antigen processing and presentation via MHC II were found in both lists (Table 1 and Table 3, H2-eb1, Ctse, H2-aa, H2-ab1, and Cd74). As mentioned above, MHC II is a marker for primed microglia. Interestingly, recent transcriptome and proteome analyses of human aged microglia have shown that genes and proteins related to antigen processing and presentation are significantly affected by aging [113]. Furthermore, overall age-related increases in the genes associated with antigen processing and presentation via MHC I have been reported in astrocytes (Table 2, [114]). Therefore, it is possible that the antigen processing and presentation pathway is a possible mechanistic link between DR and neuroinflammation.

3.2. The Effects of Dietary Restriction in Age-Related Neurodegenerative Diseases

Neuroinflammation is a major pathological hallmark of many neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS) [115,116,117,118]. So far, several studies have evaluated the beneficial effects of DR on AD in different mouse and monkey models. AD is pathologically characterized with abnormal accumulation of amyloid plaques and neurofibrillary tangles (NTFs), mainly composed of extracellular amyloid beta (Aβ) and intracellular tau, respectively [119]. Short-term DR before amyloid plaque accumulation significantly decreased accumulation of amyloid plaques and GFAP levels in two different mouse models of amyloidosis, J20 (DR for 6 weeks) and Tg2576 (DR for 14 weeks) [120]. J20 line expresses human APPswe/ind with the K670N/M671L (Swedish) and the V717F (Indiana) mutations [121], whereas Tg2576 line expresses human APPswe mutant with PS1 with M146L mutation [122]. Chronic DR for 12 months similarly decreased amyloid plaque burden in the brain of Tg25765 mice [123,124]. In another study, Mouton and colleagues sought to evaluate the therapeutic potential of DR at advanced pathologic stage in APPswe/PS1ΔE9 mouse model of AD [125]. APPswe/PS1ΔE9 mice aged 13–14 months with severe accumulation of amyloid plaques were subjected to DR for 4.5 months. DR significantly reduced amyloid plaque deposition in the brain, suggesting that DR may represent a novel therapeutic strategy for patients with advanced AD. It is noteworthy that DR attenuated amyloid pathology only in female Tg2576 and not in male mice [123]. By contrast, DR attenuated amyloid deposition in male APPswe/PS1ΔE9 line [125]. This conflicting finding may be due to different mouse models, different experimental settings, and/or different DR regimen in those two studies. Further investigations are needed to fully address whether sex influences the neuroprotective effects of DR on AD. Wu and colleagues demonstrated that DR attenuated astrogliosis in a double knockout of Psen1 and Psen2, another AD mouse model [126]. Ghrelin agonist, which induces hunger, also reduced the levels of insoluble Aβ and microgliosis in hippocampus of APPSwDI mice [127]. Whether DR reduces amyloid deposition in nonhuman primates is debatable. Although life-long DR reduced Aβ levels in temporal cortices of Squirrel monkeys [128], short-term DR in middle age did not reduce amyloid plaque burden in aged rhesus macaques [5]. However, DR attenuated astrogliosis in hippocampal CA region and entorhinal cortex in aged rhesus macaques [5]. In mouse models of tau pathology, DR has generated inconsistent results. Chronic DR reduced both Aβ and phospho-tau levels in hippocampus in the triple transgenic mouse model of AD, 3xTg mice expressing human APP KM670/671NL, TAU P301L, and PSEN1 M146V mutants with concomitant improvement in memory [129]. In a mouse model only expressing human mutant Tau (Tg4510), DR partially rescued memory deficits without altering tau accumulation, neuronal loss, or the levels of astroglial (GFAP) and microglial activation (Iba-1) [130].
PD is characterized by selective loss of dopaminergic neurons in the substantia nigra region [131]. DR ameliorated the loss of dopaminergic neuron and motor deficits in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced moue model of PD [132]. By contrast, no beneficial effects of DR were observed in 6-hydroxydopamine-induced rat model of PD [133]. In a MPTP-induced PD model of rhesus monkey, six months of DR improved locomotor activity and increased the levels of dopamine and its metabolites in striatum [134]. It is notable that DR attenuated MPTP-induced loss of dopaminergic neuron, astrogliosis, and microgliosis in substantia nigra in WT but not ghrelin KO mice, suggesting that ghrelin mediates the neuroprotective effects of DR in MPTP-induced mouse model of PD [135]. DR also attenuated pathologic events in a mouse model of Huntington’s disease (HD) [136].
In animal models of ALS, DR has generated conflicting results in different studies. Chronic DR accelerated clinical onset and progression and shortened the lifespan while transiently improving motor performance in a mouse model of ALS expressing human SOD1 G93A mutant [137], possibly by increasing lipid peroxidation, inflammation, and apoptosis [138]. In another study with the same model, no benefit of DR on disease onset or progression was observed [139]. However, DR significantly delayed the onset of disease and extended the lifespan in a different mouse model of ALS expressing SOD1 H46R/H48Q mutant [140]. Because of the substantial discrepancies across studies, it is difficult to draw a firm conclusion as to whether DR is beneficial in experimental models of ALS.
MS is a chronic inflammatory neurodegenerative disorder characterized by demyelination in the CNS [141]. In an experimental allergic encephalomyelitis (EAE), a rodent model of MS, 15 days of severe DR (66% food restriction) before EAE induction in 6 week-old male rats significantly attenuated progression of EAE [142]. In contrast to severe calorie restriction (66% food restriction), 15 days of mild DR (33% food restriction) before EAE induction in 6-week-old male rats had no inhibitory effect on the development of acute EAE [143]. Five weeks of mild DR (40% food restriction) before EAE induction in 5-week-old mice ameliorated clinical EAE with less severe inflammation, demyelination, and axon injury [144], suggesting that the effects of DR on EAE are dependent on severity and duration of regimen. A recent study demonstrated that periodic cycles of fasting mimicking diet with very-low-calorie and low-protein lasting 3 days every 7 days ameliorated demyelination and promoted oligodendrocyte precursor cell regeneration in a mouse model of EAE [145]. Although there is a report suggesting the beneficial effect of saturated fat restriction on MS in humans [146], it is currently not clear whether DR has protective and/or therapeutic effect on MS in humans.

4. A Potential Mechanistic Link between Dietary Restriction and Neuroinflammation

Although the precise mechanisms of DR’s neuroprotective functions are not fully elucidated, it has been suggested that DR exerts neuroprotective effects through multiple pathways, such as modulating metabolic rates, reducing oxidative stress, increasing anti-inflammatory responses, regulating insulin sensitivity, and improving synaptic plasticity and neurogenesis (reviewed in [15,16]). All of the molecular changes induced by DR may directly or indirectly contribute to the regulation of neuroinflammation associated with aging and neurodegenerative diseases. DR may directly mitigate activation of glial cells and modulate expression of inflammatory cytokines and indirectly regulate neuroinflammation by reducing inflammatory stresses, such as accumulation of toxic proteins and oxidative stress.
A previous gene profiling study provided evidence that DR increased IκBα, a NF-κB inhibitor, and decreased the p65 subunit of NF-κB in mouse neocortex [147]. Besides regulating expression of NF-κB, DR also reduced phosphorylation and activity of NF-κB in the brain of a mouse model of experimental astrocytoma [148]. These findings suggest that DR suppresses inflammation by inhibiting NF-κB signaling in the brain. It is well established that inflammation induces ROS generation in various cell types. In turn, ROS can activate redox-sensitive NF-κB, forming a positive feed-forward loop [149,150]. An increase of oxidative stress in the brain is a hallmark of aging as well as neurodegenerative diseases. It is evident that DR reduces oxidative stress in senescent astrocytes as well as in aged brains, as evidenced by reduction in ROS and protein oxidation [16,151,152,153,154]. Although how DR reduces oxidative stress in the brain remains elusive, several potential mechanisms of DR’s antioxidative functions have been proposed [155]. A recent transcriptome study showed that DR increased expression of ROS scavengers, such as glutathione S-transferases and thioredoxins, in cortices of rats [111]. In another study, Hyun and colleagues demonstrated that DR increased activities of multiple enzymes related to plasma membrane redox system and antioxidants, such as α-tocopherol and coenzyme Q10 [152]. DR also attenuated age-dependent induction of NADPH oxidase 2 (NOX2) in hypothalamus, which may contribute to the reduction of ROS by DR in the aged brain [32]. Taken together, it is likely that DR ameliorates neuroinflammation associated with aging and neurodegenerative diseases at least in part by reducing oxidative stress and thereby suppressing inflammatory responses in the brain.
Cellular redox status can also regulate SIRT1, a regulator of oxidative stress and inflammation [156]. SIRT1-mediated deacetylation of p65 subunit of NF-κB inhibits inflammatory responses via suppressing NF-κB signaling pathway [157]. SIRT1 can also regulate oxidative stress by modulating FOXO3, which regulates expression of antioxidant genes, such as MnSOD [158]. It is also notable that SIRT1 can regulate cellular redox status by modulating mitochondrial biogenesis by inducing PGC1-α and nitric oxide synthase [156,159]. Oxidative stress is known to suppress expression and activity of SIRT1 at the transcriptional and posttranslational levels [14,156]. In line with this, reduced SIRT1 expression has been reported in the brains of aged rodents [103,160]. Several lines of evidence have shown that SIRT1 level is increased by DR in multiple brain regions, including hypothalamus, hippocampus, and cortex [103,104,161,162]. Of note, it has been reported that Sirt1 transgenic mice have phenotypes that resemble DR [163] and show better physical activity in response to DR than wild-type mice [104]. By contrast, Sirt1-deficient mice exhibit defects in somatotropic and behavioral responses to DR [104,164]. SIRT1, together with NF-κB, seem to lie at the hub of antioxidative and anti-inflammatory responses mediated by DR in the brain.
Besides NF-κB and SIRT1-mediated pathways, several other pathways have been suggested as potential mechanisms mediating the anti-inflammatory action of DR, such as modulation of BBB permeability and regulation of steroid hormones in hypothalamic–pituitary–adrenal axis, such as glucocorticoid [20,165]. However, it is not clear whether these pathways indeed mediate anti-inflammatory action of DR in the brain due to a lack of solid evidence. Further studies are warranted to comprehensively understand mechanisms of anti-neuroinflammatory action of DR.

5. Conclusions

There is overwhelming evidence that DR attenuates inflammatory responses associated with aging in the brain. Figure 1 shows the possible relationship between DR and neuroinflammation during aging. However, the molecular bases of DR-mediated anti-inflammatory responses in the brain remain elusive. Moreover, it is not clear whether DR is neuroprotective for age-related neurodegenerative diseases because of mixed results. Thus, further in-depth studies are warranted to fully elucidate the molecular mechanisms of anti-inflammatory responses mediated by DR and whether DR can represent a novel therapeutic intervention for neurodegenerative diseases.

Author Contributions

E.B., M.J., J.K. and H.-J.K. collection material, writing the manuscript and revision; S.L. and B.-R.L. revision the manuscript, final editing.

Funding

This work was supported in part by the KBRI Research Program of the Ministry of Science, ICT & Future Planning, 19-BR-02-03 (H.-J.K.) and 19-BR-02-04 (J.K.), and the Basic Science Research Program through the National Research Foundation of Korea (NRF), funded by the Ministry of Science, ICT & Future Planning NRF-2017R1C1B2007941 (H.-J.K.).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ROSReactive oxygen species
DRDietary restriction
CALERIEComprehensive Assessment of Long-term Effects of Reducing Intake of Energy
BBBBlood–brain barrier
IL-1βInterleukin-1 beta
TNF-αTumor necrosis factor alpha
IL-6Interleukin-6
GFAPGlial fibrillary acidic protein
MHC IIMajor histocompatibility complex II
TLRsToll-like receptors
Iba-1Ionized calcium-binding adapter molecule 1
LPSLipopolysaccharide
TBITraumatic brain injury
CCL4C-C chemokine ligand 4
DEGDifferentially expressed genes
ORM2Orosomucoid-2
BDNFBrain-derived neurotrophic factor
SIRT1Sirtuin 1
ADAlzheimer’s disease
PDParkinson’s disease
ALSAmyotrophic lateral sclerosis
NTFsNeurofibrillary tangles
Amyloid beta
MPTPMethyl-4-phenyl-1,2,3,6-tetrahydropyridine
MSMultiple sclerosis
HDHuntington’s disease
ALAd libitum
EAEExperimental allergic encephalomyelitis
NOX2NADPH oxidase 2

References

  1. Hadem, I.K.H.; Majaw, T.; Kharbuli, B.; Sharma, R. Beneficial effects of dietary restriction in aging brain. J. Chem. Neuroanat. 2019, 95, 123–133. [Google Scholar] [CrossRef]
  2. Prolla, T.A.; Mattson, M.P. Molecular mechanisms of brain aging and neurodegenerative disorders: Lessons from dietary restriction. Trends Neurosci. 2001, 24, S21–S31. [Google Scholar] [CrossRef]
  3. McCay, C.M.; Crowell, M.F.; Maynard, L.A. The effect of retarded growth upon the length of life span and upon the ultimate body size. 1935. Nutrition 1989, 5, 155–171. [Google Scholar] [PubMed]
  4. Speakman, J.R.; Mitchell, S.E.; Mazidi, M. Calories or protein? The effect of dietary restriction on lifespan in rodents is explained by calories alone. Exp. Gerontol. 2016, 86, 28–38. [Google Scholar] [CrossRef]
  5. Sridharan, A.; Pehar, M.; Salamat, M.S.; Pugh, T.D.; Bendlin, B.B.; Willette, A.A.; Anderson, R.M.; Kemnitz, J.W.; Colman, R.J.; Weindruch, R.H.; et al. Calorie restriction attenuates astrogliosis but not amyloid plaque load in aged rhesus macaques: A preliminary quantitative imaging study. Brain Res. 2013, 1508, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Kastman, E.K.; Willette, A.A.; Coe, C.L.; Bendlin, B.B.; Kosmatka, K.J.; McLaren, D.G.; Xu, G.; Canu, E.; Field, A.S.; Alexander, A.L.; et al. A calorie-restricted diet decreases brain iron accumulation and preserves motor performance in old rhesus monkeys. J. Neurosci. 2012, 32, 11897–11904. [Google Scholar] [CrossRef] [PubMed]
  7. Mattison, J.A.; Roth, G.S.; Beasley, T.M.; Tilmont, E.M.; Handy, A.M.; Herbert, R.L.; Longo, D.L.; Allison, D.B.; Young, J.E.; Bryant, M.; et al. Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. Nature 2012, 489, 318–321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Willette, A.A.; Bendlin, B.B.; Colman, R.J.; Kastman, E.K.; Field, A.S.; Alexander, A.L.; Sridharan, A.; Allison, D.B.; Anderson, R.; Voytko, M.L.; et al. Calorie restriction reduces the influence of glucoregulatory dysfunction on regional brain volume in aged rhesus monkeys. Diabetes 2012, 61, 1036–1042. [Google Scholar] [CrossRef]
  9. Willette, A.A.; Coe, C.L.; Birdsill, A.C.; Bendlin, B.B.; Colman, R.J.; Alexander, A.L.; Allison, D.B.; Weindruch, R.H.; Johnson, S.C. Interleukin-8 and interleukin-10, brain volume and microstructure, and the influence of calorie restriction in old rhesus macaques. Age 2013, 35, 2215–2227. [Google Scholar] [CrossRef] [Green Version]
  10. Colman, R.J.; Anderson, R.M.; Johnson, S.C.; Kastman, E.K.; Kosmatka, K.J.; Beasley, T.M.; Allison, D.B.; Cruzen, C.; Simmons, H.A.; Kemnitz, J.W.; et al. Caloric restriction delays disease onset and mortality in rhesus monkeys. Science 2009, 325, 201–204. [Google Scholar] [CrossRef]
  11. Colman, R.J.; Beasley, T.M.; Kemnitz, J.W.; Johnson, S.C.; Weindruch, R.; Anderson, R.M. Caloric restriction reduces age-related and all-cause mortality in rhesus monkeys. Nat. Commun 2014, 5, 3557. [Google Scholar] [CrossRef] [PubMed]
  12. Most, J.; Tosti, V.; Redman, L.M.; Fontana, L. Calorie restriction in humans: An update. Ageing Res. Rev. 2017, 39, 36–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Redman, L.M.; Smith, S.R.; Burton, J.H.; Martin, C.K.; Il'yasova, D.; Ravussin, E. Metabolic Slowing and Reduced Oxidative Damage with Sustained Caloric Restriction Support the Rate of Living and Oxidative Damage Theories of Aging. Cell Metab. 2018, 27, 805–815.e4. [Google Scholar] [CrossRef] [PubMed]
  14. Fusco, S.; Pani, G. Brain response to calorie restriction. Cell. Mol. Life Sci. 2013, 70, 3157–3170. [Google Scholar] [CrossRef] [PubMed]
  15. Pani, G. Neuroprotective effects of dietary restriction: Evidence and mechanisms. Semin. Cell Dev. Biol. 2015, 40, 106–114. [Google Scholar] [CrossRef] [PubMed]
  16. Van Cauwenberghe, C.; Vandendriessche, C.; Libert, C.; Vandenbroucke, R.E. Caloric restriction: Beneficial effects on brain aging and Alzheimer’s disease. Mamm. Genome 2016, 27, 300–319. [Google Scholar] [CrossRef] [PubMed]
  17. Farooqui, A.A.; Horrocks, L.A.; Farooqui, T. Modulation of inflammation in brain: A matter of fat. J. Neurochem. 2007, 101, 577–599. [Google Scholar] [CrossRef] [PubMed]
  18. Franceschi, C.; Bonafe, M.; Valensin, S.; Olivieri, F.; De Luca, M.; Ottaviani, E.; De Benedictis, G. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann. N. Y. Acad. Sci. 2000, 908, 244–254. [Google Scholar] [CrossRef]
  19. Hotamisligil, G.S.; Erbay, E. Nutrient sensing and inflammation in metabolic diseases. Nat. Rev. Immunol. 2008, 8, 923–934. [Google Scholar] [CrossRef] [Green Version]
  20. Miller, A.A.; Spencer, S.J. Obesity and neuroinflammation: A pathway to cognitive impairment. Brain Behav. Immun. 2014, 42, 10–21. [Google Scholar] [CrossRef]
  21. Mattson, M.P.; Arumugam, T.V. Hallmarks of Brain Aging: Adaptive and Pathological Modification by Metabolic States. Cell Metab. 2018, 27, 1176–1199. [Google Scholar] [CrossRef] [PubMed]
  22. Di Benedetto, S.; Muller, L.; Wenger, E.; Duzel, S.; Pawelec, G. Contribution of neuroinflammation and immunity to brain aging and the mitigating effects of physical and cognitive interventions. Neurosci. Biobehav. Rev. 2017, 75, 114–128. [Google Scholar] [CrossRef] [PubMed]
  23. Hosoi, T.; Okuma, Y.; Nomura, Y. The mechanisms of immune-to-brain communication in inflammation as a drug target. Curr. Drug Targets Inflamm. Allergy 2002, 1, 257–262. [Google Scholar] [CrossRef] [PubMed]
  24. Sankowski, R.; Mader, S.; Valdes-Ferrer, S.I. Systemic inflammation and the brain: Novel roles of genetic, molecular, and environmental cues as drivers of neurodegeneration. Front. Cell. Neurosci. 2015, 9, 28. [Google Scholar] [CrossRef] [PubMed]
  25. Berchtold, N.C.; Cribbs, D.H.; Coleman, P.D.; Rogers, J.; Head, E.; Kim, R.; Beach, T.; Miller, C.; Troncoso, J.; Trojanowski, J.Q.; et al. Gene expression changes in the course of normal brain aging are sexually dimorphic. Proc. Natl. Acad. Sci. USA 2008, 105, 15605–15610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Cribbs, D.H.; Berchtold, N.C.; Perreau, V.; Coleman, P.D.; Rogers, J.; Tenner, A.J.; Cotman, C.W. Extensive innate immune gene activation accompanies brain aging, increasing vulnerability to cognitive decline and neurodegeneration: A microarray study. J. Neuroinflamm. 2012, 9, 179. [Google Scholar] [CrossRef] [PubMed]
  27. Soreq, L.; Consortium, U.K.B.E.; North American Brain Expression, C.; Rose, J.; Soreq, E.; Hardy, J.; Trabzuni, D.; Cookson, M.R.; Smith, C.; Ryten, M.; et al. Major Shifts in Glial Regional Identity Are a Transcriptional Hallmark of Human Brain Aging. Cell Rep. 2017, 18, 557–570. [Google Scholar] [CrossRef]
  28. Abraham, J.; Johnson, R.W. Consuming a diet supplemented with resveratrol reduced infection-related neuroinflammation and deficits in working memory in aged mice. Rejuvenation Res. 2009, 12, 445–453. [Google Scholar] [CrossRef]
  29. Godbout, J.P.; Chen, J.; Abraham, J.; Richwine, A.F.; Berg, B.M.; Kelley, K.W.; Johnson, R.W. Exaggerated neuroinflammation and sickness behavior in aged mice following activation of the peripheral innate immune system. FASEB J. 2005, 19, 1329–1331. [Google Scholar] [CrossRef]
  30. Swanson, K.S.; Vester, B.M.; Apanavicius, C.J.; Kirby, N.A.; Schook, L.B. Implications of age and diet on canine cerebral cortex transcription. Neurobiol. Aging 2009, 30, 1314–1326. [Google Scholar] [CrossRef]
  31. Chen, J.; Buchanan, J.B.; Sparkman, N.L.; Godbout, J.P.; Freund, G.G.; Johnson, R.W. Neuroinflammation and disruption in working memory in aged mice after acute stimulation of the peripheral innate immune system. Brain Behav. Immun. 2008, 22, 301–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Horrillo, D.; Sierra, J.; Arribas, C.; Garcia-San Frutos, M.; Carrascosa, J.M.; Lauzurica, N.; Fernandez-Agullo, T.; Ros, M. Age-associated development of inflammation in Wistar rats: Effects of caloric restriction. Arch. Physiol. Biochem. 2011, 117, 140–150. [Google Scholar] [CrossRef] [PubMed]
  33. Campuzano, O.; Castillo-Ruiz, M.M.; Acarin, L.; Castellano, B.; Gonzalez, B. Increased levels of proinflammatory cytokines in the aged rat brain attenuate injury-induced cytokine response after excitotoxic damage. J. Neurosci. Res. 2009, 87, 2484–2497. [Google Scholar] [CrossRef] [PubMed]
  34. Ye, S.M.; Johnson, R.W. Increased interleukin-6 expression by microglia from brain of aged mice. J. Neuroimmunol. 1999, 93, 139–148. [Google Scholar] [CrossRef]
  35. Maher, F.O.; Nolan, Y.; Lynch, M.A. Downregulation of IL-4-induced signalling in hippocampus contributes to deficits in LTP in the aged rat. Neurobiol. Aging 2005, 26, 717–728. [Google Scholar] [CrossRef] [PubMed]
  36. Ye, S.M.; Johnson, R.W. An age-related decline in interleukin-10 may contribute to the increased expression of interleukin-6 in brain of aged mice. Neuroimmunomodulation 2001, 9, 183–192. [Google Scholar] [CrossRef] [PubMed]
  37. Henry, C.J.; Huang, Y.; Wynne, A.M.; Godbout, J.P. Peripheral lipopolysaccharide (LPS) challenge promotes microglial hyperactivity in aged mice that is associated with exaggerated induction of both pro-inflammatory IL-1beta and anti-inflammatory IL-10 cytokines. Brain Behav. Immun. 2009, 23, 309–317. [Google Scholar] [CrossRef]
  38. Morgan, T.E.; Rozovsky, I.; Goldsmith, S.K.; Stone, D.J.; Yoshida, T.; Finch, C.E. Increased transcription of the astrocyte gene GFAP during middle-age is attenuated by food restriction: Implications for the role of oxidative stress. Free Radic. Biol. Med. 1997, 23, 524–528. [Google Scholar] [CrossRef]
  39. Morgan, T.E.; Wong, A.M.; Finch, C.E. Anti-inflammatory mechanisms of dietary restriction in slowing aging processes. Interdiscip. Top. Gerontol. 2007, 35, 83–97. [Google Scholar]
  40. Nichols, N.R.; Day, J.R.; Laping, N.J.; Johnson, S.A.; Finch, C.E. GFAP mRNA increases with age in rat and human brain. Neurobiol. Aging 1993, 14, 421–429. [Google Scholar] [CrossRef]
  41. Morgan, T.E.; Xie, Z.; Goldsmith, S.; Yoshida, T.; Lanzrein, A.S.; Stone, D.; Rozovsky, I.; Perry, G.; Smith, M.A.; Finch, C.E. The mosaic of brain glial hyperactivity during normal ageing and its attenuation by food restriction. Neuroscience 1999, 89, 687–699. [Google Scholar] [CrossRef]
  42. Sheng, J.G.; Mrak, R.E.; Griffin, W.S. Enlarged and phagocytic, but not primed, interleukin-1 alpha-immunoreactive microglia increase with age in normal human brain. Acta Neuropathol. 1998, 95, 229–234. [Google Scholar] [CrossRef] [PubMed]
  43. Goss, J.R.; Finch, C.E.; Morgan, D.G. GFAP RNA increases during a wasting state in old mice. Exp. Neurol. 1990, 108, 266–268. [Google Scholar] [CrossRef]
  44. O’Callaghan, J.P.; Miller, D.B. The concentration of glial fibrillary acidic protein increases with age in the mouse and rat brain. Neurobiol. Aging 1991, 12, 171–174. [Google Scholar] [CrossRef]
  45. Kohama, S.G.; Goss, J.R.; Finch, C.E.; McNeill, T.H. Increases of glial fibrillary acidic protein in the aging female mouse brain. Neurobiol. Aging 1995, 16, 59–67. [Google Scholar] [CrossRef]
  46. Haley, G.E.; Kohama, S.G.; Urbanski, H.F.; Raber, J. Age-related decreases in SYN levels associated with increases in MAP-2, apoE, and GFAP levels in the rhesus macaque prefrontal cortex and hippocampus. Age 2010, 32, 283–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Griffin, R.; Nally, R.; Nolan, Y.; McCartney, Y.; Linden, J.; Lynch, M.A. The age-related attenuation in long-term potentiation is associated with microglial activation. J. Neurochem. 2006, 99, 1263–1272. [Google Scholar] [CrossRef] [Green Version]
  48. Sloane, J.A.; Hollander, W.; Moss, M.B.; Rosene, D.L.; Abraham, C.R. Increased microglial activation and protein nitration in white matter of the aging monkey. Neurobiol. Aging 1999, 20, 395–405. [Google Scholar] [CrossRef]
  49. Hinman, J.D.; Duce, J.A.; Siman, R.A.; Hollander, W.; Abraham, C.R. Activation of calpain-1 in myelin and microglia in the white matter of the aged rhesus monkey. J. Neurochem. 2004, 89, 430–441. [Google Scholar] [CrossRef] [Green Version]
  50. Perry, V.H.; Matyszak, M.K.; Fearn, S. Altered antigen expression of microglia in the aged rodent CNS. Glia 1993, 7, 60–67. [Google Scholar] [CrossRef]
  51. Hanisch, U.K.; Kettenmann, H. Microglia: Active sensor and versatile effector cells in the normal and pathologic brain. Nat. Neurosci. 2007, 10, 1387–1394. [Google Scholar] [CrossRef] [PubMed]
  52. Wake, H.; Moorhouse, A.J.; Jinno, S.; Kohsaka, S.; Nabekura, J. Resting microglia directly monitor the functional state of synapses in vivo and determine the fate of ischemic terminals. J. Neurosci. 2009, 29, 3974–3980. [Google Scholar] [CrossRef] [PubMed]
  53. Nimmerjahn, A.; Kirchhoff, F.; Helmchen, F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 2005, 308, 1314–1318. [Google Scholar] [CrossRef] [PubMed]
  54. Schulz, C.; Gomez Perdiguero, E.; Chorro, L.; Szabo-Rogers, H.; Cagnard, N.; Kierdorf, K.; Prinz, M.; Wu, B.; Jacobsen, S.E.; Pollard, J.W.; et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 2012, 336, 86–90. [Google Scholar] [CrossRef] [PubMed]
  55. Kierdorf, K.; Erny, D.; Goldmann, T.; Sander, V.; Schulz, C.; Perdiguero, E.G.; Wieghofer, P.; Heinrich, A.; Riemke, P.; Holscher, C.; et al. Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways. Nat. Neurosci. 2013, 16, 273–280. [Google Scholar] [CrossRef] [PubMed]
  56. Ajami, B.; Bennett, J.L.; Krieger, C.; Tetzlaff, W.; Rossi, F.M. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat. Neurosci. 2007, 10, 1538–1543. [Google Scholar] [CrossRef] [PubMed]
  57. Hashimoto, D.; Chow, A.; Noizat, C.; Teo, P.; Beasley, M.B.; Leboeuf, M.; Becker, C.D.; See, P.; Price, J.; Lucas, D.; et al. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity 2013, 38, 792–804. [Google Scholar] [CrossRef] [PubMed]
  58. Fuger, P.; Hefendehl, J.K.; Veeraraghavalu, K.; Wendeln, A.C.; Schlosser, C.; Obermuller, U.; Wegenast-Braun, B.M.; Neher, J.J.; Martus, P.; Kohsaka, S.; et al. Microglia turnover with aging and in an Alzheimer’s model via long-term in vivo single-cell imaging. Nat. Neurosci. 2017, 20, 1371–1376. [Google Scholar] [CrossRef] [PubMed]
  59. Reu, P.; Khosravi, A.; Bernard, S.; Mold, J.E.; Salehpour, M.; Alkass, K.; Perl, S.; Tisdale, J.; Possnert, G.; Druid, H.; et al. The Lifespan and Turnover of Microglia in the Human Brain. Cell Rep. 2017, 20, 779–784. [Google Scholar] [CrossRef] [PubMed]
  60. Norden, D.M.; Muccigrosso, M.M.; Godbout, J.P. Microglial priming and enhanced reactivity to secondary insult in aging, and traumatic CNS injury, and neurodegenerative disease. Neuropharmacology 2015, 96, 29–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Norden, D.M.; Godbout, J.P. Review: Microglia of the aged brain: Primed to be activated and resistant to regulation. Neuropathol. Appl Neurobiol. 2013, 39, 19–34. [Google Scholar] [CrossRef] [PubMed]
  62. Barrientos, R.M.; Kitt, M.M.; Watkins, L.R.; Maier, S.F. Neuroinflammation in the normal aging hippocampus. Neuroscience 2015, 309, 84–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Perry, V.H. A revised view of the central nervous system microenvironment and major histocompatibility complex class II antigen presentation. J. Neuroimmunol. 1998, 90, 113–121. [Google Scholar] [CrossRef]
  64. Facci, L.; Barbierato, M.; Marinelli, C.; Argentini, C.; Skaper, S.D.; Giusti, P. Toll-like receptors 2, -3 and -4 prime microglia but not astrocytes across central nervous system regions for ATP-dependent interleukin-1beta release. Sci. Rep. 2014, 4, 6824. [Google Scholar] [CrossRef] [PubMed]
  65. Streit, W.J.; Xue, Q.S. The Brain’s Aging Immune System. Aging Dis. 2010, 1, 254–261. [Google Scholar] [PubMed]
  66. Hwang, I.K.; Lee, C.H.; Li, H.; Yoo, K.Y.; Choi, J.H.; Kim, D.W.; Kim, D.W.; Suh, H.W.; Won, M.H. Comparison of ionized calcium-binding adapter molecule 1 immunoreactivity of the hippocampal dentate gyrus and CA1 region in adult and aged dogs. Neurochem. Res. 2008, 33, 1309–1315. [Google Scholar] [CrossRef] [PubMed]
  67. Choi, J.H.; Lee, C.H.; Hwang, I.K.; Won, M.H.; Seong, J.K.; Yoon, Y.S.; Lee, H.S.; Lee, I.S. Age-related changes in ionized calcium-binding adapter molecule 1 immunoreactivity and protein level in the gerbil hippocampal CA1 region. J. Vet. Med. Sci. 2007, 69, 1131–1136. [Google Scholar] [CrossRef]
  68. Streit, W.J.; Sammons, N.W.; Kuhns, A.J.; Sparks, D.L. Dystrophic microglia in the aging human brain. Glia 2004, 45, 208–212. [Google Scholar] [CrossRef]
  69. Davies, D.S.; Ma, J.; Jegathees, T.; Goldsbury, C. Microglia show altered morphology and reduced arborization in human brain during aging and Alzheimer’s disease. Brain Pathol. 2017, 27, 795–808. [Google Scholar] [CrossRef]
  70. Barrientos, R.M.; Frank, M.G.; Hein, A.M.; Higgins, E.A.; Watkins, L.R.; Rudy, J.W.; Maier, S.F. Time course of hippocampal IL-1 beta and memory consolidation impairments in aging rats following peripheral infection. Brain Behav. Immun. 2009, 23, 46–54. [Google Scholar] [CrossRef]
  71. Onyszchuk, G.; He, Y.Y.; Berman, N.E.; Brooks, W.M. Detrimental effects of aging on outcome from traumatic brain injury: A behavioral, magnetic resonance imaging, and histological study in mice. J. Neurotrauma 2008, 25, 153–171. [Google Scholar] [CrossRef] [PubMed]
  72. Kumar, A.; Stoica, B.A.; Sabirzhanov, B.; Burns, M.P.; Faden, A.I.; Loane, D.J. Traumatic brain injury in aged animals increases lesion size and chronically alters microglial/macrophage classical and alternative activation states. Neurobiol. Aging 2013, 34, 1397–1411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Pelinka, L.E.; Kroepfl, A.; Leixnering, M.; Buchinger, W.; Raabe, A.; Redl, H. GFAP versus S100B in serum after traumatic brain injury: Relationship to brain damage and outcome. J. Neurotrauma 2004, 21, 1553–1561. [Google Scholar] [CrossRef] [PubMed]
  74. Holtman, I.R.; Raj, D.D.; Miller, J.A.; Schaafsma, W.; Yin, Z.; Brouwer, N.; Wes, P.D.; Moller, T.; Orre, M.; Kamphuis, W.; et al. Induction of a common microglia gene expression signature by aging and neurodegenerative conditions: A co-expression meta-analysis. Acta Neuropathol. Commun. 2015, 3, 31. [Google Scholar] [CrossRef] [PubMed]
  75. Pekny, M.; Pekna, M.; Messing, A.; Steinhauser, C.; Lee, J.M.; Parpura, V.; Hol, E.M.; Sofroniew, M.V.; Verkhratsky, A. Astrocytes: A central element in neurological diseases. Acta Neuropathol. 2016, 131, 323–345. [Google Scholar] [CrossRef] [PubMed]
  76. Jyothi, H.J.; Vidyadhara, D.J.; Mahadevan, A.; Philip, M.; Parmar, S.K.; Manohari, S.G.; Shankar, S.K.; Raju, T.R.; Alladi, P.A. Aging causes morphological alterations in astrocytes and microglia in human substantia nigra pars compacta. Neurobiol. Aging 2015, 36, 3321–3333. [Google Scholar] [CrossRef] [PubMed]
  77. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Munch, 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] [PubMed] [Green Version]
  78. Sofroniew, M.V.; Vinters, H.V. Astrocytes: Biology and pathology. Acta Neuropathol. 2010, 119, 7–35. [Google Scholar] [CrossRef]
  79. Colombo, E.; Farina, C. Astrocytes: Key Regulators of Neuroinflammation. Trends Immunol. 2016, 37, 608–620. [Google Scholar] [CrossRef]
  80. Jo, M.; Kim, J.H.; Song, G.J.; Seo, M.; Hwang, E.M.; Suk, K. Astrocytic Orosomucoid-2 Modulates Microglial Activation and Neuroinflammation. J. Neurosci. 2017, 37, 2878–2894. [Google Scholar] [CrossRef]
  81. Liddelow, S.A.; Barres, B.A. Reactive Astrocytes: Production, Function, and Therapeutic Potential. Immunity 2017, 46, 957–967. [Google Scholar] [CrossRef] [PubMed]
  82. Clarke, L.E.; Liddelow, S.A.; Chakraborty, C.; Munch, A.E.; Heiman, M.; Barres, B.A. Normal aging induces A1-like astrocyte reactivity. Proc. Natl. Acad. Sci. USA 2018, 115, E1896–E1905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Boisvert, M.M.; Erikson, G.A.; Shokhirev, M.N.; Allen, N.J. The Aging Astrocyte Transcriptome from Multiple Regions of the Mouse Brain. Cell Rep. 2018, 22, 269–285. [Google Scholar] [CrossRef] [PubMed]
  84. Hori, N.; Hirotsu, I.; Davis, P.J.; Carpenter, D.O. Long-term potentiation is lost in aged rats but preserved by calorie restriction. Neuroreport 1992, 3, 1085–1088. [Google Scholar] [CrossRef] [PubMed]
  85. Ingram, D.K.; Weindruch, R.; Spangler, E.L.; Freeman, J.R.; Walford, R.L. Dietary restriction benefits learning and motor performance of aged mice. J. Gerontol. 1987, 42, 78–81. [Google Scholar] [CrossRef]
  86. Joseph, J.A.; Whitaker, J.; Roth, G.S.; Ingram, D.K. Life-long dietary restriction affects striatally-mediated behavioral responses in aged rats. Neurobiol. Aging 1983, 4, 191–196. [Google Scholar] [CrossRef]
  87. Pitsikas, N.; Algeri, S. Deterioration of spatial and nonspatial reference and working memory in aged rats: Protective effect of life-long calorie restriction. Neurobiol. Aging 1992, 13, 369–373. [Google Scholar] [CrossRef]
  88. Roth, G.S.; Ingram, D.K.; Joseph, J.A. Delayed loss of striatal dopamine receptors during aging of dietarily restricted rats. Brain Res. 1984, 300, 27–32. [Google Scholar] [CrossRef]
  89. Beatty, W.W.; Clouse, B.A.; Bierley, R.A. Effects of long-term restricted feeding on radial maze performance by aged rats. Neurobiol. Aging 1987, 8, 325–327. [Google Scholar] [CrossRef]
  90. Bond, N.W.; Everitt, A.V.; Walton, J. Effects of dietary restriction on radial-arm maze performance and flavor memory in aged rats. Neurobiol. Aging 1989, 10, 27–30. [Google Scholar] [CrossRef]
  91. May, P.C.; Telford, N.; Salo, D.; Anderson, C.; Kohama, S.G.; Finch, C.E.; Walford, R.L.; Weindruch, R. Failure of dietary restriction to retard age-related neurochemical changes in mice. Neurobiol. Aging 1992, 13, 787–791. [Google Scholar] [CrossRef]
  92. Shi, L.; Poe, B.H.; Constance Linville, M.; Sonntag, W.E.; Brunso-Bechtold, J.K. Caloric restricted male rats demonstrate fewer synapses in layer 2 of sensorimotor cortex. Brain Res. 2002, 931, 32–40. [Google Scholar] [CrossRef]
  93. Minor, R.K.; Villarreal, J.; McGraw, M.; Percival, S.S.; Ingram, D.K.; de Cabo, R. Calorie restriction alters physical performance but not cognition in two models of altered neuroendocrine signaling. Behav. Brain Res. 2008, 189, 202–211. [Google Scholar] [CrossRef] [PubMed]
  94. Joseph, J.; Cole, G.; Head, E.; Ingram, D. Nutrition, brain aging, and neurodegeneration. J. Neurosci. 2009, 29, 12795–12801. [Google Scholar] [CrossRef] [PubMed]
  95. Vauzour, D.; Camprubi-Robles, M.; Miquel-Kergoat, S.; Andres-Lacueva, C.; Banati, D.; Barberger-Gateau, P.; Bowman, G.L.; Caberlotto, L.; Clarke, R.; Hogervorst, E.; et al. Nutrition for the ageing brain: Towards evidence for an optimal diet. Ageing Res. Rev. 2017, 35, 222–240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Nichols, N.R.; Finch, C.E.; Nelson, J.F. Food restriction delays the age-related increase in GFAP mRNA in rat hypothalamus. Neurobiol. Aging 1995, 16, 105–110. [Google Scholar] [CrossRef]
  97. Kaur, M.; Sharma, S.; Kaur, G. Age-related impairments in neuronal plasticity markers and astrocytic GFAP and their reversal by late-onset short term dietary restriction. Biogerontology 2008, 9, 441–454. [Google Scholar] [CrossRef]
  98. Major, D.E.; Kesslak, J.P.; Cotman, C.W.; Finch, C.E.; Day, J.R. Life-long dietary restriction attenuates age-related increases in hippocampal glial fibrillary acidic protein mRNA. Neurobiol. Aging 1997, 18, 523–526. [Google Scholar] [CrossRef]
  99. Rozovsky, I.; Wei, M.; Morgan, T.E.; Finch, C.E. Reversible age impairments in neurite outgrowth by manipulations of astrocytic GFAP. Neurobiol. Aging 2005, 26, 705–715. [Google Scholar] [CrossRef]
  100. Radler, M.E.; Wright, B.J.; Walker, F.R.; Hale, M.W.; Kent, S. Calorie restriction increases lipopolysaccharide-induced neuropeptide Y immunolabeling and reduces microglial cell area in the arcuate hypothalamic nucleus. Neuroscience 2015, 285, 236–247. [Google Scholar] [CrossRef]
  101. Matsuzaki, J.; Kuwamura, M.; Yamaji, R.; Inui, H.; Nakano, Y. Inflammatory responses to lipopolysaccharide are suppressed in 40% energy-restricted mice. J. Nutr. 2001, 131, 2139–2144. [Google Scholar] [CrossRef] [PubMed]
  102. Spaulding, C.C.; Walford, R.L.; Effros, R.B. Calorie restriction inhibits the age-related dysregulation of the cytokines TNF-alpha and IL-6 in C3B10RF1 mice. Mech. Ageing Dev. 1997, 93, 87–94. [Google Scholar] [CrossRef]
  103. Quintas, A.; de Solis, A.J.; Diez-Guerra, F.J.; Carrascosa, J.M.; Bogonez, E. Age-associated decrease of SIRT1 expression in rat hippocampus: Prevention by late onset caloric restriction. Exp. Gerontol. 2012, 47, 198–201. [Google Scholar] [CrossRef] [PubMed]
  104. Satoh, A.; Brace, C.S.; Ben-Josef, G.; West, T.; Wozniak, D.F.; Holtzman, D.M.; Herzog, E.D.; Imai, S. SIRT1 promotes the central adaptive response to diet restriction through activation of the dorsomedial and lateral nuclei of the hypothalamus. J. Neurosci. 2010, 30, 10220–10232. [Google Scholar] [CrossRef] [PubMed]
  105. Lee, J.; Duan, W.; Long, J.M.; Ingram, D.K.; Mattson, M.P. Dietary restriction increases the number of newly generated neural cells, and induces BDNF expression, in the dentate gyrus of rats. J. Mol. Neurosci. 2000, 15, 99–108. [Google Scholar] [CrossRef]
  106. Duan, W.; Lee, J.; Guo, Z.; Mattson, M.P. Dietary restriction stimulates BDNF production in the brain and thereby protects neurons against excitotoxic injury. J. Mol. Neurosci. 2001, 16, 1–12. [Google Scholar] [CrossRef]
  107. Kim, K.Y.; Ju, W.K.; Neufeld, A.H. Neuronal susceptibility to damage: Comparison of the retinas of young, old and old/caloric restricted rats before and after transient ischemia. Neurobiol. Aging 2004, 25, 491–500. [Google Scholar] [CrossRef]
  108. Sharma, S.; Kaur, G. Dietary restriction enhances kainate-induced increase in NCAM while blocking the glial activation in adult rat brain. Neurochem. Res. 2008, 33, 1178–1188. [Google Scholar] [CrossRef]
  109. Wahl, D.; Solon-Biet, S.M.; Wang, Q.P.; Wali, J.A.; Pulpitel, T.; Clark, X.; Raubenheimer, D.; Senior, A.M.; Sinclair, D.A.; Cooney, G.J.; et al. Comparing the Effects of Low-Protein and High-Carbohydrate Diets and Caloric Restriction on Brain Aging in Mice. Cell Rep. 2018, 25, 2234–2243. [Google Scholar] [CrossRef]
  110. Wu, P.; Jiang, C.; Shen, Q.; Hu, Y. Systematic gene expression profile of hypothalamus in calorie-restricted mice implicates the involvement of mTOR signaling in neuroprotective activity. Mech. Ageing Dev. 2009, 130, 602–610. [Google Scholar] [CrossRef]
  111. Wood, S.H.; van Dam, S.; Craig, T.; Tacutu, R.; O'Toole, A.; Merry, B.J.; de Magalhaes, J.P. Transcriptome analysis in calorie-restricted rats implicates epigenetic and post-translational mechanisms in neuroprotection and aging. Genome Biol. 2015, 16, 285. [Google Scholar] [CrossRef] [PubMed]
  112. Schafer, M.J.; Dolgalev, I.; Alldred, M.J.; Heguy, A.; Ginsberg, S.D. Calorie Restriction Suppresses Age-Dependent Hippocampal Transcriptional Signatures. PLoS ONE 2015, 10, e0133923. [Google Scholar] [CrossRef] [PubMed]
  113. Olah, M.; Patrick, E.; Villani, A.C.; Xu, J.; White, C.C.; Ryan, K.J.; Piehowski, P.; Kapasi, A.; Nejad, P.; Cimpean, M.; et al. A transcriptomic atlas of aged human microglia. Nat. Commun. 2018, 9, 539. [Google Scholar] [CrossRef] [PubMed]
  114. Mangold, C.A.; Masser, D.R.; Stanford, D.R.; Bixler, G.V.; Pisupati, A.; Giles, C.B.; Wren, J.D.; Ford, M.M.; Sonntag, W.E.; Freeman, W.M. CNS-wide Sexually Dimorphic Induction of the Major Histocompatibility Complex 1 Pathway With Aging. J. Gerontol. A Biol. Sci. Med. Sci. 2017, 72, 16–29. [Google Scholar] [CrossRef] [PubMed]
  115. DiSabato, D.J.; Quan, N.; Godbout, J.P. Neuroinflammation: The devil is in the details. J. Neurochem. 2016, 139 (Suppl. 2), 136–153. [Google Scholar] [CrossRef] [PubMed]
  116. Gelders, G.; Baekelandt, V.; Van der Perren, A. Linking Neuroinflammation and Neurodegeneration in Parkinson’s Disease. J. Immunol. Res. 2018, 2018, 4784268. [Google Scholar] [CrossRef]
  117. Heneka, M.T.; Carson, M.J.; El Khoury, J.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef]
  118. Liu, J.; Wang, F. Role of Neuroinflammation in Amyotrophic Lateral Sclerosis: Cellular Mechanisms and Therapeutic Implications. Front. Immunol. 2017, 8, 1005. [Google Scholar] [CrossRef]
  119. Holtzman, D.M.; Morris, J.C.; Goate, A.M. Alzheimer’s disease: The challenge of the second century. Sci. Transl. Med. 2011, 3, 77sr1. [Google Scholar] [CrossRef]
  120. Patel, N.V.; Gordon, M.N.; Connor, K.E.; Good, R.A.; Engelman, R.W.; Mason, J.; Morgan, D.G.; Morgan, T.E.; Finch, C.E. Caloric restriction attenuates Abeta-deposition in Alzheimer transgenic models. Neurobiol. Aging 2005, 26, 995–1000. [Google Scholar] [CrossRef]
  121. Mucke, L.; Masliah, E.; Yu, G.Q.; Mallory, M.; Rockenstein, E.M.; Tatsuno, G.; Hu, K.; Kholodenko, D.; Johnson-Wood, K.; McConlogue, L. High-level neuronal expression of abeta 1-42 in wild-type human amyloid protein precursor transgenic mice: Synaptotoxicity without plaque formation. J. Neurosci. 2000, 20, 4050–4058. [Google Scholar] [CrossRef] [PubMed]
  122. Holcomb, L.; Gordon, M.N.; McGowan, E.; Yu, X.; Benkovic, S.; Jantzen, P.; Wright, K.; Saad, I.; Mueller, R.; Morgan, D.; et al. Accelerated Alzheimer-type phenotype in transgenic mice carrying both mutant amyloid precursor protein and presenilin 1 transgenes. Nat. Med. 1998, 4, 97–100. [Google Scholar] [CrossRef] [PubMed]
  123. Schafer, M.J.; Alldred, M.J.; Lee, S.H.; Calhoun, M.E.; Petkova, E.; Mathews, P.M.; Ginsberg, S.D. Reduction of beta-amyloid and gamma-secretase by calorie restriction in female Tg2576 mice. Neurobiol. Aging 2015, 36, 1293–1302. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, J.; Ho, L.; Qin, W.; Rocher, A.B.; Seror, I.; Humala, N.; Maniar, K.; Dolios, G.; Wang, R.; Hof, P.R.; et al. Caloric restriction attenuates beta-amyloid neuropathology in a mouse model of Alzheimer’s disease. FASEB J. 2005, 19, 659–661. [Google Scholar] [CrossRef] [PubMed]
  125. Mouton, P.R.; Chachich, M.E.; Quigley, C.; Spangler, E.; Ingram, D.K. Caloric restriction attenuates amyloid deposition in middle-aged dtg APP/PS1 mice. Neurosci. Lett. 2009, 464, 184–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Wu, P.; Shen, Q.; Dong, S.; Xu, Z.; Tsien, J.Z.; Hu, Y. Calorie restriction ameliorates neurodegenerative phenotypes in forebrain-specific presenilin-1 and presenilin-2 double knockout mice. Neurobiol. Aging 2008, 29, 1502–1511. [Google Scholar] [CrossRef] [PubMed]
  127. Dhurandhar, E.J.; Allison, D.B.; van Groen, T.; Kadish, I. Hunger in the absence of caloric restriction improves cognition and attenuates Alzheimer’s disease pathology in a mouse model. PLoS ONE 2013, 8, e60437. [Google Scholar] [CrossRef]
  128. Qin, W.; Chachich, M.; Lane, M.; Roth, G.; Bryant, M.; de Cabo, R.; Ottinger, M.A.; Mattison, J.; Ingram, D.; Gandy, S.; et al. Calorie restriction attenuates Alzheimer’s disease type brain amyloidosis in Squirrel monkeys (Saimiri sciureus). J. Alzheimers Dis. 2006, 10, 417–422. [Google Scholar] [CrossRef]
  129. Halagappa, V.K.; Guo, Z.; Pearson, M.; Matsuoka, Y.; Cutler, R.G.; Laferla, F.M.; Mattson, M.P. Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer’s disease. Neurobiol. Dis. 2007, 26, 212–220. [Google Scholar] [CrossRef]
  130. Brownlow, M.L.; Joly-Amado, A.; Azam, S.; Elza, M.; Selenica, M.L.; Pappas, C.; Small, B.; Engelman, R.; Gordon, M.N.; Morgan, D. Partial rescue of memory deficits induced by calorie restriction in a mouse model of tau deposition. Behav. Brain Res. 2014, 271, 79–88. [Google Scholar] [CrossRef]
  131. Shulman, J.M.; De Jager, P.L.; Feany, M.B. Parkinson’s disease: Genetics and pathogenesis. Annu. Rev. Pathol. 2011, 6, 193–222. [Google Scholar] [CrossRef] [PubMed]
  132. Duan, W.; Mattson, M.P. Dietary restriction and 2-deoxyglucose administration improve behavioral outcome and reduce degeneration of dopaminergic neurons in models of Parkinson’s disease. J. Neurosci. Res. 1999, 57, 195–206. [Google Scholar] [CrossRef]
  133. Armentero, M.T.; Levandis, G.; Bramanti, P.; Nappi, G.; Blandini, F. Dietary restriction does not prevent nigrostriatal degeneration in the 6-hydroxydopamine model of Parkinson’s disease. Exp. Neurol. 2008, 212, 548–551. [Google Scholar] [CrossRef] [PubMed]
  134. Maswood, N.; Young, J.; Tilmont, E.; Zhang, Z.; Gash, D.M.; Gerhardt, G.A.; Grondin, R.; Roth, G.S.; Mattison, J.; Lane, M.A.; et al. Caloric restriction increases neurotrophic factor levels and attenuates neurochemical and behavioral deficits in a primate model of Parkinson’s disease. Proc. Natl. Acad. Sci. USA 2004, 101, 18171–18176. [Google Scholar] [CrossRef] [PubMed]
  135. Bayliss, J.A.; Lemus, M.B.; Stark, R.; Santos, V.V.; Thompson, A.; Rees, D.J.; Galic, S.; Elsworth, J.D.; Kemp, B.E.; Davies, J.S.; et al. Ghrelin-AMPK Signaling Mediates the Neuroprotective Effects of Calorie Restriction in Parkinson’s Disease. J. Neurosci. 2016, 36, 3049–3063. [Google Scholar] [CrossRef] [PubMed]
  136. Duan, W.; Guo, Z.; Jiang, H.; Ware, M.; Li, X.J.; Mattson, M.P. Dietary restriction normalizes glucose metabolism and BDNF levels, slows disease progression, and increases survival in huntingtin mutant mice. Proc. Natl. Acad. Sci. USA 2003, 100, 2911–2916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Hamadeh, M.J.; Rodriguez, M.C.; Kaczor, J.J.; Tarnopolsky, M.A. Caloric restriction transiently improves motor performance but hastens clinical onset of disease in the Cu/Zn-superoxide dismutase mutant G93A mouse. Muscle Nerve 2005, 31, 214–220. [Google Scholar] [CrossRef] [PubMed]
  138. Patel, B.P.; Safdar, A.; Raha, S.; Tarnopolsky, M.A.; Hamadeh, M.J. Caloric restriction shortens lifespan through an increase in lipid peroxidation, inflammation and apoptosis in the G93A mouse, an animal model of ALS. PLoS ONE 2010, 5, e9386. [Google Scholar] [CrossRef] [PubMed]
  139. Pedersen, W.A.; Mattson, M.P. No benefit of dietary restriction on disease onset or progression in amyotrophic lateral sclerosis Cu/Zn-superoxide dismutase mutant mice. Brain Res. 1999, 833, 117–120. [Google Scholar] [CrossRef]
  140. Bhattacharya, A.; Bokov, A.; Muller, F.L.; Jernigan, A.L.; Maslin, K.; Diaz, V.; Richardson, A.; Van Remmen, H. Dietary restriction but not rapamycin extends disease onset and survival of the H46R/H48Q mouse model of ALS. Neurobiol. Aging 2012, 33, 1829–1832. [Google Scholar] [CrossRef]
  141. Correale, J.; Gaitan, M.I.; Ysrraelit, M.C.; Fiol, M.P. Progressive multiple sclerosis: From pathogenic mechanisms to treatment. Brain A J. Neurol. 2017, 140, 527–546. [Google Scholar] [CrossRef] [PubMed]
  142. Esquifino, A.I.; Cano, P.; Jimenez, V.; Cutrera, R.A.; Cardinali, D.P. Experimental allergic encephalomyelitis in male Lewis rats subjected to calorie restriction. J. Physiol. Biochem. 2004, 60, 245–252. [Google Scholar] [CrossRef] [PubMed]
  143. Esquifino, A.I.; Cano, P.; Jimenez-Ortega, V.; Fernandez-Mateos, M.P.; Cardinali, D.P. Immune response after experimental allergic encephalomyelitis in rats subjected to calorie restriction. J. Neuroinflamm. 2007, 4, 6. [Google Scholar] [CrossRef] [PubMed]
  144. 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] [PubMed] [Green Version]
  145. Choi, I.Y.; Piccio, L.; Childress, P.; Bollman, B.; Ghosh, A.; Brandhorst, S.; Suarez, J.; Michalsen, A.; Cross, A.H.; Morgan, T.E.; et al. A Diet Mimicking Fasting Promotes Regeneration and Reduces Autoimmunity and Multiple Sclerosis Symptoms. Cell Rep. 2016, 15, 2136–2146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Swank, R.L.; Goodwin, J. Review of MS patient survival on a Swank low saturated fat diet. Nutrition 2003, 19, 161–162. [Google Scholar] [CrossRef]
  147. Lee, C.K.; Weindruch, R.; Prolla, T.A. Gene-expression profile of the ageing brain in mice. Nat. Genet. 2000, 25, 294–297. [Google Scholar] [CrossRef]
  148. Mulrooney, T.J.; Marsh, J.; Urits, I.; Seyfried, T.N.; Mukherjee, P. Influence of caloric restriction on constitutive expression of NF-kappaB in an experimental mouse astrocytoma. PLoS ONE 2011, 6, e18085. [Google Scholar] [CrossRef]
  149. Chung, H.Y.; Kim, H.J.; Kim, K.W.; Choi, J.S.; Yu, B.P. Molecular inflammation hypothesis of aging based on the anti-aging mechanism of calorie restriction. Microsc. Res. Tech. 2002, 59, 264–272. [Google Scholar] [CrossRef]
  150. Mhatre, M.; Floyd, R.A.; Hensley, K. Oxidative stress and neuroinflammation in Alzheimer’s disease and amyotrophic lateral sclerosis: Common links and potential therapeutic targets. J. Alzheimers Dis. 2004, 6, 147–157. [Google Scholar] [CrossRef]
  151. Santin, K.; da Rocha, R.F.; Cechetti, F.; Quincozes-Santos, A.; de Souza, D.F.; Nardin, P.; Rodrigues, L.; Leite, M.C.; Moreira, J.C.; Salbego, C.G.; et al. Moderate exercise training and chronic caloric restriction modulate redox status in rat hippocampus. Brain Res. 2011, 1421, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Hyun, D.H.; Emerson, S.S.; Jo, D.G.; Mattson, M.P.; de Cabo, R. Calorie restriction up-regulates the plasma membrane redox system in brain cells and suppresses oxidative stress during aging. Proc. Natl. Acad. Sci. USA 2006, 103, 19908–19912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Dubey, A.; Forster, M.J.; Lal, H.; Sohal, R.S. Effect of age and caloric intake on protein oxidation in different brain regions and on behavioral functions of the mouse. Arch. Biochem. Biophys. 1996, 333, 189–197. [Google Scholar] [CrossRef] [PubMed]
  154. Garcia-Matas, S.; Paul, R.K.; Molina-Martinez, P.; Palacios, H.; Gutierrez, V.M.; Corpas, R.; Pallas, M.; Cristofol, R.; de Cabo, R.; Sanfeliu, C. In vitro caloric restriction induces protective genes and functional rejuvenation in senescent SAMP8 astrocytes. Aging Cell 2015, 14, 334–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Walsh, M.E.; Shi, Y.; Van Remmen, H. The effects of dietary restriction on oxidative stress in rodents. Free Radic. Biol. Med. 2014, 66, 88–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Hwang, J.W.; Yao, H.; Caito, S.; Sundar, I.K.; Rahman, I. Redox regulation of SIRT1 in inflammation and cellular senescence. Free Radic. Biol. Med. 2013, 61, 95–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Yeung, F.; Hoberg, J.E.; Ramsey, C.S.; Keller, M.D.; Jones, D.R.; Frye, R.A.; Mayo, M.W. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 2004, 23, 2369–2380. [Google Scholar] [CrossRef] [PubMed]
  158. Brunet, A.; Sweeney, L.B.; Sturgill, J.F.; Chua, K.F.; Greer, P.L.; Lin, Y.; Tran, H.; Ross, S.E.; Mostoslavsky, R.; Cohen, H.Y.; et al. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 2004, 303, 2011–2015. [Google Scholar] [CrossRef] [PubMed]
  159. Rasouri, S.; Lagouge, M.; Auwerx, J. [SIRT1/PGC-1: A neuroprotective axis?]. Med. Sci. (Paris) 2007, 23, 840–844. [Google Scholar] [CrossRef]
  160. Lafontaine-Lacasse, M.; Richard, D.; Picard, F. Effects of age and gender on Sirt 1 mRNA expressions in the hypothalamus of the mouse. Neurosci. Lett. 2010, 480, 1–3. [Google Scholar] [CrossRef]
  161. Cunha-Santos, J.; Duarte-Neves, J.; Carmona, V.; Guarente, L.; Pereira de Almeida, L.; Cavadas, C. Caloric restriction blocks neuropathology and motor deficits in Machado-Joseph disease mouse models through SIRT1 pathway. Nat. Commun. 2016, 7, 11445. [Google Scholar] [CrossRef] [PubMed]
  162. Graff, J.; Kahn, M.; Samiei, A.; Gao, J.; Ota, K.T.; Rei, D.; Tsai, L.H. A dietary regimen of caloric restriction or pharmacological activation of SIRT1 to delay the onset of neurodegeneration. J. Neurosci. 2013, 33, 8951–8960. [Google Scholar] [CrossRef] [PubMed]
  163. Bordone, L.; Cohen, D.; Robinson, A.; Motta, M.C.; van Veen, E.; Czopik, A.; Steele, A.D.; Crowe, H.; Marmor, S.; Luo, J.; et al. SIRT1 transgenic mice show phenotypes resembling calorie restriction. Aging Cell 2007, 6, 759–767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Cohen, D.E.; Supinski, A.M.; Bonkowski, M.S.; Donmez, G.; Guarente, L.P. Neuronal SIRT1 regulates endocrine and behavioral responses to calorie restriction. Genes Dev. 2009, 23, 2812–2817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Vasconcelos, A.R.; Cabral-Costa, J.V.; Mazucanti, C.H.; Scavone, C.; Kawamoto, E.M. The Role of Steroid Hormones in the Modulation of Neuroinflammation by Dietary Interventions. Front. Endocrinol. (Lausanne) 2016, 7, 9. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of the potential mechanistic links between age-induced neuroinflammation and dietary restriction. All arrows indicate stimulation/consequences, and red T bar indicates mitigation.
Figure 1. Overview of the potential mechanistic links between age-induced neuroinflammation and dietary restriction. All arrows indicate stimulation/consequences, and red T bar indicates mitigation.
Ijms 20 00464 g001
Table 1. List of differentially expressed genes (DEGs) associated with inflammation/immune response in aged microglia.
Table 1. List of differentially expressed genes (DEGs) associated with inflammation/immune response in aged microglia.
Gene NameGO TermExpression Level Old vs. YoungAge/Brain RegionSpeciesReference
AxlGO:0002376, GO:0045087, GO:0006954Up24 months/whole brainMus musculus[74]
CampGO:0045087Up24 months/whole brainMus musculus[74]
Ccl3GO:0006954, GO:0050729, GO:0006955Up24 months/whole brainMus musculus[74]
Cd274GO:0006955Up24 months/whole brainMus musculus[74]
Cd36GO:0006955Up24 months/whole brainMus musculus[74]
Cd74GO:0002376, GO:0006955, GO:0019886, GO:0042613Up24 months/whole brainMus musculus[74]
Chst1GO:0006954Up24 months/whole brainMus musculus[74]
Clec7aGO:0045087, GO:0006954Up24 months/whole brainMus musculus[74]
CtseGO:0019886Up24 months/whole brainMus musculus[74]
Cxcl13GO:0006954, GO:0006955Up24 months/whole brainMus musculus[74]
CybbGO:0045087, GO:0006954Up24 months/whole brainMus musculus[74]
H2-aaGO:0002376, GO:0006955, GO:0019886, GO:0042613Up24 months/whole brainMus musculus[74]
H2-ab1GO:0002376, GO:0006955, GO:0019886, GO:0042613Up24 months/whole brainMus musculus[74]
H2-eb1GO:0002376, GO:0006955, GO:0019886, GO:0042613Up24 months/whole brainMus musculus[74]
Ifit3GO:0002376, GO:0045087Up24 months/whole brainMus musculus[74]
Ifitm2GO:0002376Up24 months/whole brainMus musculus[74]
Ifitm3GO:0002376, GO:0045087Up24 months/whole brainMus musculus[74]
Lcn2GO:0002376, GO:0045087Up24 months/whole brainMus musculus[74]
Lgals3GO:0002376, GO:0045087Up24 months/whole brainMus musculus[74]
LtfGO:0002376Up24 months/whole brainMus musculus[74]
Ly9GO:0002376, GO:0045087Up24 months/whole brainMus musculus[74]
Oasl2GO:0002376, GO:0045087, GO:0006955Up24 months/whole brainMus musculus[74]
Rsad2GO:0002376, GO:0045087Up24 months/whole brainMus musculus[74]
S100a8GO:0002376, GO:0045087, GO:0006954, GO:0050729Up24 months/whole brainMus musculus[74]
S100a9GO:0002376, GO:0045087, GO:0006954, GO:0050729Up24 months/whole brainMus musculus[74]
Spp1GO:0006954Up24 months/whole brainMus musculus[74]
GO:0002376, immune system process; GO:0006954, inflammatory response; GO:0006955, immune response; GO:0019886, antigen processing and presentation of exogenous peptide antigen via MHC class II; GO:0042613, MHC class II protein complex; GO:0045087, innate immune response; GO:0050729, positive regulation of inflammatory response. All gene listed passed the rule that log2 fold change >1 and adjusted p value <0.05.
Table 2. List of DEGs associated with inflammation/immune response in aged astrocytes.
Table 2. List of DEGs associated with inflammation/immune response in aged astrocytes.
Gene NameGO TermExpression Level Old vs. YoungAge/Brain RegionSpeciesReference
Akap8GO:0002376, GO:0045087Up24 months/StriatumMus musculus[82]
AppGO:0045087Up24 months/StriatumMus musculus[82]
B2mGO:0006955, GO:0002376, GO:0045087Up24 months/
Hippocampus, Striatum
Mus musculus[82]
Bcl6GO:0002376, GO:0006954Up24 months/
Cortex, Striatum
Mus musculus[82]
Bmp6GO:0006954Up24 months/
Visual cortex, Striatum
Mus musculus[82,83]
Bst2GO:0045087, GO:0002376Up24 months/Motor cortexMus musculus[83]
C3GO:0045087, GO:0002376, GO:0006954Up24 months/Motor cortex, Visual cortexMus musculus[83]
C4bGO:0045087, GO:0006954Up24 months/Motor cortex, Visual cortex, StriatumMus musculus[82,83]
Csf1GO:0002376, GO:0045087, GO:0006954Up24 months/StriatumMus musculus[82]
CtssGO:0019882, GO:0006955Up24 months/
Hippocampus, Striatum
Mus musculus[82]
Cxcl10GO:0006955, GO:0006954Up24 months/
Hippocampus, Striatum
Mus musculus[82]
Cxcl12GO:0006955Down24 months/HippocampusMus musculus[82]
Cxcl5GO:0006954Up24 months/Visual cortexMus musculus[83]
Defb1GO:0045087Up24 months/Motor cortex, Visual cortexMus musculus[83]
Enpp2GO:0006955Down24 months/HippocampusMus musculus[82]
Erap1GO:0002376Up24 months/StriatumMus musculus[82]
H2-d1GO:0019882, GO:0006955, GO:0002376Up24 months/Cortex, Hippocampus, StriatumMus musculus[82]
H2-k1GO:0019882, GO:0006955, GO:0002376Up24 months/
Hippocampus, Striatum
Mus musculus[82]
HfeGO:0019882Up24 months/StriatumMus musculus[82]
IcoslGO:0002376Up24 months/StriatumMus musculus[82]
Ifit1GO:0045087, GO:0002376Up24 months/Visual cortex, StriatumMus musculus[82,83]
Ifit3GO:0002376, GO:0045087Up24 months/Cortex, StriatumMus musculus[82]
Ifitm3GO:0002376, GO:0045087Up24 months/Cortex, StriatumMus musculus[82]
Ly86GO:0002376, GO:0045087, GO:0006954Up24 months/StriatumMus musculus[82]
Nlrp6GO:0045087, GO:0002376, GO:0006954Up24 months/Visual cortexMus musculus[83]
Oasl2GO:0045087, GO:0002376, GO:0006955Up24 months/Motor cortex, Visual cortex, Hippocampus, StriatumMus musculus[82,83]
Psmb8GO:0002376, GO:0019882Up24 months/Visual cortex, Hippocampus, StriatumMus musculus[82,83]
Psmb9GO:0002376, GO:0019882Up24 months/
Hippocampus, Striatum
Mus musculus[82]
Rsad2GO:0045087, GO:0002376Up24 months/Visual cortexMus musculus[83]
Serinc3GO:0002376, GO:0045087Up24 months/StriatumMus musculus[82]
Serping1GO:0002376, GO:0045087Up24 months/StriatumMus musculus[82]
Tspan2GO:0006954Up24 months/StriatumMus musculus[82]
TyrobpGO:0045087Up24 months/StriatumMus musculus[82]
Zc3hav1GO:0045087, GO:0002376Up24 months/Visual cortex, StriatumMus musculus[82,83]
GO:0019882, antigen processing and presentation; GO:0002376, immune system process; GO:0006954, inflammatory response; GO:0006955, immune response; GO:0045087, innate immune response. All gene listed passed the rule that log2 fold change >1 and adjusted p value <0.05.
Table 3. List of DEGs associated with inflammation/immune response in dietary restriction during aging.
Table 3. List of DEGs associated with inflammation/immune response in dietary restriction during aging.
Gene NameGO TermExpression Level DR vs. ALAge/Brain RegionSpeciesReference
PrlrGO:0034097Down15 months/HippocampusMus musculus[109]
Il2raGO:0034097Up15 months/HippocampusMus musculus[109]
SigirrGO:0034097Down15 months/HippocampusMus musculus[109]
Ptk2bGO:0002376Up15 months/Hippocampus (CA1)Mus musculus[112]
Bcl6GO:0006954, GO:0050727, GO:0002376Up15 months/Hippocampus (CA1)Mus musculus[112]
Ccr1GO:0006954Up15 months/Hippocampus (CA1)Mus musculus[112]
Il1r1GO:0050727Up15 months/Hippocampus (CA1)Mus musculus[112]
Tnfrsf25GO:0006954Up15 months/Hippocampus (CA1)Mus musculus[112]
GalGO:0006954Up15 months/Hippocampus (CA1)Mus musculus[112]
H2-q10GO:0002376Up15 months/Hippocampus (CA1)Mus musculus[112]
S100a8GO:0006954, GO:0050727, GO:0002376Up15 months/Hippocampus (CA1)Mus musculus[112]
S100a9GO:0006954, GO:0050727, GO:0002376Up15 months/Hippocampus (CA1)Mus musculus[112]
C1qbpGO:0006955Up28 months/Cerebral cortexRattus norvegicus[111]
Rt1-db1GO:0006955, GO:0019886, GO:0042613, GO:0002504Up28 months/Cerebral cortexRattus norvegicus[111]
Rt1-baGO:0006955, GO:0019886, GO:0042613, GO:0002504, GO:0019882Up28 months/Cerebral cortexRattus norvegicus[111]
Cxcl12GO:0006955Up28 months/Cerebral cortexRattus norvegicus[111]
Rt1-daGO:0006955, GO:0042613, GO:0002504, GO:0019882Up28 months/Cerebral cortexRattus norvegicus[111]
Cd74GO:0006955, GO:0019886, GO:0042613, GO:0019882Up28 months/Cerebral cortexRattus norvegicus[111]
Rt1-bbGO:0006955, GO:0019886, GO:0042613, GO:0002504, GO:0019882Up28 months/Cerebral cortexRattus norvegicus[111]
Fcer1gGO:0019886Up28 months/Cerebral cortexRattus norvegicus[111]
Rab3bGO:0019882Up28 months/Cerebral cortexRattus norvegicus[111]
Tnfaip6GO:0006954Down19 months/HypothalamusMus musculus[110]
C1qgGO:0002376Up19 months/HypothalamusMus musculus[110]
GO:0002376, immune system process; GO:0002504, antigen processing and presentation of peptide or polysaccharide antigen via MHC class II; GO:0006954, inflammatory response; GO:0006955, immune response; GO:0019882, antigen processing and presentation; GO:0019886, antigen processing and presentation of exogenous peptide antigen via MHC class II; GO:0034097, response to cytokine; GO:0042613, MHC class II protein complex; GO:0050727, regulation of inflammatory response. All gene listed passed the rule that log2 fold change >0.5 and adjusted p value <0.05.

Share and Cite

MDPI and ACS Style

Bok, E.; Jo, M.; Lee, S.; Lee, B.-R.; Kim, J.; Kim, H.-J. Dietary Restriction and Neuroinflammation: A Potential Mechanistic Link. Int. J. Mol. Sci. 2019, 20, 464. https://doi.org/10.3390/ijms20030464

AMA Style

Bok E, Jo M, Lee S, Lee B-R, Kim J, Kim H-J. Dietary Restriction and Neuroinflammation: A Potential Mechanistic Link. International Journal of Molecular Sciences. 2019; 20(3):464. https://doi.org/10.3390/ijms20030464

Chicago/Turabian Style

Bok, Eugene, Myungjin Jo, Shinrye Lee, Bo-Ram Lee, Jaekwang Kim, and Hyung-Jun Kim. 2019. "Dietary Restriction and Neuroinflammation: A Potential Mechanistic Link" International Journal of Molecular Sciences 20, no. 3: 464. https://doi.org/10.3390/ijms20030464

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop