Mechanisms Underlying Brain Aging Under Normal and Pathological Conditions

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Neurosci. Bull. February, 2023, 39(2):303–314 www.neurosci.

cn
https://doi.org/10.1007/s12264-022-00969-9 www.springer.com/12264

REVIEW

Mechanisms Underlying Brain Aging Under Normal


and Pathological Conditions
Menglong Jin1,2 · Shi‑Qing Cai1 

Received: 9 March 2022 / Accepted: 17 June 2022 / Published online: 27 November 2022
© The Author(s) 2022

Abstract  Aging is a major risk factor for many human developing cognitive impairment like Alzheimer’s disease
diseases, including cognitive impairment, which affects a (AD), the most common dementia that affects ~55 mil-
large population of the elderly. In the past few decades, our lion people worldwide and 15 million in China [5]. Thus,
understanding of the molecular and cellular mechanisms the development of new drugs or treatments to prevent or
underlying the changes associated with aging and age- reverse cognitive impairment requires a comprehensive
related diseases has expanded greatly, shedding light on the understanding of the biological events associated with the
potential role of these changes in cognitive impairment. In aging process in the brain.
this article, we review recent advances in understanding of In the past decades, our understanding of the molecu-
the mechanisms underlying brain aging under normal and lar mechanism underlying the modulation of aging has
pathological conditions, compare their similarities and dif- been greatly expanded. Most remarkably, several signaling
ferences, discuss the causative and adaptive mechanisms of pathways have been identified as key aging modulators in
brain aging, and finally attempt to find some rules to guide Caenorhabditis elegans (C. elegans), flies, and mammals,
us on how to promote healthy aging and prevent age-related suggesting that the biology of aging is conserved across
diseases. species and the rate of aging is plastic [6, 7]. Nine com-
mon age-related molecular and cellular alterations have been
Keywords  Healthy aging · Pathological aging · Cognitive proposed as hallmarks of aging [3]. Most of these hallmarks
impairment occur in aging neurons, except for cellular senescence and
telomere attrition (both of which usually occur in prolifera-
tive peripheral tissues), which remain to be established [8].
Introduction A myriad of evidence has implied that these hallmarks also
occur in pathological aging brains at a more severe level
As we age, our bodies undergo many changes [1–3] that can [9]. However, although brain atrophy and cognitive decline
be physiological and pathological. Just like other organs, appear both in normal and pathological aging brains, the
the brain is susceptible to aging and it undergoes a series mechanisms underlying these phenomena are not exactly the
of structural and functional changes during aging [2, 4]. In same. Cognitive decline in normal aging is attributed to the
the context of brain biology aging is a major risk factor for disruption of circuits and synapses, without significant neu-
ron loss, and this is in contrast to AD and related dementias,
which are associated with extensive neuronal death [10, 11].
* Shi‑Qing Cai
In this review, we mainly focus on several molecular and
sqcai@ion.ac.cn
1
cellular events (genomic instability, epigenetic alteration,
Institute of Neuroscience and State Key Laboratory
mitochondrial dysfunction, deregulated nutrient sensing,
of Neuroscience, CAS Center for Excellence in Brain Science
and Intelligence Technology, Chinese Academy of Sciences, altered cell proliferative capacity, altered intercellular com-
Shanghai 200031, China munication, and loss of proteostasis; Fig. 1) to review the
2
University of Chinese Academy of Sciences, Beijing 100049, mechanisms of normal aging and how those processes inter-
China face with pathological aging of the brain.

13
Vol.:(0123456789)

304 Neurosci. Bull. February, 2023, 39(2):303–314

the DNA damage by inducing aberrant synaptic activity,


which elicits more ROS [19]. Similarly, enhanced oxida-
tive DNA lesions and damage to mitochondrial DNA have
been reported in the neurons of amyotrophic lateral sclero-
sis (ALS) and Parkinson disease (PD) patients, respectively
[20–22].
Genomic instability is a key cause of age-related cogni-
tive impairment. Accumulating evidence has demonstrated
that impairment of DNA repair is sufficient to accelerate
aging phenotypes and induce neurodegeneration [23]. Cen-
tral dogma states that DNA transfers genetic information to
RNA and produces functional proteins. Thus, DNA dam-
age eventually affects the function of proteins, and therefore
Fig. 1  Molecular and cellular events occur in aging brains under nor- accumulation of DNA damage is a likely major cause of
mal and pathological conditions. Molecular and cellular alterations in
genomic stability, mitochondrial function, nutrient sensing, epigenetic chronic systemic failures, which are further worsened under
modification, cell proliferative capacity, intercellular communication, pathological conditions. This inevitably confers insults on
and proteostasis can be similar or different in aging brains under nor- the aging brain, consequently impairing cognitive function,
mal and pathological conditions. and may drive the onset or progression of neurodegenera-
tive diseases.
Genomic Instability

One common feature of aging is the accumulation of genetic Mitochondrial Dysfunction


damage throughout life [12]. Genomic DNA is regularly
damaged by radiation, reactive oxygen species (ROS), Mitochondrial impairment has been regarded as one of the
DNA replication errors, and other exogenous or endogenous hallmarks of aging [3]. Neurons are more susceptible to
threats, and damaged DNAs are progressively accumulated mitochondrial impairment as they need high energy to be
during the normal course of aging. Such accumulation is functional. In the aging brain, mitochondria exhibit a myriad
more serious in aging neurons because excitatory synaptic of structural and functional changes, including morphologi-
activity produces more ROS, causing more DNA damage. cal enlargement or fragmentation, membrane depolarization,
Furthermore, neurons are post-mitotic cells and can not an impaired electron transport chain, and increased oxida-
repair double-strand breaks by the more accurate homolo- tive damage to mitochondrial DNA (mtDNA) [3, 24]. The
gous recombination pathway. Indeed, analyses of tissue molecular mechanism underlying age-related dysfunction of
samples from human and rodent brains have revealed that mitochondria remains controversial. It has been suggested
the amount of damaged nuclear and mitochondrial DNA that mitochondrial dysfunction is due to the gradual accumu-
increases and the capacity for DNA repair decreases during lation of somatic mtDNA mutations. However, this hypoth-
aging [8]. In the cortex of the aging human brain, promoter esis has been challenged by the fact that a cell has multiple
regions of a considerable proportion of genes are damaged, mitochondrial genomes, which can tolerate the coexistence
resulting in reduction of expression of a subset of genes of wild-type and some mutant genomes [25]. Clearly, dys-
associated with synaptic plasticity, mitochondrial function, function of the mitochondrion is at least partially attributable
and neuronal survival [13]. Interestingly, a similar pattern to the age-related decline in the expression of mitochondrial
of DNA damage in vulnerable genes has also been found proteins [13]. A recent study provides evidence that epige-
in C. elegans, Drosophila, and mouse [14–17], suggesting netic changes contribute to mitochondrial dysfunction in the
that selective DNA damage to gene promoter sequences is aging brain. The age-related upregulation of bromodomain
a conserved mechanism underlying age-related cognitive adjacent to zinc finger domain 2B (BAZ2B) and euchro-
impairment. matic histone lysine methyltransferase 1 (EHMT1) underlie
Compared with the moderate DNA damage occurring the down-regulation of genes involving core metabolism
in normal aging brains, the damage to genomic DNA in (mainly ribosome and oxidative phosphorylation), which
aging brains under pathological conditions is much more have been consistently reported in patients with AD and
severe. Studies have shown that in human brains with AD, mild cognitive impairment [26].
the genomic DNA is evidently disrupted, and the exten- Pathological aging brains show more severe dysfunction
sive DNA damage may eventually lead to the death of a of mitochondria. Many lines of evidence have suggested
large number of neurons [18]. Aggregated β-amyloid pep- that mitochondrial dysfunction plays a central role in the
tide (Aβ), a major pathological hallmark of AD, worsens onset and development of neurodegenerative diseases. The

13
M. Jin, S.-Q. Cai: Mechanisms Underlying Brain Aging 305

age-related reductions in oxidative phosphorylation and


accumulation of mtDNA damage are aggravated in the
brains of AD patients [27–29]. Impaired mitochondrial
biogenesis and fission–fusion balance, as well as decreased
mitophagy, have also been reported in AD patients’ brains
[30, 31]. When it comes to the relationship between mito-
chondria and pathological brain aging, PD should not be
ignored [32, 33]. Although the exact cause of the disease
is not clear, aberrant mitochondrial clearance by autophagy
is a major factor in the death of dopaminergic neurons. The
protein kinase PTEN-induced putative kinase 1 (PINK1)
accumulates on the outer membrane of the mitochondria,
and then phosphorylates and activates the E3 ligase activity
of Parkin, which ubiquitinates outer mitochondrial mem-
brane proteins to trigger selective autophagy [34]. Healthy Fig. 2  Nutrient-sensing pathways and aging. Molecules proposed
to favor aging and prevent aging are shown in orange and green,
neurons effectively remove damaged mitochondria through respectively. IR, insulin receptor; IGF-1, insulin-like growth factor 1;
mitochondrial phagocytosis; while in PD neurons, exces- IGF1R, IGF-1 receptor; IRS, IR substrates; PI3K, phosphoinositide
sive mitochondrial damage causes a failure in the clearance 3-kinase; Akt, AKT serine/threonine kinase; FOXO, forkhead box
of damaged mitochondria, resulting in extensive neuronal O; GSK3, glycogen synthase kinase-3; mTOR, mammalian target
of rapamycin; AMPK, adenosine 5′ monophosphate-activated pro-
death. tein kinase; ULK1, Unc-51 like autophagy activating kinase 1; Sirt1,
Mitochondria establish and maintain the aging rate of the sirtuin 1; PGC1ɑ, PPARG coactivator 1 alpha; AMP, adenosine
entire organism [24], but the effect of mitochondrial damage monophosphate; ATP, adenosine triphosphate; N ­ AD+, nicotinamide
on the aging phenotypes is not simply linear. Previously, it adenine dinucleotide; NADH, nicotinamide adenine dinucleotide
hydride.
was supposed that an energetic crisis in aging brains caused
by accumulating damaged mitochondria accelerates func-
tional deterioration and triggers neurodegenerative diseases. The IIS pathway, which represents nutrient abundance
However, recent genetic and pharmacological evidence has and anabolism, is the most conserved aging-modifying path-
discovered that, although severe mitochondrial dysfunc- way in the process of evolution, and its downstream targets,
tion may cause cell death, mild respiratory defects extend forkhead box O (FOXO) family transcription factors, are
the lifespan. Down-regulating the function of the electron also highly conserved in regulating longevity across species:
transport chain in neurons has been reported to extend inhibiting the activity of the IIS pathway extends the lifespan
the lifespan of C. elegans [24]. On the other hand, several of yeast, worms, flies, and mammals [40]. Brain IIS plays a
reports have demonstrated that caloric restriction, genetic/ key role in sensing and regulating body metabolism, as well
pharmacological restoration of nicotinamide adenine dinu- as regulating synaptic plasticity, learning, and neural devel-
cleotide ­(NAD+), or manipulation of a glia-neuron signal opment [41]. It is worth noting that, although the IIS path-
can increase mitochondrial function and activation of the way plays a small role in glucose metabolism in the brain, it
mitochondrial unfolded protein response (­ UPRmt) to miti- is essential for maintaining a healthy brain metabolism [42].
gate many of the detrimental effects of aging and/or prolong The IIS pathway sends signals of the body’s energy status
the healthspan [35–39]. Perhaps maintaining mitochondrial to the brain and regulates eating behavior: excessive insulin
function at the proper level, not too low or too high, is most inhibits eating, while a decreased insulin level causes over-
beneficial for promoting healthy aging. eating through the IIS pathway in the hypothalamus [43, 44].
Interestingly, growth hormone and IGF-1 levels decrease
during normal aging, and this phenomenon has also been
Deregulated Nutrient Sensing reported in mouse models of premature aging [45]. One pos-
sible explanation for this is that cells compensate for aging
The function of the nutrient-sensing system changes with and age-related declines by downregulating the function of
age. Previous studies have shown that several nutrient-sens- IIS pathways. mTOR kinase, another highly-conserved fac-
ing pathways play a role in regulating longevity and aging tor for sensing and responding to nutrient signals (including
[40]. As shown in Fig. 2, these pathways include insulin and signals related to the availability of amino-acids), also plays
the insulin-like growth factor 1 (IGF-1) signaling (IIS) path- a key role in modulating aging, and administration of the
way, the mammalian target of rapamycin (mTOR) pathway, mTOR inhibitor rapamycin is considered to be one of the
the adenosine 5′ monophosphate-activated protein kinase most effective approaches to increasing the lifespan in mam-
(AMPK) pathway, and the sirtuin signaling pathway. mals [46, 47]. Of note, the activity of the mTOR signaling

13

306 Neurosci. Bull. February, 2023, 39(2):303–314

pathway also decreases during normal brain aging [48], but models of AD and stroke, dietary restriction protects neu-
in AD patients, the mTOR pathway is hyperactivated [49]. rons in the brain from degeneration [67]; however, no benefit
Some other pathways (like AMPK and sirtuins) sense nutri- of dietary restriction on alleviation of the disease has been
ent scarcity and catabolism, and they act in an opposite way reported in an animal model of ALS [68]. Recently, stud-
to modulate aging. Studies have shown that the responsive- ies on intermittent fasting (IF) are gaining increasing inter-
ness of AMPK signaling decreases with age and enhancing est [69]. Studies suggest that IF has no positive short-term
AMPK activity delays aging [50]. In general, the available effects on cognition in healthy subjects, but has benefits in
evidence has demonstrated that anabolic signaling accel- relieving the symptoms of epilepsy, AD, and multiple scle-
erates the rate of aging and reduced nutritional signaling rosis [70]. Although the exact mechanism of IF in healthy
increases longevity [51]. aging is still unclear, the process is thought to be associated
In aging brains, reduced glucose metabolism is found in with the IIS pathway, the mTOR pathway, and the like [71].
many brain regions, particularly marked in the temporal and
parietal lobes, and the motor cortex [52]. This alteration is
much more profound in these regions of elderly patients with
dementia [53]. In patients with neurodegenerative diseases, Epigenetic Alteration
such as AD and PD, reduced brain glucose consumption and
energy production are associated with altered IIS, mTOR, Epigenetics is the best choice for organisms to adapt to a
and AMPK signaling. Studies have shown that brain insulin changeable environment: heritable changes in gene expres-
resistance accelerates aging, while maintaining brain insu- sion occur while the DNA sequence does not change. Epi-
lin sensitivity delays aging [54]. An abnormal distribution genetic changes include alterations in DNA methylation
and cellular localization of insulin receptors (IR and IGF1R) patterns, post-translational modification of histones, chro-
have been reported in AD neurons, suggesting possible insu- matin remodeling, and expression of non-coding RNAs [72].
lin resistance in the AD brain [55]. The dysfunction of insu- Generally, diversity of modification helps individuals adapt
lin and IGF-1 signaling may also contribute to the progress to their environment; however, studies have shown that as
of AD, as insulin signaling plays a critical role in the clear- individuals age, heterogeneity of DNA modifications in the
ance of Aβ and inhibiting Tau aggregation [56]. Disrup- brain decreases at both the individual and tissue levels, and
tion of insulin receptor substrates 2 (IRS2) promotes Tau convergence of neighboring DNA modifications suggests
phosphorylation by activating glycogen synthase kinase-3β that there exist some rules of age-related alteration in DNA
in the Irs2−/− mouse brain [57]. Interestingly, other lines of modifications during brain aging [73]. Interestingly, changes
evidence support the idea that inhibition of IIS alleviates in the levels of DNA methylation at 353 CpG sites have been
AD-related phenotypes [58]. Neuron-specific knockout of proposed as an epigenetic clock (known as DNA methylation
IRS2 (but not IR or IGF1R) delays Aβ accumulation in a age) to predict the biological age of human tissues including
mouse model of AD [59], suggesting that the role of IIS in the brain [74]. Generally, histone methylation levels in the
AD pathology is distinct in different cell types. Inconsistent promoter regions of many synaptic function-related genes
changes in the AMPK pathway have also been reported in increase, while histone acetylation levels decrease in aging
the AD brain. Generally, AMPK activity decreases in the brains [75]. These epigenetic changes ultimately affect gene
AD brain, but it is abnormally activated in neurons bearing expression, leading to cognitive decline during aging.
neurofibrillary pre-tangles and tangles; activated AMPK Epigenetic alterations have also been widely reported in
accumulates near Aβ plaques and tau tangles instead of in pathological brain aging [76], and these alterations may be
the nucleus, where AMPK usually functions [60]. Thus, different from those that occur in normal aging. For exam-
moderating the sensitivity of AMPK pathways rather than ple, the methylation level in the promotor region of many
simply inhibiting or activating them (which may cause AD risk genes (such as APP, PS1, and BACE1) changes
resistance) may be more effective for relieving AD symp- significantly in AD patients’ brains compared with that
toms [54]. of age-matched normally aging brains [77]. However, the
The manipulation of nutritional signaling pathways may global changes in DNA methylation under AD conditions is
provide effective approaches to alleviate brain aging and pre- controversial: a decreased DNA methylation level has been
vent age-related neurodegenerative diseases. Dietary restric- found in the cell model of AD, but an increased DNA meth-
tion is probably the best known way to extend the lifes- ylation level has been reported in a mouse model of AD [78].
pan and the healthspan [61–63]. A reduced inflammatory Normal aging results in significant accumulation, while AD
response and improved capacity of DNA repair are found entails a marked loss, of H4K16ac in the proximity of genes
in dietary-restricted mice and rats, as a result of upregula- associated with the aging process and AD, suggesting that
tion of genes associated with inhibition of the inflamma- the marker H4K16ac plays a distinct role in normal aging
tory response and DNA synthesis/repair [64–66]. In animal and age-associated neurodegeneration [79].

13
M. Jin, S.-Q. Cai: Mechanisms Underlying Brain Aging 307

On the other hand, some epigenetic changes could be a of OSKM for a short period of time improves the aging phe-
continuous regulatory mechanism for normal and patho- notypes of aged mice and prolongs the life span of progeroid
logical brain aging. Studies have reported an increase in the mice [90], and the effect on delaying aging increases with
levels of H3K9me2 and H3K9me3 in aging brains of aged the cumulative time of partial reprogramming in aged mice,
mice, mouse models of AD and Huntington’s disease (HD), suggesting that epigenetic changes occur early during aging
as well as AD and HD patients [80, 81]. Pharmacological and that earlier interventions may get better results [91].
inhibition of the histone methyl transferases SUV39H1 and Notably, despite controversy, reprogramming glial cells into
EHMT1/EHMT2 decreases the levels of H3K9me3 in the neurons is an exciting and challenging approach to treating
hippocampus and improves cognitive function in aged mice neurodegenerative diseases [92, 93]. However, more work
and a mouse model of AD, respectively [82, 83]. Strikingly, remains to be done to understand epigenetic regulation in
a recent study found that the expression levels of EHMT1 aging brains under both normal and pathological conditions.
and an epigenetic reader BAZ2B increase with age, and are
positively correlated with the progress of AD in the pre-
frontal cortex of human brains. Ablation of Baz2b enhances Altered Cell Proliferative Capacity
mitochondrial function in the brain and improves cognitive
function in aged mice by affecting the H3K9 methylation In most organs, one of the most obvious age-related hall-
level in the promotor regions of genes associated with core marks is the decline in the regenerative potential of tissues,
metabolism [26]. which is a result of telomere attrition, stem cell exhaustion,
MicroRNAs (miRNAs) provide another epigenetic mech- and cellular senescence [94, 95]. Evidence shows that tel-
anism for regulating brain aging. Although dysfunction of omere alteration affects the rate of aging. It has been dem-
miRNAs affects neuroinflammation, cognitive decline, and onstrated that mice with shortened and lengthened telom-
other age-related deteriorations, currently little is known eres show decreased and increased lifespan, respectively.
about functional changes in brain miRNAs during aging Pathological dysfunction of telomeres accelerates aging,
[84]. Microarray data indicate that some miRNAs, including while experimental stimulation of telomerase delays aging
miR-139-5p, miR-342-3p, and miR-204, exhibit increased [96, 97]. However, neurons are permanently postmitotic and
expression levels in the hippocampus of aged mice, and neuronal telomeres do not shorten in aging brains. Interest-
they may suppress the surface expression of neurotransmit- ingly, changes in the telomere have shown an opposite role
ter receptors and contribute to age-related cognitive decline in different studies of pathological brain aging. Telomere
[85, 86]. Recent evidence suggests that miRNAs can be lengths are shorter in AD patients, and telomerase-deficient
packed into exosomes, which can be transported cross tis- mice showed signs of neurodegeneration [98, 99]. However,
sues and probably coordinate the ageing rate of different another study in the APP23 transgenic mouse have shown
organs. Astrocytic apolipoprotein E3 (APOE3) vesicles have that, although telomere shortening disrupts adult neurogen-
been reported to carry diverse regulatory miRNAs, which esis and the maintenance of neurons after mitosis, it reduces
specifically silence genes involved in neuronal cholesterol the formation of amyloid plaques and alleviates the deficits
biosynthesis, and ultimately promote histone acetylation and in learning and memory [100].
activate gene expression in neurons. In contrast, APOE4, a In the adult brain, neurogenesis only occurs in a few
risk factor for AD, carries fewer miRNAs and thus down- regions, including the hippocampal dentate gyrus, sub-ven-
regulates the acetylation level of histone in the promoter tricular zone (SVZ), and olfactory bulb [101, 102]. Age-
regions of synapse-related genes, ultimately leading to related changes in neurogenesis have been found in both
repressed expression of these genes and a decline in cogni- normal and pathological aging brains. During normal and
tive ability [87]. pathological aging, a decrease of neurogenesis in the hip-
Overall, the potential reversibility of epigenetic altera- pocampus, SVZ, and olfactory bulb may lead to cognitive
tions provides exciting chances to alter the trajectory of brain and olfactory deficits [103]. Neural stem cell transplanta-
aging under both normal and pathological conditions. Stud- tion has been considered as a potential treatment for some
ies have shown that some epigenetic factors regulate healthy age-related neurodegenerative diseases associated with neu-
aging, such as BAZ2B and EHMT1, and they may be tar- ronal loss [104]. However, aberrant neurogenesis has been
gets for slowing brain aging and treating age-related diseases reported in aging brains with neurodegenerative disorders,
[26]. Expression of octamer-binding transcription factor 3/4, including AD, PD, and HD [105]. For example, the human
SRY-Box transcription factor 2 (Sox2), Kruppel-like factor HD brain exhibits greater neural progenitor cell (NPC)
4, and the proto-oncogene c-Myc (OSKM, also known as proliferation that is proportional to the severity of the dis-
Yamanaka factors) in cells reshapes epigenetic markers and ease, and modifies the pathology of the disease [106]. Both
reprograms cells to a pluripotent state, as well as erasing increased and decreased neurogenesis have been reported in
cellular markers of senescence [88–90]. Periodic expression the brains of AD model mice and AD patients [105, 107].

13

308 Neurosci. Bull. February, 2023, 39(2):303–314

Some studies on postmortem brain tissue from AD patients is composed of bundles of axons) is compromised during
found a higher level of neurogenesis in the hippocampus normal aging of the human brain [121]. Not surprisingly,
[108], while other studies found that neurogenic markers like these alterations may render neurons more vulnerable to
Musashi-1, Sox2, and doublecortin are downregulated in the degeneration under pathological conditions [122], rein-
brains of AD patients [109, 110]. This discrepancy may be forcing the importance of maintaining synaptic function
explained by difference in the animal model used and the to achieve healthy aging of the brain. In support of this
stage of the disease. Overall, the capacity for neurogenesis notion, recent studies have demonstrated that improving
may change differently in normal and pathological brain synaptic function by enhancing neurotransmitter levels
aging, despite the fact that their states are both abnormal alleviates the age-related deterioration of cognitive func-
and may contribute to the cognitive impairments. tion in old people and animal models [123, 124]. How-
NPCs and proliferation-competent glial cells can undergo ever, other lines of evidence have shown that inhibition of
senescence. Cells with features of senescence have been neuronal excitability delays aging. Mutations that cause
found in aging brains under both normal and pathological defects in specific sensory neurons extends the lifespan
conditions [111]. Senescence alters cellular morphology and in C. elegans, and ablation of RE1 silencing transcription
proteostasis, decreases autophagy-lysosome function, and factor causes abnormal neural excitation in the aged mouse
enhances the secretory activity of cells, which leads to a brain and accelerates aging through the IIS pathway [125,
pro-inflammatory senescence-associated secretory pheno- 126]. These findings suggest that higher neural activity is
type [112]. Senescent cells accumulate during aging and not always better, and the changes of neural connections
negatively influence a heathy lifespan. In AD model mice, may also be an adaptive mechanism in response to aging.
the telomeres in microglia shorten during continuous repli- In addition to synaptic connections, factors such as neu-
cation, leading to replicative senescence and contributing to rotrophic and inflammation factors from other cells influ-
AD pathology [113]. Although neurons no longer undergo ence neuronal survival. In neurodegenerative diseases,
mitosis, they show some features of senescence in aging synaptic inputs may be altered by inhibiting anterograde
brains; the level of senescence-associated beta-galactosidase axonal transport and/or axonal degeneration, resulting in
activity increases in neurons in the CA3 region of aged rat reduced release of transmitters and neurotrophic peptides
brains, and a DNA damage-induced senescence-like phe- [127]. During aging, glial cells, particularly microglia, often
notype has also been reported in aging neurons [114, 115]. exhibit an activated state, causing long-term and chronic
Removal of senescent cells in the brain by pharmacological inflammation in the nervous system [128], which may lead
intervention, chimeric antigen receptor T cell therapy, or to cognitive impairments as evidenced by the findings that
genetic/antibody-mediated depletion alleviates the inflam- in both normal and pathological brain aging, inhibition of
mation and cognitive impairment in naturally aging mice, the inflammatory responses has been reported to improve
and prevents tau-dependent pathology and cognitive decline learning and cognition [129, 130]. For example, activating
in mouse models of neurodegenerative diseases [116–119]. the cholinergic anti-inflammatory pathway, inhibiting the
activation of microglia, or restoring the metabolic levels of
myeloid cells in aging mice alleviates inflammation in the
Altered Intercellular Communication nervous system and restores cognitive function [131–133].
NVUs also degenerate with age [134]. Changes of NVUs
Maintaining connections with other cells is critical for in aging usually result in impaired integrity of the blood-
preserving the functions of neurons. With increasing age, brain barrier (BBB) and infiltration of blood-derived pro-
the communication between neuronal cells changes signifi- teins, affecting the blood oxygen supply to nerve cells. The
cantly, causing decreased synaptic connections, increased degeneration of NVUs eventually impairs cognitive function
inflammation, and degenerated neurovascular units (NVUs) and increases the risk of stroke and cerebral small-vessel dis-
[3, 8]. ease, among others [135, 136]. Interestingly, unique changes
The fidelity of neural networks within and between in NVUs occur in some neurodegenerative diseases. For
brain regions is disturbed with age. Functional imaging example, Aβ42 has been reported to disturb the organization
studies have revealed a global loss of integrative func- of junctional complexes containing tight and adherent junc-
tion in aging human brains. Age-related dysfunction of tions between endothelial cells, causing leakage of plasma
neural networks is not due to a loss of neurons (which is protein and more severe damage in brains with AD [137].
minimal in normal aging brains), but to abnormal synaptic The deposition of α-synuclein and matrix metallopeptidase
connections that form the basis of neural circuits in the 3 damages the BBB and exacerbates inflammation in PD
hippocampus and prefrontal cortex [120]. The expression patients’ brains [138–140]. Thus, it is reasonable to assume
levels of most synaptic function-related genes are down- that protecting the functions of NVUs is a promising strategy
regulated [13] and the integrity of the white matter (which for preventing some neurodegenerative diseases.

13
M. Jin, S.-Q. Cai: Mechanisms Underlying Brain Aging 309

Loss of Proteostasis neuronal degeneration in post-mortem brain tissues from


patients with neurodegenerative diseases [145, 146]. Thus,
The proteostasis network regulates cellular protein homeo- activation of the UPR in the ER has distinct and even oppo-
stasis and it consists of the protein biosynthetic machinery, site effects on the progress of neurodegeneration, probably
the molecular chaperone system, and protein clearance path- depending on the stage of disease.
ways including the proteasome and autophagy systems. The It is worth noting that the relation between cognitive
loss of proteostasis, a failure of aging cells in responding impairment and toxic protein aggregation remains controver-
to proteotoxic challenges, has been considered a hallmark sial. In about a quarter of patients diagnosed as having AD-
of organismal aging [3]. A recent study has proposed that like diseases, the level of Aβ plaques and tau tangles does
age-related changes in translational efficiency initially drive not reach the diagnostic threshold of AD [147, 148]. Many
the deterioration of proteostasis [141]. Genetic and pharma- old people who exhibit extensive Aβ plaque accumulation
cological strategies to improve proteostasis promote healthy only have minimal neuronal loss and a marginal decline in
aging in animal models and delay the onset of age-related cognitive function [149, 150]. These suggest that there may
pathologies associated with protein aggregation [142]. be neuroprotective mechanisms that we have not yet found
In aged brains, the functions of protein chaperones and that influence the role of proteostasis during brain aging.
proteolytic systems decrease significantly [143]. Once the
capacity of these systems decreases below a threshold level,
some aggregation-prone proteins aggregate and exist in an Conclusions and Perspective
insoluble state. The threshold is lowered in some stress con-
ditions, such as the presence of disease-associated mutations Brain aging is a major risk factor for many neurodegenera-
in genes encoding aggregation-prone proteins [142]. Indeed, tive diseases, but the mechanisms underlying the transi-
in age-related neurodegenerative diseases, loss of protein tion from normal aging to neurodegenerative disorders are
homeostasis often shows more severe pathological features. complex and remain to be fully elucidated. The hallmarks
For example, aggregation of Aβ and hyperphosphorylation of brain aging have been associated with the pathogenesis
of Tau are the most obvious pathological features of AD; of neurodegenerative diseases, which are quite common
and a large amount of α-synuclein aggregates in dopamin- among older populations, leading to the hypothesis that neu-
ergic neurons in PD patients [144]. Stress response path- rodegeneration is accelerated brain ageing [151]. Indeed,
ways shape the proteostasis network, affecting the process as shown in Fig. 3, some hallmarks of brain aging occur
of aging and the onset/progress of neurodegeneration. The in both normal and pathological brain aging, but they are
aggregation of unfolded proteins perturbs the homeostasis more exacerbated in a pathological state. These are genomic
in the endoplasmic reticulum (ER) and triggers ER stress- instability, deregulated nutrient sensing, loss of proteosta-
induced activation of the unfolded protein response (UPR), sis, and mitochondrial dysfunction. However, some unique
an initially adaptive mechanism that restores cellular proteo- molecular and cellular processes occur in normal aging or
stasis [145, 146]. However, studies have demonstrated that neurodegenerative diseases. For instance, some changes in
chronic activation of the UPR in the ER is associated with intercellular communication (such as NUV damage caused

Fig. 3  Similar and different


molecular and cellular changes
in aging brains under normal
and pathological conditions.
Changes occurring in normal
and pathological aging brains
are shown in green and in
orange, respectively. These
changes are only examples,
not the full range of changes
during normal and pathologi-
cal brain aging. IIS, the insulin
and IGF-1 signaling pathway;
mTOR, mammalian target of
rapamycin.

13

310 Neurosci. Bull. February, 2023, 39(2):303–314

on single-cell transcriptomics [152] of normal, protected,


and pathological aging human brains may help to further
clarify the causative and adaptive age-related molecular and
cellular events in a cell type-specific pattern.
It is worth noting that the hallmarks of brain aging are
interrelated and interdependent, and none of them occur in
isolation. For example, age-related accumulation of DNA
damage and epigenetic aberrations alters gene expression,
resulting in dysfunction of mitochondria, deregulation of
nutrient sensing, and loss of proteostasis; all of which in
turn lead to more severe cytotoxicity and genomic insta-
bility, finally causing pathological changes in intercellular
communication and transforming physiological changes
Fig. 4  Functional interconnections between the molecular and cel- into pathological changes in the whole brain. Accumulating
lular events occurring in aging brains under normal and pathologi-
cal conditions. The seven molecular and cellular events occurring in
evidence has demonstrated that caloric restriction protects
aging brains can be divided into three categories. Genome instabil- aging brains from deterioration by stimulating mitochondrial
ity, loss of proteostasis, and altered cell proliferative capacity are sup- biogenesis and neural signaling pathways in neurons [8],
posed to be causative mechanisms; deregulated nutrient sensing and supporting a link between nutrient sensing and mitochon-
altered intercellular communication are mostly adaptive mechanisms;
while mitochondrial dysfunction and epigenetic alteration could be
drial function in modulating aging of the brain.
both causative and adaptive for brain aging and neurodegeneration. Given that normal and pathological brain aging are
closely linked, approaches that promote healthy aging are
worth trying to treat aging-related diseases. Recent stud-
by Aβ in AD [137]) and epigenetic modifications (such as ies have demonstrated that exposure of an aged animal to
methylation level in the promotor region of many AD risk young blood improves the aging phenotypes, and plasma
genes [77]) specifically occur in aging brains under patho- replacement has also been reported to be beneficial for treat-
logical conditions. Strikingly, other molecular and cellular ing AD [153–155]. A combination of removal of senescent
changes (such as alteration in telomeres and stem cells) in cells with transient somatic reprogramming synergistically
the brain are distinct between normal aging and neurode- extends lifespan and improves health [156]. On the other
generation, showing an antagonistic mechanism. It is worth hand, brain aging is a complex and integral process, there-
noting that the opposite changes that occur in neurodegen- fore future development of therapeutic approaches for treat-
erative diseases ultimately exacerbate the pathological phe- ing neurodegenerative diseases needs a greater understand-
notype rather than alleviate the aging phenotype. ing of not only the distinct molecular changes occurring in
Brain aging is a key regulator of longevity and functional pathological aging brains, but also the biological context
deterioration. Molecular and cellular alterations associated of normal aging of the brain. Many mouse models of AD
with the aging process in the brain could be adaptive fac- develop disease symptoms too early, which may not be suit-
tors for maintaining neural function or causative factors for able for the search for the true pathogenesis of the disease
deteriorating systemic functions [3]. Generally speaking, as [157, 158]. Researchers must take these questions into
shown in Fig. 4, genome instability, loss of proteostasis, and account when using these model mice.
altered cell proliferative capacity are causative mechanisms
for cognitive impairment [23, 95, 142]; while deregulated Acknowledgements  This review was supported by grants from
the National Key R&D Program of China (2018YFC2000400),
nutrient sensing and altered intercellular communication are
the National Natural Science Foundation of China (31925022 and
mainly considered to be adaptive mechanisms for brain aging 91949206), the Strategic Priority Research Program of the Chinese
because down-regulation of nutrient sensing and neuronal Academy of Sciences (XDB32020100), the Shanghai Municipal Sci-
excitability have been reported to extend lifespan [45, 125]. ence and Technology Major Project (2018SHZDZX05), and the Inno-
vative Research Team of High-level Local Universities in Shanghai
Mitochondrial dysfunction and epigenetic changes could be
(SHSMU-ZDCX20212501).
both causative and adaptive mechanisms for brain aging [24,
25, 72]. For example, mild downregulation of mitochondrial Open Access  This article is licensed under a Creative Commons
function promotes longevity, while severe impairment of Attribution 4.0 International License, which permits use, sharing, adap-
mitochondria leads to neuronal death and neurodegenera- tation, distribution and reproduction in any medium or format, as long
as you give appropriate credit to the original author(s) and the source,
tion. Yet, the causative and adaptive mechanisms are not provide a link to the Creative Commons licence, and indicate if changes
absolutely inconvertible; the adaptive molecular and cellular were made. The images or other third party material in this article are
changes in the aging brain may also contribute to the decline included in the article’s Creative Commons licence, unless indicated
of some subsets of behaviors. Future comprehensive studies otherwise in a credit line to the material. If material is not included in

13
M. Jin, S.-Q. Cai: Mechanisms Underlying Brain Aging 311

the article’s Creative Commons licence and your intended use is not 18. Adamec E, Vonsattel JP, Nixon RA. DNA strand breaks in Alz-
permitted by statutory regulation or exceeds the permitted use, you will heimer’s disease. Brain Res 1999, 849: 67–77.
need to obtain permission directly from the copyright holder. To view a 19. Merlo D, Cuchillo-Ibañez I, Parlato R, Rammes G. DNA damage,
copy of this licence, visit http://​creat​iveco​mmons.​org/​licen​ses/​by/4.​0/. neurodegeneration, and synaptic plasticity. Neural Plast 2016,
2016: 1206840.
20. Bender A, Krishnan KJ, Morris CM, Taylor GA, Reeve AK,
Perry RH, et al. High levels of mitochondrial DNA deletions
References in substantia nigra neurons in aging and Parkinson disease. Nat
Genet 2006, 38: 515–517.
21. Fitzmaurice PS, Shaw IC, Kleiner HE, Miller RT, Monks TJ,
1. de Almeida A, Ribeiro TP, de Medeiros IA. Aging: Molecular
Lau SS, et al. Evidence for DNA damage in amyotrophic lateral
Pathways and Implications on the Cardiovascular System. Oxid
sclerosis. Muscle Nerve 1996, 19: 797–798.
Med Cell Longev 2017, 2017: 7941563. https://​doi.​org/​10.​1155/​
22. Kraytsberg Y, Kudryavtseva E, McKee AC, Geula C, Kowall
2017/​79415​63.
NW, Khrapko K. Mitochondrial DNA deletions are abundant
2. Eikelboom WS, Bertens D, Kessels RPC. Cognitive rehabilita-
and cause functional impairment in aged human substantia nigra
tion in normal aging and individuals with subjective cognitive
neurons. Nat Genet 2006, 38: 518–520.
decline. Cognitive Rehabilitation and Neuroimaging. Cham:
23. Madabhushi R, Pan L, Tsai LH. DNA damage and its links to
Springer International Publishing, 2020: 37–67.
neurodegeneration. Neuron 2014, 83: 266–282.
3. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G.
24. Durieux J, Wolff S, Dillin A. The cell-non-autonomous nature
The hallmarks of aging. Cell 2013, 153: 1194–1217.
of electron transport chain-mediated longevity. Cell 2011, 144:
4. Nouchi R, Taki Y, Takeuchi H, Sekiguchi A, Hashizume
79–91.
H, Nozawa T. Four weeks of combination exercise training
25. Lin MT, Beal MF. Mitochondrial dysfunction and oxidative
improved executive functions, episodic memory, and process-
stress in neurodegenerative diseases. Nature 2006, 443: 787–795.
ing speed in healthy elderly people: Evidence from a randomized
26. Yuan J, Chang SY, Yin SG, Liu ZY, Cheng X, Liu XJ, et al. Two
controlled trial. Age (Dordr) 2014, 36: 787–799.
conserved epigenetic regulators prevent healthy ageing. Nature
5. Jia L, Du Y, Chu L, Zhang Z, Li F, Lyu D, et al. Prevalence, risk
2020, 579: 118–122.
factors, and management of dementia and mild cognitive impair-
27. Bonda DJ, Wang X, Lee HG, Smith MA, Perry G, Zhu X. Neu-
ment in adults aged 60 years or older in China: A cross-sectional
ronal failure in Alzheimer’s disease: A view through the oxida-
study. Lancet Public Health 2020, 5: e661–e671.
tive stress looking-glass. Neurosci Bull 2014, 30: 243–252.
6. Kenyon CJ. The genetics of ageing. Nature 2010, 464: 504–512.
28. Chen Z, Zhong C. Oxidative stress in Alzheimer’s disease. Neu-
7. Gems D, Partridge L. Genetics of longevity in model organ-
rosci Bull 2014, 30: 271–281.
isms: Debates and paradigm shifts. Annu Rev Physiol 2013, 75:
29. Wang W, Zhao F, Ma X, Perry G, Zhu X. Mitochondria dysfunc-
621–644.
tion in the pathogenesis of Alzheimer’s disease: Recent advances.
8. Mattson MP, Arumugam TV. Hallmarks of brain aging: Adaptive
Mol Neurodegeneration 2020, 15: 30.
and pathological modification by metabolic states. Cell Metab
30. Wang X, Su B, Lee HG, Li X, Perry G, Smith MA, et al.
2018, 27: 1176–1199.
Impaired balance of mitochondrial fission and fusion in Alzhei-
9. Hou Y, Dan X, Babbar M, Wei Y, Hasselbalch SG, Croteau DL,
mer’s disease. J Neurosci 2009, 29: 9090–9103.
et al. Ageing as a risk factor for neurodegenerative disease. Nat
31. Cai Q, Tammineni P. Alterations in mitochondrial quality control
Rev Neurol 2019, 15: 565–581.
in Alzheimer’s disease. Front Cell Neurosci 2016, 10: 24.
10. Dumitriu D, Hao J, Hara Y, Kaufmann J, Janssen WGM, Lou W,
32. Hirsch E, Graybiel AM, Agid YA. Melanized dopaminergic neu-
et al. Selective changes in thin spine density and morphology in
rons are differentially susceptible to degeneration in Parkinson’s
monkey prefrontal cortex correlate with aging-related cognitive
disease. Nature 1988, 334: 345–348.
impairment. J Neurosci 2010, 30: 7507–7515.
33. Galvan A, Wichmann T. Pathophysiology of Parkinsonism. Clin
11. Burke SN, Barnes CA. Neural plasticity in the ageing brain. Nat
Neurophysiol 2008, 119: 1459–1474.
Rev Neurosci 2006, 7: 30–40.
34. Pickrell AM, Youle RJ. The roles of PINK1, parkin, and mito-
12. Moskalev AA, Shaposhnikov MV, Plyusnina EN, Zhavoronkov
chondrial fidelity in Parkinson’s disease. Neuron 2015, 85:
A, Budovsky A, Yanai H, et al. The role of DNA damage and
257–273.
repair in aging through the prism of Koch-like criteria. Ageing
35. Nisoli E, Tonello C, Cardile A, Cozzi V, Bracale R, Tedesco L,
Res Rev 2013, 12: 661–684.
et al. Calorie restriction promotes mitochondrial biogenesis by
13. Lu T, Pan Y, Kao SY, Li C, Kohane I, Chan J, et al. Gene regula-
inducing the expression of eNOS. Science 2005, 310: 314–317.
tion and DNA damage in the ageing human brain. Nature 2004,
36. Civitarese AE, Carling S, Heilbronn LK, Hulver MH, Ukropcova
429: 883–891.
B, Deutsch WA, et al. Calorie restriction increases muscle mito-
14. Lopes AFC, Bozek K, Herholz M, Trifunovic A, Rieckher M,
chondrial biogenesis in healthy humans. PLoS Med 2007, 4: e76.
Schumacher BAC. elegans model for neurodegeneration in Coc-
37. Lanza IR, Zabielski P, Klaus KA, Morse DM, Heppelmann CJ,
kayne syndrome. Nucleic Acids Res 2020, 48: 10973–10985.
Bergen HR III, et al. Chronic caloric restriction preserves mito-
15. Shaposhnikov M, Proshkina E, Shilova L, Zhavoronkov A,
chondrial function in senescence without increasing mitochon-
Moskalev A. Lifespan and stress resistance in Drosophila with
drial biogenesis. Cell Metab 2012, 16: 777–788.
overexpressed DNA repair genes. Sci Rep 2015, 5: 15299.
38. Mouchiroud L, Houtkooper RH, Moullan N, Katsyuba E, Ryu D,
16. Oberdoerffer P, Michan S, McVay M, Mostoslavsky R, Vann
Cantó C, et al. The N ­ AD+/sirtuin pathway modulates longevity
J, Park SK, et al. SIRT1 redistribution on chromatin promotes
through activation of mitochondrial UPR and FOXO signaling.
genomic stability but alters gene expression during aging. Cell
Cell 2013, 154: 430–441.
2008, 135: 907–918.
39. Yin JA, Gao G, Liu XJ, Hao ZQ, Li K, Kang XL, et al. Genetic
17. Schumacher B, Pothof J, Vijg J, Hoeijmakers JHJ. The central
variation in glia–neuron signalling modulates ageing rate. Nature
role of DNA damage in the ageing process. Nature 2021, 592:
2017, 551: 198–203.
695–703.

13

312 Neurosci. Bull. February, 2023, 39(2):303–314

40. Fontana L, Partridge L, Longo VD. Extending healthy life span— 60. Vingtdeux V, Davies P, Dickson DW, Marambaud P. AMPK is
from yeast to humans. Science 2010, 328: 321–326. abnormally activated in tangle- and pre-tangle-bearing neurons
41. Akintola AA, van Heemst D. Insulin, aging, and the brain: Mech- in Alzheimer’s disease and other tauopathies. Acta Neuropathol
anisms and implications. Front Endocrinol (Lausanne) 2015, 6: 2011, 121: 337–349.
13. 61. Caprara G. Diet and longevity: The effects of traditional eating
42. Heni M, Kullmann S, Preissl H, Fritsche A, Häring HU. Impaired habits on human lifespan extension. Mediterr J Nutr Metab 2018,
insulin action in the human brain: Causes and metabolic conse- 11: 261–294.
quences. Nat Rev Endocrinol 2015, 11: 701–711. 62. McCay C, Crowell MF. Prolonging the life span. Science
43. Schwartz MW, Sipols AJ, Marks JL, Sanacora G, White JD, Monthly 1934, 39: 405–414.
Scheurink A, et al. Inhibition of hypothalamic neuropep- 63. Hansen M, Kennedy BK. Does longer lifespan mean longer
tide Y gene expression by insulin. Endocrinology 1992, 130: healthspan? Trends Cell Biol 2016, 26: 565–568.
3608–3616. 64. Vermeij WP, Dollé MET, Reiling E, Jaarsma D, Payan-Gomez C,
44. Brüning JC, Gautam D, Burks DJ, Gillette J, Schubert M, Orban Bombardieri CR, et al. Restricted diet delays accelerated ageing
PC, et al. Role of brain insulin receptor in control of body weight and genomic stress in DNA-repair-deficient mice. Nature 2016,
and reproduction. Science 2000, 289: 2122–2125. 537: 427–431.
45. Schumacher B, van der Pluijm I, Moorhouse MJ, Kosteas T, 65. Green CL, Lamming DW, Fontana L. Molecular mechanisms
Robinson AR, Suh Y, et al. Delayed and accelerated aging of dietary restriction promoting health and longevity. Nat Rev
share common longevity assurance mechanisms. PLoS Genet Mol Cell Biol 2022, 23: 56–73.
2008, 4: e1000161. 66. Morgan TE, Wong AM, Finch CE. Anti-inflammatory mecha-
46. Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flur- nisms of dietary restriction in slowing aging processes. Inter-
key K, et al. Rapamycin fed late in life extends lifespan in discip Top Gerontol 2007, 35: 83–97.
genetically heterogeneous mice. Nature 2009, 460: 392–395. 67. Prolla TA, Mattson MP. Molecular mechanisms of brain aging
47. Papadopoli D, Boulay K, Kazak L, Pollak M, Mallette F, Topi- and neurodegenerative disorders: Lessons from dietary restric-
sirovic I, et al. mTOR as a central regulator of lifespan and tion. Trends Neurosci 2001, 24: S21–S31.
aging. F1000Res 2019, 8: F1000FacultyRev–F1000Faculty998. 68. Pedersen WA, Mattson MP. No benefit of dietary restriction on
48. Romine J, Gao X, Xu XM, So KF, Chen J. The proliferation disease onset or progression in amyotrophic lateral sclerosis
of amplifying neural progenitor cells is impaired in the aging Cu/Zn-superoxide dismutase mutant mice. Brain Res 1999,
brain and restored by the mTOR pathway activation. Neurobiol 833: 117–120.
Aging 2015, 36: 1716–1726. 69. Varady K. Intermittent fasting is Gaining interest fast. Nat Rev
49. Li X, Alafuzoff I, Soininen H, Winblad B, Pei JJ. Levels of Mol Cell Biol 2021, 22: 587.
mTOR and its downstream targets 4E-BP1, eEF2, and eEF2 70. Gudden J, Arias Vasquez A, Bloemendaal M. The effects of
kinase in relationships with tau in Alzheimer’s disease brain. intermittent fasting on brain and cognitive function. Nutrients
FEBS J 2005, 272: 4211–4220. 2021, 13: 3166.
50. Salminen A, Kaarniranta K. AMP-activated protein kinase 71. Longo VD, di Tano M, Mattson MP, Guidi N. Intermittent and
(AMPK) controls the aging process via an integrated signal- periodic fasting, longevity and disease. Nat Aging 2021, 1:
ing network. Ageing Res Rev 2012, 11: 230–241. 47–59.
51. Barzilai N, Huffman DM, Muzumdar RH, Bartke A. The criti- 72. Bollati V, Baccarelli A. Environmental epigenetics. Heredity
cal role of metabolic pathways in aging. Diabetes 2012, 61: 2010, 105: 105–112.
1315–1322. 73. Oh G, Ebrahimi S, Wang SC, Cortese R, Kaminsky ZA, Gottes-
52. Goyal MS, Vlassenko AG, Blazey TM, Su Y, Couture LE, man II, et al. Epigenetic assimilation in the aging human brain.
Durbin TJ, et al. Loss of brain aerobic glycolysis in normal Genome Biol 2016, 17: 76.
human aging. Cell Metab 2017, 26: 353-360.e3. 74. Horvath S. DNA methylation age of human tissues and cell
53. Kato T, Inui Y, Nakamura A, Ito K. Brain fluorodeoxyglucose types. Genome Biol 2013, 14: R115.
(FDG) PET in dementia. Ageing Res Rev 2016, 30: 73–84. 75. Maity S, Farrell K, Navabpour S, Narayanan SN, Jarome TJ.
54. Masternak MM, Panici JA, Bonkowski MS, Hughes LF, Bartke Epigenetic mechanisms in memory and cognitive decline asso-
A. Insulin sensitivity as a key mediator of growth hormone ciated with aging and Alzheimer’s disease. Int J Mol Sci 2021,
actions on longevity. J Gerontol A Biol Sci Med Sci 2009, 22: 12280.
64A: 516–521. 76. Tecalco-Cruz AC, Ramírez-Jarquín JO, Alvarez-Sánchez ME,
55. Moloney AM, Griffin RJ, Timmons S, O’Connor R, Ravid Zepeda-Cervantes J. Epigenetic basis of Alzheimer disease.
R, O’Neill C. Defects in IGF-1 receptor, insulin receptor and World J Biol Chem 2020, 11: 62–75.
IRS-1/2 in Alzheimer’s disease indicate possible resistance 77. Chouliaras L, Rutten BPF, Kenis G, Peerbooms O, Visser PJ,
to IGF-1 and insulin signalling. Neurobiol Aging 2010, 31: Verhey F, et al. Epigenetic regulation in the pathophysiology
224–243. of Alzheimer’s disease. Prog Neurobiol 2010, 90: 498–510.
56. Gasparini L, Xu H. Potential roles of insulin and IGF-1 in Alz- 78. Sanchez-Mut JV, Gräff J. Epigenetic alterations in Alzheimer’s
heimer’s disease. Trends Neurosci 2003, 26: 404–406. disease. Front Behav Neurosci 2015, 9: 347.
57. Schubert M, Brazil DP, Burks DJ, Kushner JA, Ye J, Flint CL, 79. Nativio R, Donahue G, Berson A, Lan Y, Amlie-Wolf A, Tuzer
et al. Insulin receptor substrate-2 deficiency impairs brain F, et al. Dysregulation of the epigenetic landscape of normal
growth and promotes tau phosphorylation. J Neurosci 2003, 23: aging in Alzheimer’s disease. Nat Neurosci 2018, 21: 497–505.
7084–7092. 80. Ryu H, Lee J, Hagerty SW, Soh BY, McAlpin SE, Cormier
58. Candeias E, Duarte AI, Carvalho C, Correia SC, Cardoso S, San- KA, et al. ESET/SETDB1 gene expression and histone H3 (K9)
tos RX, et al. The impairment of insulin signaling in Alzheimer’s trimethylation in Huntington’s disease. Proc Natl Acad Sci U
disease. IUBMB Life 2012, 64: 951–957. S A 2006, 103: 19176–19181.
59. Freude S, Hettich MM, Schumann C, Stöhr O, Koch L, Köhler 81. Walker MP, LaFerla FM, Oddo SS, Brewer GJ. Reversible epi-
C, et al. Neuronal IGF-1 resistance reduces Abeta accumulation genetic histone modifications and Bdnf expression in neurons
and protects against premature death in a model of Alzheimer’s with aging and from a mouse model of Alzheimer’s disease.
disease. FASEB J 2009, 23: 3315–3324. AGE 2013, 35: 519–531.

13
M. Jin, S.-Q. Cai: Mechanisms Underlying Brain Aging 313

82. Snigdha S, Prieto GA, Petrosyan A, Loertscher BM, Dieskau neurogenesis in some brain regions. J Comp Neurol 1966, 126:
AP, Overman LE, et al. H3K9me3 inhibition improves mem- 337–389.
ory, promotes spine formation, and increases BDNF levels in 103. Lazarov O, Mattson MP, Peterson DA, Pimplikar SW, van Praag
the aged hippocampus. J Neurosci 2016, 36: 3611–3622. H. When neurogenesis encounters aging and disease. Trends
83. Zheng Y, Liu A, Wang ZJ, Cao Q, Wang W, Lin L, et al. Inhibi- Neurosci 2010, 33: 569–579.
tion of EHMT1/2 rescues synaptic and cognitive functions for 104. Lunn JS, Sakowski SA, Hur J, Feldman EL. Stem cell technology
Alzheimer’s disease. Brain 2019, 142: 787–807. for neurodegenerative diseases. Ann Neurol 2011, 70: 353–361.
84. Danka Mohammed CP, Park JS, Nam HG, Kim K. microRNAs 105. Liszewska E, Jaworski J. Neural stem cell dysfunction in human
in brain aging. Mech Ageing Dev 2017, 168: 3–9. brain disorders. Results Probl Cell Differ 2018, 66: 283–305.
85. Cosín-Tomás M, Alvarez-López MJ, Sanchez-Roige S, Lalanza 106. Curtis MA, Penney EB, Pearson AG, van Roon-Mom WMC,
JF, Bayod S, Sanfeliu C, et al. Epigenetic alterations in hip- Butterworth NJ, Dragunow M, et al. Increased cell proliferation
pocampus of SAMP8 senescent mice and modulation by vol- and neurogenesis in the adult human Huntington’s disease brain.
untary physical exercise. Front Aging Neurosci 2014, 6: 51. Proc Natl Acad Sci U S A 2003, 100: 9023–9027.
86. Mohammed CPD, Rhee H, Phee BK, Kim K, Kim HJ, Lee H, et 107. Liu H, Song N. Molecular mechanism of adult neurogenesis and
al. miR-204 downregulates EphB2 in aging mouse hippocampal its association with human brain diseases. J Cent Nerv Syst Dis
neurons. Aging Cell 2016, 15: 380–388. 2016, 8: 5–11.
87. Li X, Zhang J, Li D, He C, He K, Xue T, et al. Astrocytic ApoE 108. Ghosal K, Stathopoulos A, Pimplikar SW. APP intracellular
reprograms neuronal cholesterol metabolism and histone-acety- domain impairs adult neurogenesis in transgenic mice by induc-
lation-mediated memory. Neuron 2021, 109: 957-970.e8. ing neuroinflammation. PLoS One 2010, 5: e11866.
88. Takahashi K, Yamanaka S. Induction of pluripotent stem cells 109. Crews L, Adame A, Patrick C, Delaney A, Pham E, Rockenstein
from mouse embryonic and adult fibroblast cultures by defined E, et al. Increased BMP6 levels in the brains of Alzheimer’s
factors. Cell 2006, 126: 663–676. disease patients and APP transgenic mice are accompanied by
89. Buganim Y, Faddah DA, Jaenisch R. Mechanisms and models of impaired neurogenesis. J Neurosci 2010, 30: 12252–12262.
somatic cell reprogramming. Nat Rev Genet 2013, 14: 427–439. 110. Perry EK, Johnson M, Ekonomou A, Perry RH, Ballard C,
90. Ocampo A, Reddy P, Martinez-Redondo P, Platero-Luengo A, Attems J. Neurogenic abnormalities in Alzheimer’s disease dif-
Hatanaka F, Hishida T, et al. In vivo amelioration of age-associ- fer between stages of neurogenesis and are partly related to cho-
ated hallmarks by partial reprogramming. Cell 2016, 167: 1719- linergic pathology. Neurobiol Dis 2012, 47: 155–162.
1733.e12. 111. Sikora E, Bielak-Zmijewska A, Dudkowska M, Krzystyniak A,
91. Browder KC, Reddy P, Yamamoto M, Haghani A, Guillen IG, Mosieniak G, Wesierska M, et al. Cellular senescence in brain
Sahu S, et al. In vivo partial reprogramming alters age-associ- aging. Front Aging Neurosci 2021, 13: 646924.
ated molecular changes during physiological aging in mice. Nat 112. Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O,
Aging 2022, 2: 243–253. Bishop C, et al. Cellular senescence: Defining a path forward.
92. Wang F, Cheng L, Zhang X. Reprogramming glial cells into Cell 2019, 179: 813–827.
functional neurons for neuro-regeneration: Challenges and prom- 113. Hu Y, Fryatt GL, Ghorbani M, Obst J, Menassa DA, Martin-
ise. Neurosci Bull 2021, 37: 1625–1636. Estebane M, et al. Replicative senescence dictates the emergence
93. Jiang Y, Wang Y, Huang Z. Targeting PTB as a one-step proce- of disease-associated microglia and contributes to Aβ pathology.
dure for in situ astrocyte-to-dopamine neuron reprogramming in Cell Rep 2021, 35: 109228.
Parkinson’s disease. Neurosci Bull 2021, 37: 430–432. 114. Geng YQ, Guan JT, Xu XH, Fu YC. Senescence-associated beta-
94. Sousounis K, Baddour JA, Tsonis PA. Aging and regeneration in galactosidase activity expression in aging hippocampal neurons.
vertebrates. Curr Top Dev Biol 2014, 108: 217–246. Biochem Biophys Res Commun 2010, 396: 866–869.
95. Rando TA, Jones DL. Regeneration, rejuvenation, and replace- 115. Jurk D, Wang C, Miwa S, Maddick M, Korolchuk V, Tsolou A, et
ment: Turning back the clock on tissue aging. Cold Spring Harb al. Postmitotic neurons develop a p21-dependent senescence-like
Perspect Biol 2021, 13: a040907. phenotype driven by a DNA damage response. Aging Cell 2012,
96. Calado RT, Dumitriu B. Telomere dynamics in mice and humans. 11: 996–1004.
Semin Hematol 2013, 50: 165–174. 116. Bussian TJ, Aziz A, Meyer CF, Swenson BL, van Deursen JM,
97. Liu J, Wang L, Wang Z, Liu JP. Roles of telomere biology in cell Baker DJ. Clearance of senescent glial cells prevents tau-depend-
senescence, replicative and chronological ageing. Cells 2019, 8: ent pathology and cognitive decline. Nature 2018, 562: 578–582.
54. 117. Jin WN, Shi K, He W, Sun JH, van Kaer L, Shi FD, et al. Neu-
98. Hochstrasser T, Marksteiner J, Humpel C. Telomere length is roblast senescence in the aged brain augments natural killer cell
age-dependent and reduced in monocytes of Alzheimer patients. cytotoxicity leading to impaired neurogenesis and cognition. Nat
Exp Gerontol 2012, 47: 160–163. Neurosci 2021, 24: 61–73.
99. Whittemore K, Derevyanko A, Martinez P, Serrano R, Pumarola 118. Jin JL, Liou AK, Shi Y, Yin KL, Chen L, Li LL, et al. CART
M, Bosch F, et al. Telomerase gene therapy ameliorates the treatment improves memory and synaptic structure in APP/PS1
effects of neurodegeneration associated to short telomeres in mice. Sci Rep 2015, 5: 10224.
mice. Aging 2019, 11: 2916–2948. 119. Amor C, Feucht J, Leibold J, Ho YJ, Zhu C, Alonso-Curbelo
100. Rolyan H, Scheffold A, Heinrich A, Begus-Nahrmann Y, Lang- D, et al. Senolytic CAR T cells reverse senescence-associated
kopf BH, Hölter SM, et al. Telomere shortening reduces Alz- pathologies. Nature 2020, 583: 127–132.
heimer’s disease amyloid pathology in mice. Brain 2011, 134: 120. Bishop NA, Lu T, Yankner BA. Neural mechanisms of ageing
2044–2056. and cognitive decline. Nature 2010, 464: 529–535.
101. Altman J, Das GD. Autoradiographic and histological evidence 121. Cox SR, Ritchie SJ, Tucker-Drob EM, Liewald DC, Hagenaars
of postnatal hippocampal neurogenesis in rats. J Comp Neurol SP, Davies G, et al. Ageing and brain white matter structure in
1965, 124: 319–335. 3, 513 UK Biobank participants. Nat Commun 2016, 7: 13629.
102. Altman J, Das GD. Autoradiographic and histological studies 122. Bennett IJ, Madden DJ. Disconnected aging: Cerebral white mat-
of postnatal neurogenesis. I. A longitudinal investigation of the ter integrity and age-related differences in cognition. Neurosci-
kinetics, migration and transformation of cells incorporating triti- ence 2014, 276: 187–205.
ated thymidine in neonate rats, with special reference to postnatal

13

314 Neurosci. Bull. February, 2023, 39(2):303–314

123. Chowdhury R, Guitart-Masip M, Lambert C, Dayan P, Huys Q, 141. Stein KC, Morales-Polanco F, van der Lienden J, Rainbolt TK,
Düzel E, et al. Dopamine restores reward prediction errors in old Frydman J. Ageing exacerbates ribosome pausing to disrupt
age. Nat Neurosci 2013, 16: 648–653. cotranslational proteostasis. Nature 2022, 601: 637–642.
124. Yin JA, Liu XJ, Yuan J, Jiang J, Cai SQ. Longevity manipula- 142. Hipp MS, Kasturi P, Hartl FU. The proteostasis network and its
tions differentially affect serotonin/dopamine level and behav- decline in ageing. Nat Rev Mol Cell Biol 2019, 20: 421–435.
ioral deterioration in aging Caenorhabditis elegans. J Neurosci 143. Powers ET, Morimoto RI, Dillin A, Kelly JW, Balch WE. Bio-
2014, 34: 3947–3958. logical and chemical approaches to diseases of proteostasis defi-
125. Zullo JM, Drake D, Aron L, O’Hern P, Dhamne SC, Davidsohn ciency. Annu Rev Biochem 2009, 78: 959–991.
N, et al. Regulation of lifespan by neural excitation and REST. 144. Rodriguez-Oroz MC, Jahanshahi M, Krack P, Litvan I, Macias
Nature 2019, 574: 359–364. R, Bezard E, et al. Initial clinical manifestations of Parkinson’s
126. Apfeld J, Kenyon C. Regulation of lifespan by sensory perception disease: Features and pathophysiological mechanisms. Lancet
in Caenorhabditis elegans. Nature 1999, 402: 804–809. Neurol 2009, 8: 1128–1139.
127. Garden GA, la Spada AR. Intercellular (mis)communication in 145. Stranahan AM, Mattson MP. Recruiting adaptive cellular stress
neurodegenerative disease. Neuron 2012, 73: 886–901. responses for successful brain ageing. Nat Rev Neurosci 2012,
128. Norden DM, Godbout JP. Review: Microglia of the aged brain: 13: 209–216.
Primed to be activated and resistant to regulation. Neuropathol 146. Hetz C, Saxena S. ER stress and the unfolded protein response
Appl Neurobiol 2013, 39: 19–34. in neurodegeneration. Nat Rev Neurol 2017, 13: 477–491.
129. Ownby RL. Neuroinflammation and cognitive aging. Curr Psy- 147. Mattson MP. Late-onset dementia: A mosaic of prototypical
chiatry Rep 2010, 12: 39–45. pathologies modifiable by diet and lifestyle. NPJ Aging Mech
130. Colonna M, Butovsky O. Microglia function in the central nerv- Dis 2015, 1: 15003.
ous system during health and neurodegeneration. Annu Rev 148. Geddes JW, Tekirian TL, Soultanian NS, Ashford JW, Davis DG,
Immunol 2017, 35: 441–468. Markesbery WR. Comparison of neuropathologic criteria for the
131. Minhas PS, Latif-Hernandez A, McReynolds MR, Durairaj AS, diagnosis of Alzheimer’s disease. Neurobiol Aging 1997, 18:
Wang Q, Rubin A, et al. Restoring metabolism of myeloid cells S99–S105.
reverses cognitive decline in ageing. Nature 2021, 590: 122–128. 149. Bennett DA, Schneider JA, Arvanitakis Z, Kelly JF, Aggarwal
132. Jang S, Dilger RN, Johnson RW. Luteolin inhibits microglia and NT, Shah RC, et al. Neuropathology of older persons without
alters hippocampal-dependent spatial working memory in aged cognitive impairment from two community-based studies. Neu-
mice. J Nutr 2010, 140: 1892–1898. rology 2006, 66: 1837–1844.
133. Benfante R, Lascio SD, Cardani S, Fornasari D. Acetylcholinest- 150. Kok E, Haikonen S, Luoto T, Huhtala H, Goebeler S, Haapasalo
erase inhibitors targeting the cholinergic anti-inflammatory path- H, et al. Apolipoprotein E-dependent accumulation of Alzheimer
way: A new therapeutic perspective in aging-related disorders. disease-related lesions begins in middle age. Ann Neurol 2009,
Aging Clin Exp Res 2021, 33: 823–834. 65: 650–657.
134. Li Y, Xie L, Huang T, Zhang Y, Zhou J, Qi B, et al. Aging 151. Wyss-Coray T. Ageing, neurodegeneration and brain rejuvena-
neurovascular unit and potential role of DNA damage and repair tion. Nature 2016, 539: 180–186.
in combating vascular and neurodegenerative disorders. Front 152. Ximerakis M, Lipnick SL, Innes BT, Simmons SK, Adiconis X,
Neurosci 2019, 13: 778. Dionne D, et al. Single-cell transcriptomic profiling of the aging
135. Yousufuddin M, Young N. Aging and ischemic stroke. Aging mouse brain. Nat Neurosci 2019, 22: 1696–1708.
2019, 11: 2542–2544. 153. Ding XL, Lei P. Plasma replacement therapy for Alzheimer’s
136. De Silva TM, Faraci FM. Contributions of aging to cerebral small disease. Neurosci Bull 2020, 36: 89–90.
vessel disease. Annu Rev Physiol 2020, 82: 275–295. 154. Sha SJ, Deutsch GK, Tian L, Richardson K, Coburn M, Gaudioso
137. Carrano A, Hoozemans JJM, van der Vies SM, Rozemuller AJM, JL, et al. Safety, tolerability, and feasibility of young plasma infu-
van Horssen J, de Vries HE. Amyloid Beta induces oxidative sion in the plasma for alzheimer symptom amelioration study: A
stress-mediated blood-brain barrier changes in capillary amyloid randomized clinical trial. JAMA Neurol 2019, 76: 35–40.
angiopathy. Antioxid Redox Signal 2011, 15: 1167–1178. 155. Villeda SA, Plambeck KE, Middeldorp J, Castellano JM, Mosher
138. Chung YC, Kim YS, Bok E, Yune TY, Maeng S, Jin BK. MMP-3 KI, Luo J, et al. Young blood reverses age-related impairments
contributes to nigrostriatal dopaminergic neuronal loss, BBB in cognitive function and synaptic plasticity in mice. Nat Med
damage, and neuroinflammation in an MPTP mouse model of 2014, 20: 659–663.
Parkinson’s disease. Mediators Inflamm 2013, 2013: 370526. 156. Kaur P, Otgonbaatar A, Ramamoorthy A, Chua EHZ, Harmston
139. Jangula A, Murphy EJ. Lipopolysaccharide-induced blood brain N, Gruber J, et al. Combining stem cell rejuvenation and senes-
barrier permeability is enhanced by alpha-synuclein expression. cence targeting to synergistically extend lifespan. bioRxiv 2022,
Neurosci Lett 2013, 551: 23–27. DOI:https://​doi.​org/​10.​1101/​2022.​04.​21.​488994.
140. Dohgu S, Takata F, Matsumoto J, Kimura I, Yamauchi A, 157. King A. The search for better animal models of Alzheimer’s dis-
Kataoka Y. Monomeric α-synuclein induces blood-brain barrier ease. Nature 2018, 559: S13–S15.
dysfunction through activated brain pericytes releasing inflam- 158. Hall AM, Roberson ED. Mouse models of Alzheimer’s disease.
matory mediators in vitro. Microvasc Res 2019, 124: 61–66. Brain Res Bull 2012, 88: 3–12.

13

You might also like