Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

REVIEWS

Ageing as a risk factor for


neurodegenerative disease
Yujun Hou   1, Xiuli Dan1, Mansi Babbar1, Yong Wei1, Steen G. Hasselbalch   2,
Deborah L. Croteau1 and Vilhelm A. Bohr   1,3*
Abstract | Ageing is the primary risk factor for most neurodegenerative diseases, including
Alzheimer disease (AD) and Parkinson disease (PD). One in ten individuals aged ≥65 years has
AD and its prevalence continues to increase with increasing age. Few or no effective treatments
are available for ageing-​related neurodegenerative diseases, which tend to progress in an
irreversible manner and are associated with large socioeconomic and personal costs. This Review
discusses the pathogenesis of AD, PD and other neurodegenerative diseases, and describes their
associations with the nine biological hallmarks of ageing: genomic instability , telomere attrition,
epigenetic alterations, loss of proteostasis, mitochondrial dysfunction, cellular senescence,
deregulated nutrient sensing, stem cell exhaustion and altered intercellular communication.
The central biological mechanisms of ageing and their potential as targets of novel therapies
for neurodegenerative diseases are also discussed, with potential therapies including NAD+
precursors, mitophagy inducers and inhibitors of cellular senescence.

Ageing is associated with physical deterioration that manifestation of accelerated ageing. Conversely, ageing
leads to an increased risk of disease and death1. Ageing is a major risk factor for neurodegeneration4. The most
occurs at different rates in different species, and inter-​ common neurodegenerative diseases, Alzheimer disease
individual variations exist within a species and in the (AD) and Parkinson disease (PD), are predominantly
different tissues of an individual2. Potential biomark- observed in elderly individuals, and the risk of these
ers of ageing align with the central molecular mech- diseases increases with age (Fig. 1). Molecular studies
anisms of ageing, and studies have identified nine have revealed that brain tissue from older individuals
critical hallmarks of the ageing process, as we discuss contains abnormal deposits of aggregated proteins such
in this Review2. Among the many risk factors for neuro­ as hyperphosphorylated tau (p-​tau), amyloid-​β (Aβ)
degeneration, the ageing process itself has by far the most and α-​synuclein; however, it remains unclear whether
impact. Thus, in order to develop successful interven- the levels of these deposits are linked with the degree
tions, it is important to consider the basic mechanisms of cognitive impairment5. Some studies have indicated
of ageing and their role in the onset and progression of that the risk of neurodegenerative disease is associated
neurodegenerative disease. with early developmental defects, showing that brain
The US population aged ≥65 years is estimated to structural changes might take place much earlier than
increase from 53 million in 2018 to 88 million in 2050 cognitive impairment6. MRI measurements of white
(ref.3). As this elderly population increases, the financial matter myelin water fraction and grey matter volume in
burden of age-​related health disorders will increase, and various brain regions of infants who were carriers of the
1
Laboratory of Molecular
Gerontology, National
effective preventive and/or therapeutic approaches are apolipoprotein E (APOE) ε4 allele, a major suscepti­bility
Institute on Aging, NIH, urgently needed. Among the different age-​related dis- gene for sporadic (late-​onset) AD, were significantly diff­
Baltimore, MD, USA. eases, neurodegeneration and the associated cognitive erent from measurements in non-​carriers6. Exposure
2
Danish Dementia Research decline is particularly relevant owing to its great influence to adverse environmental stimuli — such as trauma,
Centre, Rigshospitalet, on healthspan and quality of life. drugs or environmental toxins — during development
University of Copenhagen,
Given that in the elderly population, neurodegen- has been proposed to have consequences in later life
Copenhagen, Denmark.
erative diseases are common and disease-​free brains (for example, by affecting neuroplasticity)7.
3
Center for Healthy Aging,
University of Copenhagen,
are rare, especially in very old individuals, brain age- Common age-​related neurodegenerative diseases and
Copenhagen, Denmark. ing might form a continuum with neurodegeneration. their estimated prevalences, as reported in the primary
*e-​mail: vbohr@nih.gov Human genetic and environmental factors determine the literature, are shown in Table 1. The prevalence of AD
https://doi.org/10.1038/ progression of neurodegenerative disease4. It is tempt- in individuals aged ≥95 years in the USA is ~50%3,8,9
s41582-019-0244-7 ing, therefore, to regard neurodegenerative diseases as a (Fig. 1a). Globally, PD prevalence increases steadily with

Nature Reviews | Neurology


Reviews

Key points neurodegenerative processes. Limiting the progression


of neuronal ageing through diet and exercise might be
• Ageing is the main risk factor for most neurodegenerative diseases, including possible14. Ageing neurons frequently show evidence of
Alzheimer disease (AD) and Parkinson disease (PD). mitochondrial damage and dysfunction, both of which
• Tissues composed primarily of postmitotic cells, such as the brain, are especially are associated with neurodegenerative disease and with
sensitive to the effects of ageing. normal ageing. This Review outlines aspects of our cur-
• Hallmarks of ageing — genomic instability, telomere attrition, epigenetic alterations, rent understanding of the molecular and cellular basis
loss of proteostasis, mitochondrial dysfunction, cellular senescence, deregulated of human ageing, with a focus on neurodegenerative
nutrient sensing, stem cell exhaustion and altered intercellular communication — disease.
correlate with susceptibility to neurodegenerative disease.
• NAD+ deficiency is a key biomarker for mitochondrial dysfunction, and agents that Primary hallmarks of ageing
elevate intracellular NAD+ have shown promising results against many features of
Genomic instability and DNA damage. Several types
neurodegeneration.
of DNA damage, including bulky adducts, abasic sites,
• Genomic instability, mitophagy, cellular senescence, protein aggregation and
DNA single-​strand breaks, DNA double-​strand breaks,
inflammation are being explored as therapeutic targets for neurodegenerative
disease.
base mismatches, insertions and deletions, are associated
with neurodegeneration16. Oxidative DNA damage from
endogenous reactive oxygen species (ROS) can increase
age8,10 (Fig. 1b), and according to data from 2014, the inflammation, accelerate ageing, and increase suscepti-
prevalence of amyotrophic lateral sclerosis (ALS) in bility to cancer and neurodegenerative diseases17. The
the USA increases with age up to 80 years11 (Fig. 1c). This five key DNA repair pathways are base excision repair
Review synthesizes current knowledge about the bio- (BER), nucleotide excision repair (NER), mismatch
logical processes linked with normal brain ageing and repair, DNA double-​strand break repair (DSBR) and
neurodegeneration and discusses promising therapeu- direct reversal16,18.
tic avenues. The main focus is on the two most preva- BER is the primary mechanism for the repair of small
lent neurodegenerative diseases, AD and PD, but other DNA base modifications, such as oxidative base damage
neuro­degenerative diseases that show important links and single-​strand breaks19. Mutations in BER pathway
with ageing hallmarks are also discussed12. genes cause a mutator phenotype, potentially increas-
ing the risk of neurodegeneration and ageing20,21. In the
The biology of ageing first step in BER, a DNA glycosylase recognizes and
Ageing is an inevitable and irreversible process, associ- removes the damaged DNA base, leaving an abasic site.
ated with readily identifiable and characteristic changes BER proceeds via one of two BER subpathways, known
in physical appearance and function in the individual as short-​patch and long-​patch BER. The repair process
organism. Studies have defined molecular and cellular involves the following core steps: excision of the base,
processes and biomarkers that are associated with ageing incision, end processing and repair synthesis, including
in diverse mammalian species and might be linked with gap filling and ligation22. In principle, BER can be recons­-
basic biological mechanisms underlying human ageing. tituted in vitro using four proteins: a DNA glycosylase,
López-​Otin et al.13 identified nine so-​called ‘hallmarks AP endonuclease 1 (APE1), DNA polymerase β (Polβ)
of ageing’ and categorized them into primary, anta­ and DNA ligase III (Lig III)22.
gonistic and integrative hallmarks. These hallmarks are DNA damage not only generates genomic instabil-
widely used in the field of ageing research and beyond. ity, but also initiates signalling cascades that permeate
The primary hallmarks are genomic instability, telomere throughout the cell. Persistent DNA damage induces
attrition, epigenetic alterations and loss of proteostasis. PARP1, increases PARylation (the formation of PAR
Genomic instability is considered to be a major driving polymers at sites of DNA damage or DNA alterations),
force of ageing and is the central focus of one of the dom- and depletes NAD+. NAD+ is an essential cofactor for
inant theories of ageing14. The antagonistic hallmarks sirtuins, DNA repair, mitophagy and mitochondrial
— mitochondrial dysfunction, cellular senescence and health. Sirtuin 3, sirtuin 1, and sirtuins 1 and 6 pro-
deregulated nutrient sensing — are compensatory or mote efficient BER, NER and DSBR, respectively23.
antagonistic responses to the primary damage. Initially, DNA damage promotes cellular senescence and inflam-
these responses mitigate the damage, but eventually mation, which together exacerbate ageing-​related
they can become deleterious themselves. The integrative neurodegeneration (Fig. 3).
hallmarks — stem cell exhaustion and altered intercel-
lular communication — arise as a result of cumulative Telomere attrition. Telomeres are regions composed of
damage induced by the primary and antagonistic hall- DNA and protein at the ends of the linear chromosomes.
marks and are ultimately responsible for the functional Telomeres become shorter as cells divide, unless either
decline associated with ageing (Fig. 2). the parental cell expresses telomerase or another mecha­
Tissues composed primarily of postmitotic cells, such nism is present to prevent telomere attrition. Telomere
as the brain, which contains postmitotic neurons and shortening causes cellular senescence and is probably
oligodendrocytes, are especially sensitive to the effects of involved in organismal ageing. Defects in telomere
ageing, primarily because postmitotic cells are believed maintenance accelerate ageing in mice and humans13.
to be more vulnerable to DNA damage than proliferat- Further clarification of telomere-​related pathogenesis in
ing cells15. DNA damage increases with ageing, which neurodegenerative diseases is greatly needed, as has been
might be important in the aetiology of ageing-​associated discussed elsewhere24,25.

www.nature.com/nrneurol
Reviews

a Alzheimer disease b Parkinson disease c Amyotrophic lateral sclerosis


600 2,500 25

Cases per 100,000 population

Cases per 100,000 population


Cases per 1,000 population
500 2,000 20
400
1,500 15
300
Women 1,000 10
200 Men
Men Women
100 500 5

0 0 0
9

9
9

0
0
–6

–7

–7

–8

–8

–9

≥9

–3

–4

–5

–6

–7
–4

–5

–6

–7

≥8
≥8
65

70

75

80

85

90

18

40

50

60

70
40

50

60

70
Age (years) Age (years) Age (years)

Fig. 1 | Neurodegenerative disease prevalence. a | Prevalence of Alzheimer disease per 1,000 men and women by age in
the USA3,8,9. b | Prevalence of Parkinson disease per 100,000 men and women by age globally8,10. c | Prevalence of amyotrophic
lateral sclerosis (ALS) per 100,000 population in the USA in 2014 (ref.11).

Epigenetic alterations. Epigenetic modifications includ- fission, removal of damaged proteins by proteases, and
ing methylation, PARylation and acetylation of DNA mitophagy. Among these mechanisms, mitophagy has
and histones collectively influence chromatin tertiary received particular attention in recent years.
structure. Epigenetic markers strongly influence chro- Mitophagy is a form of autophagy that selectively
matin activity and function, including transcription and degrades dysfunctional mitochondria and has a key
replication. Epigenetic age-​related mechanisms have role in preventing age-​related disease. One mitophagy
roles in pathologies associated with neurodegenerative pathway is the PTEN-​induced putative kinase protein 1
diseases26. The emerging field of epigenetics in neuro- (PINK1)–Parkin pathway, whereby PINK1 accumu-
degeneration and neuroprotection has been reviewed lates on the outer membrane of depolarized mito-
elsewhere27. chondria and recruits the E3 ubiquitin ligase Parkin,
which ubiquitinates and targets mitochondrial proteins
Loss of proteostasis. The balance between protein syn- and promotes degradation of damaged mitochondria.
thesis and degradation creates a steady state known as Autophagosomes fuse with lysosomes to form auto­
proteostasis. The maintenance of proteostasis in eukary­ lysosomes where the enveloped proteins and/or dam-
otic cells depends on proper regulation of the protea- aged organelles or organelle fragments are degraded.
some and on the process of autophagy13, as well as on Mutations in the genes encoding PINK1 or Parkin
the ubiquitination machinery and lysosomal system. lead to locomotor deficits in Drosophila melanogaster31
Ubiquitinated proteins are often destined for degrada- and early onset of recessive PD in humans32. A 2018
tion. Autophagy is an intracellular degradation system study suggested that the activation of mitophagy to
that facilitates lysosomal degradation of misfolded or clear damaged mitochondria is critical not only for
unfolded proteins and of damaged organelles, thereby mitochondrial quality control but also for mitigating
reducing secretion of inflammatory cytokines28. Increa­ inflammation, an important risk factor for neurodegene­
sed protein misfolding, aggregation and deposition are ration33. Mitophagy can also take place in a manner that
observed in many neurodegenerative disorders. is independent of PINK1 and Parkin through the regu­
lation of FUNDC1, NIX/BNIP3, AMBRA1, BCL-2-L-13
Antagonistic hallmarks of ageing or MUL1 (ref.34). Genetic and pharmacological induc-
Mitochondrial dysfunction and mitophagy. As highly tion of the mitochondrial receptor NIX restored mito-
metabolically active cells, neurons have high energy phagy in cell lines from patients with PINK1-related or
demands to perform neuronal activities and are particu- Parkin-​related PD35. AMBRA1-mediated mitophagy
larly sensitive to changes in mitochondrial function. The has a promising neuroprotective role by limiting ROS-​
production of ROS is amplified in damaged mitochon- induced dopaminergic cell death36. In D. melanogaster,
dria and is implicated in the normal ageing process and overexpression of MUL1 can attenuate PINK1 and
in a majority of known neurodegenerative diseases29. Parkin mutant phenotypes in dopaminergic neurons
Besides ATP production, mitochondria also have key and might compensate for the loss of these proteins37.
roles in several intercellular pathways, including lipid Growing evidence suggests that defects in mitophagy
biosynthesis, calcium signalling and cell apoptosis, all contribute to neurodegeneration34. As such, mitophagy-​
of which are central processes in the development of stimulating agents are being explored for potential
neuro­degenerative disease30. Brain mitochondrial func- therapeutic benefit in animal models of several human
tion becomes impaired with age and is believed to be a neurological diseases, as discussed below.
major and early contributor to the ageing process. Studies have indicated that nuclear DNA damage
Several quality control mechanisms are involved in can lead to mitochondrial dysfunction, suggesting the
maintaining optimal mitochondrial function, including existence of nucleus–mitochondria ‘crosstalk’ signal-
proteasome degradation, export of damaged proteins in ling38 in DNA damage responses (DDRs) (Fig. 4). The
mitochondria-​derived vesicles, mitochondrial fusion and mitochondrial unfolded protein response (UPR) is a

Nature Reviews | Neurology


Reviews

Table 1 | Age-​related neurodegenerative diseases


Disease Prevalence Major symptoms risk factors Neuropathological
hallmarks
Alzheimer 5.7 million in the USA in Impairment of learning and Age, family history , genetics, history of Aβ plaques, neurofibrillary
disease 2018 (ref.3) memory , speech difficulties head trauma, female gender, vascular tangles, neuronal loss,
risk factors, environmental factors195–198 neuroinflammation
Parkinson 2–3% of the global Muscle rigidity , tremors, Environmental factors, genetics, male α-​Synuclein-containing
disease population aged alterations in speech and gait gender, ethnicity , age, psychiatric Lewy bodies and loss of
>65 years in 2017 (ref.10) symptoms199,200 dopaminergic neurons,
grey matter atrophy201
Amyotrophic 0.6 cases per million Progressive motor defects, with Physical activity , familial aggregation, TAR DNA-​binding protein
lateral sclerosis globally in 2016 (ref.202) muscle weakness, atrophy and environmental and occupational 43 aggregation
spasms exposure (for example, to pesticides,
solvents or heavy metals), smoking,
head injury , genetics202
Huntington 5–7 per 100,000 white Chorea, dystonia, loss of Genetic mutation in HTT, inheritance Striatal atrophy ,
disease people in 2007 (ref.203) coordination, cognitive decline, neuronal loss, psychiatric
behavioural difficulties symptoms204,205
Dementia with 1.3 million in the USA in Visual hallucinations, movement Age >50 years, male gender, family Lewy bodies and Lewy
Lewy bodies 2014 (ref.206) disorders, cognitive problems, history206 neurites
sleep difficulties, depression
Ataxia From 1 in 40,000 to 1 Cerebellar degeneration, Genetics (mutations in ATM gene) Ataxia and telangiectasias
telangiectasia in 100,000 live births immunodeficiency , radiation
worldwide in 2016 sensitivity , diabetes, cancer
(ref.207) predisposition
Cockayne 2–3 per million globally in Growth failure, neurological Genetics (mutations in CSA or CSB Growth retardation and
syndrome 2008 (ref.208) disorders, photosensitivity , eye gene) neurodegeneration
disorders, premature ageing

mitochondria-​to-nucleus signal transduction path- Cells with high levels of DNA damage become senes-
way. Mitochondrial stress results in dysregulated UPR, cent and stop proliferating. Owing to the high energy
which leads to neurodegeneration. Research into mecha­ demand and increased abundance of ROS in neurons,
nisms that facilitate communication between nuclear brain tissue can accumulate high levels of DNA damage,
and mitochondrial compartments will improve our which increases with age and age-​associated loss of DNA
understanding of ageing and neurodegeneration. repair capacity15,46. As postmitotic cells, neurons are par-
ticularly vulnerable to the accumulation of DNA lesions,
Cellular senescence. Cellular senescence, first identified and for DNA repair they primarily depend on non­
in fibroblasts by Hayflick in 1965 (ref.39), is a state of homologous end joining, an error-​prone pathway, rather
stress-​induced stable cell cycle arrest and a senescence-​ than homologous recombination, the other process that
associated secretory phenotype (SASP) that occurs with repairs DNA double-​strand breaks. Re-​entry into the cell
age. Cellular senescence can be considered a response cycle by neurons is commonly seen in pathological con-
that aims to maintain survival of healthy cells and remove ditions such as AD, and is followed by neuronal hyper-
damaged cells under conditions of stress40. Several senes- ploidy and synaptic failure47. Neurons with a persistently
cence pathways have been described, including stress-​ activated DDR pathway exhibit many features of cellular
induced premature senescence, replicative senescence, senescence and are referred to as senescence-​like neu-
oncogene-​induced senescence, and mitochondrial rons48. In a study in old C57BL/6 mice, which showed
dysfunction-​associated senescence41. Cellular senes- that DNA damage induces a senescence-​like state in
cence is initially a mechanism of tumour suppression mature postmitotic neurons in vivo, 40–80% of Purkinje
but might become a tumour initiation mechanism, cell neurons and 20–40% of cortical, hippocampal and
perhaps as a result of ageing42. SASP emerges when peripheral neurons had high levels of DNA damage, acti-
damaged or dying cells acquire pro-​inflammatory pro­ vated p38 MAP kinase, oxidative stress and senescence-​
perties that might promote tumour progression via the associated β-​galactosidase activity49. The researchers
expression of cytokines (such as IL-6 and IL-1), growth found that p21 serves as an important signal transducer
factors, chemokines and proteases43. Cells that undergo between the DDR and the senescence-​like phenotype in
mitochondrial dysfunction-​associated senescence have neurons, similar to its role in proliferation-​competent
reduced NAD+:NADH ratios, which promotes growth cells49. Another study suggested not only that the effi-
arrest and prevents IL-1-associated SASP formation44. ciency of DNA repair decreases with age, but also that
A 2019 study revealed that NAD+ metabolism is also more complex DNA repair mechanisms are used during
involved in the regulation of SASP through the high ageing, leading to more mutations being introduced50.
mobility group A (HMGA)–nicotinamide phosphoribo- These less efficient DNA repair processes are associated
syltransferase (NAMPT)–NAD+ signalling axis and that with ageing and cancer and might also have a role in
NAD+ metabolism governs the pro-​inflammatory SASP45. neurodegeneration50.

www.nature.com/nrneurol
Reviews

Genomic
instability
AD PD HD ALS AT
Telomere Epigenetic
attrition alterations
AD PD HD ALS AD PD HD

Loss of Altered intercellular


proteostasis Ageing hallmarks communication
AD PD and AD PD HD ALS
neurodegeneration

Deregulated Stem cell


nutrient sensing exhaustion
AD PD HD AD PD HD

Mitochondrial Cellular
dysfunction senescence
AD PD AT AD PD HD ALS

Fig. 2 | Hallmarks of ageing. Nine hallmarks of ageing — genomic instability , telomere attrition, epigenetic alterations,
mitochondrial dysfunction, deregulated nutrient sensing, loss of proteostasis, cellular senescence, stem cell exhaustion
and altered intercellular communication — seen in the main neurodegenerative diseases. AD, Alzheimer disease;
ALS, amyotrophic lateral sclerosis; AT, ataxia telangiectasia; HD, Huntington disease; PD, Parkinson disease.

Autophagy is another essential process that is linked dysfunction55. As cellular senescence exacerbates ageing
with cellular senescence51, but whether autophagy pro- and age-​related brain dysfunction, targeting of senescent
motes or inhibits senescence is still under debate. On the cells might be useful in patients with neurodegenerative
one hand, macro-​autophagy might help establish SASP diseases, as discussed later in this article.
by facilitating the synthesis of secretory proteins51, but
on the other hand, inhibition of autophagy might further Deregulated nutrient sensing and altered metabolism.
promote senescence under some circumstances, such as Calorie restriction, which downregulates nutrient sig-
through ROS and p53 (ref.52,53). GATA4 has been reported nalling pathways, extends lifespan in several species
to be activated in response to DNA damage in a manner including mice, and might have neuroprotective effects
that depends on the kinases ATM and ATR, and has vital in human tissues56. Major nutrient-​sensing pathways and
roles in triggering SASP and senescence by activating the molecules include insulin, insulin-​like growth factor 1
transcription factor nuclear factor κB (NF-​κB). In cells in (IGF1), mechanistic target of rapamycin (mTOR), AMP-​
which senescence had been induced by DNA damage, activated protein kinase (AMPK) and sirtuins13. mTOR,
GATA4 protein abundance was found to be increased AMPK and sirtuins are being explored as therapeutic tar-
owing to improved protein stability during senescence54. gets for neurodegenerative diseases. Metabolic dysfunc-
Degradation of GATA4 depends on selective autophagy tion is frequently observed in patients with neurological
and is suppressed during senescence. An age-​related disease and might correlate with low NAD+ abundance,
decline in autophagy is observed in the brain. However, mitochondrial dysfunction and oxidative stress57.
the mechanism connecting reduced autophagy and
senescence in the brain needs to be elucidated. Processes Integrative hallmarks of ageing
linked with cellular senescence that might be involved Stem cell exhaustion. Functional stem cells are needed
include telomere attrition, inflammation, autophagy defi- for optimal health in later life. However, stem cell func-
ciency, chronic DDR, oxidative stress and mitochondrial tion and proliferative capacity decline over an organism’s

Nature Reviews | Neurology


Reviews

Stress

DNA repair DNA damage


DNA damage responses
BER
NER
DSBR PARP1
PARylation
SIRT3
SIRT1
SIRT1 and SIRT6
Inflammation
Mitophagy TNF
Mitochondrial Senescence
dysfunction p16 IL-1β
NAD+
p21

Sirtuins

Ageing and neurodegeneration

Fig. 3 | NAD+, DNA damage, mitophagy , ageing and neurodegeneration pathways. Stresses such as radiation and
viral infections have been reported to induce DNA damage. Cellular senescence and inflammation are the main DNA
damage responses. Senescent cells often show increased expression of p16, p21 and other factors. Activated microglia
and astrocytes release many pro-​inflammatory factors such as tumour necrosis factor (TNF) and IL-1β. In addition to
leading to DNA damage, such stresses lead to mitochondrial dysfunction, which induces ageing and neurodegeneration.
Accumulation of DNA damage might be particularly prevalent in the CNS because of high susceptibility to oxidative
stress and low DNA repair capacity in postmitotic brain tissue. Accumulating evidence suggests that deficiencies in the
base excision repair (BER), nucleotide excision repair (NER) and DNA double-​strand break repair (DSBR) pathways
induce ageing-​associated neurodegeneration. NAD+ is an essential factor that is required for cellular function and energy
metabolism. NAD+ has been shown to have many functions in vivo and in vitro, including induction of mitophagy , promotion
of DNA repair via sirtuins (SIRTs), and inhibition of PARylation, senescence and inflammation. Interestingly , increased
mitophagy activity promotes DNA repair and degradation of dysfunctional mitochondria; increased NAD+ levels suppress
these pathologies and could have beneficial effects in ageing and age-​related neurodegenerative diseases.

lifespan. This functional loss can be caused by age-​ adaptive immune responses in neurodegenerative dis-
related high levels of DNA damage, low DNA repair eases is important as such knowledge will be crucial to
capacity, defects in proteostasis, epigenetic deregula- developing effective immune-​based interventions60. New
tion, mitochondrial dysfunction, telomerase inactivation evidence suggests that the pathological mechanisms of
and/or cell senescence58. Stem cell function is improved neurodegenerative diseases might involve interactions
by increased expression of heat shock protein HSP70 or of microglia — the innate resident immune cells in the
forkhead transcription factor FOXO4, and by treatment CNS — with other glial support cells, such as astrocytes
with rapamycin58. In aged animals, exposure to young and oligodendrocytes60. Regulatory T cells are important
blood through heterochronic parabiosis improves stem not only for maintaining peripheral immune balance but
cell function in muscle, liver, spinal cord and brain, and also for the tolerance and immune privilege of the CNS
counteracts ageing and rejuvenates cognitive processes59. itself61. Mast cells, microglia, T cells, oligodendrocytes
Therefore, targeting of aged stem cell regeneration might and the crosstalk between these cells are essential in
alleviate age-​associated neurodegenerative diseases. the function of the immune system and in preventing
neuro­degenerative disease60,62. The link between changes
Altered intercellular communication and immune func- in immune response with age and increased incidence
tion. Alterations in levels of hormones such as leptin, of neurodegenerative diseases in the ageing population
ghrelin, insulin, adiponectin and IGF1 regulate neuronal might provide important insights into this interaction.
damage and neurodegeneration. The immune system is Inflammation, a fundamental protective event
essential for shaping the brain during development, and in response to any insult, becomes upregulated with
both the nervous system and the immune system change advanced age and can be divided into five categories:
with age, so the loss of regulation of immune responses low-​grade, controlled, asymptomatic, chronic and sys-
in the brain is likely to be a factor in neurodegenera- temic states63. Low levels of inflammation can have bene-
tion60. Improved understanding of the role of innate and ficial effects, but uncontrolled or sustained inflammation

www.nature.com/nrneurol
Reviews

can be harmful and result in age-​related chronic neuro- resulting in an ageing-​associated chronic inflamma-
degenerative diseases such as AD and PD64. Gene expres- tory response68. Aggregated Aβ fibrils in AD69, mutant
sion studies in ageing human brains have emphasized α-​synuclein in PD70, mutant huntingtin in Huntington
the importance of inflammation in neurodegenerative disease (HD)71 and the superoxide dismutase 1 (SOD1)
diseases65. mutant protein SOD1G93A in familial ALS72,73 are capa-
Chronic inflammation, persistent activation of ble of inducing secretion of IL-1β via NLRP3 inflam-
microglia, sustained elevation of pro-​inflammatory masome activation. Inflammation also exacerbates Aβ
mediators and increased oxidative stress are associated deposition in AD, and both α-​synuclein truncation and
with age-​related neurodegenerative diseases64–66. In aggregation in PD, and might induce protein aggrega-
these conditions, hallmarks of chronic inflammation are tion in HD and ALS74,75. In this way, a positive feedback
observed, including the persistent activation of microglia loop can occur between the enhanced inflammation and
and subsequent sustained release of pro-​inflammatory accumulation of disease-​specific misfolded proteins in
mediators, and also increased oxidative and nitrosa- neurodegenerative diseases.
tive stress. Activated microglia elicit ROS, nitric oxide Other factors reported to contribute to the inflam-
and pro-​inflammatory cytokines such as IL-1β, IL-6 and matory response and pathogenesis of neurodegenera-
tumour necrosis factor (TNF), leading to neuroinflam- tive disorders include age-​related sex steroid hormone
mation67. The production of ROS from dysfunctional deficiency76, defective proteasome and autophagy degra-
mitochondria and increased NF-​κB signalling that occur dation systems77,78, cellular senescence79, increased oxida-
with increased age potentiate NLRP3 inflammasomes tive DNA damage and impaired DNA repair80, decreased
and subsequently lead to release of IL-1β in the brain, innate and adaptive immune system responses81, and
environmental stressors82. Targeting of these neuro­
inflammatory processes might be beneficial in age-​related
neurodegenerative diseases. Anti-​inflammatory drugs
NAD+ depletion, are already an important therapeutic strategy in age-​
Sirtuin dysfunction related neurodegenerative disease, but a further chal-
Mitophagy lenge is to develop drugs that can cross the blood–brain
defects
barrier and have fewer adverse effects than current
strategies. Immunotherapies have shown promise in
Apoptosis reducing inflammation in transgenic mouse models of
neurodegenerative disease83,84. Non-​pharmacological
DNA
Nuclear
interventions, including calorie restriction and phys-
fragmentation ical activity, have also been shown to exert an anti-​
gene
mutations inflammatory effect85,86. An improved understanding
of the relationship between the inflammatory process
Mitochondrial and age-​related neurodegenerative diseases is needed for
complex
dysfunction future potential interventions.

Neurodegeneration Ageing and neurodegenerative disease


Alzheimer disease
AD is the most common neurodegenerative disease.
Impaired The main clinical features of AD are late-​life memory
mitochondrial
biogenesis and learning deficits, disorientation, mood swings and
behavioural issues. Defects in at least three proteins,
Inactivation Mitochondrial
the amyloid precursor protein (APP), presenilin 1 and
Dysregulation
of transcription of UPR stress presenilin 2, have been identified as risk factors for AD87.
factors Mutations in the genes that encode these proteins are
linked with familial (early-​onset) AD. Sporadic (late-​
onset) AD occurs with much higher (and increasing)
Fig. 4 | Nuclear–mitochondrial signalling in neurodegenerative disease. NAD+ has prevalence and is thought to reflect complex interac-
a pivotal role in cellular homeostasis and function. NAD+ levels decrease with age and tions between genetic and environmental factors. The
are reduced in age-​related diseases. Sirtuins are NAD+-dependent deacetylases that APOE*ε4 allele has been identified as the most common
consume NAD+. Sirtuin deficiency impairs mitophagy in many diseases, including genetic risk factor for sporadic AD88.
Alzheimer disease. Mutations in some nuclear genes, especially those that encode Prominent molecular features of AD are Aβ plaques
mitochondrial proteins, destroy nuclear–mitochondrial interactions and lead to and p-​tau neurofibrillary tangles (NFTs) in the brain89.
mitochondrial dysfunction. The mitochondrial unfolded protein response (UPR) is a Aβ is a short abnormal proteolytic fragment (36–43
mitochondria-​to-nucleus signal transduction pathway resulting in a series of protective amino acids of APP) produced by sequential cleavage
events for mitochondria. Mitochondrial stress results in dysregulated UPR , which leads
of APP by β-​secretase and γ-​secretase90. The primary
to neurodegeneration. Notably , mitochondrial biogenesis is controlled mainly from the
nucleus, as most proteins involved are encoded by nuclear genes. Fragmentation of causal factors leading to AD are still a matter of intense
genomic DNA is an initial hallmark of apoptosis and a number of the crucial events in debate, but might include cholinergic dysfunction, Aβ
apoptosis commence in the mitochondria. The crosstalk between the nucleus and plaques, tau aggregation, inflammation, DNA damage
mitochondrial dysfunction might have a strong effect on neuronal function, tipping and mitochondrial dysfunction. Acetylcholinesterase
the balance from neuronal health to neuropathology and neurodegenerative diseases. inhibitors (donepezil, galantamine and rivastigmine) are

Nature Reviews | Neurology


Reviews

approved by the FDA for the management of AD, but learning, memory and behaviour, are regulated by
only alleviate some symptoms and have limited ability epigenetic mechanisms13,105, and both epigenetic and
to prevent disease progression91. environmental factors are thought to influence suscep-
The Aβ hypothesis proposes that Aβ accumulation tibility to sporadic AD. The epigenetic modifications
and toxicity is the triggering factor in AD. Aβ forms include methylation, PARylation and acetylation of
oligomers and fibrils that accumulate in the brain to DNA and histones, all of which have important roles
form Aβ plaques, resulting in neuronal loss92. Many in AD. For example, regions of the human and primate
potential anti-​Aβ drugs are under development, but APP promoters are differentially methylated in tissue
many have failed in clinical trials. P-​tau accumulates in and brain regions in a manner that correlates with
neurons in AD and causes neuronal dysfunction and is APP expression, and specific patterns of DNA methyl-
therefore considered to be another potential cause of ation and hydroxymethylation (5-methylcytosine and
AD89,93. The neuroinflammatory response — a brain 5-hydroxy­methylcytosine) are associated with features
immune response caused by internal or external stress, of AD26. Hyperactivated PARP1 can result in PARylation
excessive oxidation and accumulation of Aβ plaques or and might contribute to AD pathology106,107. Alterations
p-​tau — is a key driver of AD94 and results mainly from in the level of histones and their modification patterns
abnormal activation of astrocytes and microglia, which have been suggested to be involved in AD pathogenesis.
release pro-​inflammatory cytokines. Mitochondrial dys- Aberrant localization of phosphorylated histone H3 in
function and DNA damage might also be risk factors the cytoplasm, hyperphosphorylation of histone H3,
for AD. The clinical failure of therapeutic approaches and decreased levels of acetylated histone H4 in the hip-
based on the Aβ hypothesis suggests that other strategies pocampus in patients with AD have been reported108.
and/or novel targets are needed as the basis of successful Levels of histone deacetylase 2 (HDAC2) and HDAC6
interventions in AD95. have been shown to be significantly increased in patients
Various model systems have been used to study AD, with AD, but might be counterbalanced by HDAC inhib-
including mouse, rat, Caenorhabditis elegans, D. melano­- itors, as indicated in experimental models of AD and
gaster and human induced pluripotent stem cells other neurodegenerative diseases108. Ubiquitination
(iPSCs)96–101 (Table  2). Transgenic mouse models of of tau proteins is also observed in cells from patients
AD express variants of APP, presenilin 1, presenilin 2, with AD28. Studies have shown that protein aggrega-
tau, APOE or Aβ, or combinations of these and other tion occurs when soluble protein monomers form high
mutations (for example, APPswe/PS1ΔE9, 3×TgAD molecular weight oligomers, polymers and eventually
or 5×FAD). C. elegans and D. melanogaster models of very large insoluble fibrils (for example, Aβ aggregates
AD include knockout or overexpression of C. elegans in patients with AD)109. These protein aggregates impair
or D. melanogaster genes, or cross-​species expression of neuronal activity but the molecular mechanisms that
human transgenes (Table 2). underlie this effect are not known.
Evidence suggests that increased DNA damage and Emerging evidence suggests that mitophagy is com-
decreased DNA repair exacerbate AD progression102. promised in AD, resulting in accumulation of dys-
BER protein expression is specific to tissue type, cell functional mitochondria110. Studies have shown that
type and cell age, and might decrease with disease pro- mitophagy mitigates inflammation, thereby decreasing
gression in patients with AD. For example, levels of the risk of AD, PD and other neurodegenerative dis-
expression of uracil DNA glycosylase, β-​OGG1 glyco- eases33. A cross-​species analysis, including C. elegans and
sylase and Polβ are lower in post-​mortem brain tissue mouse models of AD and human iPSC-​derived human
from patients with AD than in brain tissue from age-​ AD neurons, showed that mitophagy is defective in AD,
matched controls without AD103. Null alleles of Polβ are and that stimulation of mitophagy reverses memory loss
lethal during embryonic development and are associated in AD nematodes and mice111.
with neurodevelopmental defects in mice104. A new AD Another ageing hallmark, cellular senescence,
mouse model was presented in 2015 (ref.104). The com- increases susceptibility to AD, PD and other neuro­
monly used 3×TgAD mouse was crossed with a mouse degenerative diseases112–115 (Fig. 5). Deposition of Aβ
that had defective DNA repair owing to heterozygosity fibrils in AD is linked with increased expression of
for the gene encoding Polβ104. The authors showed that p16INK4a and SASP-​associated factors112. Increased abun-
the resulting 3×TgAD/Polβ+/− heterozygote displayed dance of senescent astrocytes, microglia and neurons,
increased synaptic and cognitive deficits, including and expression of senescence-​associated β-​galactosidase
neuronal dysfunction and cell death, compared with activity (SA-​β-gal) is observed in brain tissue from AD
3×TgAD mice104, suggesting that DNA damage has an patients116. Studies have shown that the senescence-​like
important role in AD progression. neurons and senescent astrocyte, microglia or oligo-
Telomere instability or shortening might also have dendrocyte progenitor cells are sources of oxidative and
a role in AD. Defects in telomere maintenance acceler- inflammatory stress and negatively affect neighbouring
ate ageing in mice and humans13 and, in patients with cells by inducing a similar phenotype117,118. On the basis
AD, are associated with cognitive impairment, amyloid of these findings, the removal of senescent cells from the
pathology and hyperphosphorylation of tau via oxidative brain would be expected to show beneficial effects. This
stress and inflammation24. strategy has shown some success in AD research117,118.
As mentioned earlier, epigenetic mechanisms con- Zhang et al. reported that the exposure of aggregating
tribute to age-​related disease processes in humans. Aβ to oligodendrocyte progenitor cells in culture can
Diverse phenotypes and biological processes, including induce cell senescence, and that the selective removal of

www.nature.com/nrneurol
Reviews

Table 2 | Models for studying Alzheimer disease


organism Model genetic alterations
Mouse 96
hAPP models PDAPP mouse line: mice express hAPP695/751/770 with Florida mutation (V717F) at the
γ-​cleavage site under the regulatory control of the PDGFβ promoter
J20 mouse line: mice express hAPP695/751/770
TgCRND8 mouse line: mice express hAPP695, containing the Swedish double mutations (K670N/
M671L) at the β-​secretase cleavage site and V717F mutation at the γ-​secretase cleavage site
Tg2576, APP23 and RI.40 mouse lines: mice express hAPP695, hAPP751 and entire human APP
gene, respectively , containing the Swedish double mutations (K670N/M671L) at the β-​secretase
cleavage site
TASD-41 mouse line: mice express hAPP containing the Swedish double mutations (K670N/M671L)
at the β-​secretase cleavage site and London mutation (V717I) at the γ-​secretase cleavage site
Aβ models BRI-​Aβ40 or BRI-​Aβ42 mouse lines: mice express fused Aβ and BRI protein, causing amyloid formation
hAPP/PS1 models APPswe/PS1ΔE9 mouse line: mice express hAPP695 with Swedish mutation (K670N/M671L)
and PSEN1 with ΔE9 mutation
5XFAD mouse line: mice express hAPP with Swedish (K670N/M671L), Florida (I716V), and London
(V717I) mutation, and PSEN1 with M146L and L286V mutations
2xKI mouse line: mice express mAPP with Swedish mutation (K670N/M671L) and humanized Aβ
and express mPS1 with P264L mutation
Models with hTau TAPP mouse line: mice express hAPP695 containing Swedish mutation and human four-​repeat tau
with P301L mutation and without the amino-​terminal sequences
3xTg mouse line: mice express three mutated AD genes: hAPP695 containing Swedish mutation,
human four-​repeat tau with P301L mutation and without the N-​terminal sequences, and PS1 with
M146V mutation
Htau mouse line: mice express the entire human tau gene
Rat hAPP/PS1 rat model101 Rats express APP695 with the K670N/M671L mutations, hAPP minigene with the K670N/M671L
and V717F mutations and human PSEN1 with the M146V mutation
Caenorhabditis Knockout of Worms with deletion of APP family proteins (APP/APLP1/APLP2)
elegans C. elegans genes97
Worms with deletion of α-​secretase gene (ADAM10 or ADAM17)
Worms with deletion of β-​secretase gene (BACE1)
Worms with γ-​secretase complex gene (PSEN1/2, APH1, APH2 or PEN2)
Worms with deletion of tau gene
Overexpression of Worms express human Aβ: APL-1, APL-1 extracellular domain or APL-1 deletion of Go/E1/E2/E1 and
C. elegans genes97 E2 domain in head and tail neurons, ventral cord, hypodermis and supporting cells and vulva muscles
Worms express APL-1 neuronal expression
Worms express proteins γ-​secretase complex sel-12, ttx-3, egl-13, pen-2 and sel-12
Worms express α-​secretase protein adm-4
Expression of human Worms express hAβ1-42, dimer Aβ1-42, Met35Cys Aβ1-42
proteins in C. elegans97
Worms express human γ-​secretase complex proteins hPSEN1, hPSEN2, nicastrin, APH1, PEN2
Worm neurons express human tau variants tau (4R1N), V337M tau (4R1N), P301L tau (4R1N), R406W
tau (4R1N), Tau352 (4R1N), Tau352 pseudo-​hyperphosphorylated, Tau352 pseudo-​hypophosphorylated
Drosophila Expression of human Flies express hAPP, hBACE and D. melanogaster γ-​secretase presenilin (dPsn) containing AD mutations
melanogaster proteins: Aβ toxicity98 N141I, L235P and E280A
Flies express Aβ40/42 peptides fused with D. melanogaster necrotic gene sequence for secretion
Expression of human Flies express wild-​type hTau or hTau containing V337M and R406W mutations
proteins: tau toxicity98
Flies express phosphorylation-​resistant tau variants containing S2A/S11A or S262A mutation
Overexpression of Flies with overexpression of β-​secretase-like protein
D. melanogaster
genes: Aβ toxicity98 Flies with knockdown of ferritin

Silencing of Flies with loss of dtau or partitioning defective-1 (PAR-1)


D. melanogaster
genes98,99 Flies with genetic inhibition of copper-​importers (Ctr1C and Ctr1B) or inhibition of zinc importer
dZip1 to rescue Aβ42 pathogenesis
iPSCs iPSC-​derived iPSC-​derived neurons containing mutation at PSEN1 A246E, PSEN2 N141I, PSEN1 M136I, PSEN1 ΔI4,
neurons100 PSEN1 Y115C, PSEN1 ΔE9, APP duplication, APP V717I
Aβ, amyloid-​β; AD, Alzheimer disease; APP, amyloid precursor protein; hAPP, human APP; iPSC, induced pluripotent stem cell.

Nature Reviews | Neurology


Reviews

pathology, including cognitive deficits122. AMPK signal-


ling regulates tau phosphorylation and Aβ production
Senescent Telomere
microglia shortening and is a potent activator of autophagy, which is down-
↑ AD, PD ↑ AD, PD, HD regulated in AD. However, although AMPK signalling
can delay the onset of AD, it might also increase neuro­
Senescent SA-β-gal nal stress123. Additional studies have shown that brain
astrocytes
↑ AD, PD, cells from AD models have reduced glucose and oxygen
↑ AD, PD HD, ALS metabolic rates124. AD neurons show reduced activity of
Cellular the glucose transporters GLUT1 and GLUT3 and the
senescence
glycolytic enzyme aldolase, causing glucose hypometa­
bolism and altered insulin signalling124, insulin resis­
Senescent DDR
neurons tance, endoplasmic reticulum stress125, frequent type 2
↑ AD, PD,
↑ AD, PD HD, ALS diabetes, impaired fasting glucose and altered lipid
metabolism126. A decrease in hippocampal cholesterol
SASP factors
reduces Aβ formation in hippocampal neurons127.
p16INK4a
↑ AD, PD, Stem cell exhaustion and immune dysfunction are
↑ AD, PD
ALS, HD also linked with AD. Exhaustion of CNS stem cells is
associated with poor cognitive ability, reflecting reduced
neurogenesis in the dentate gyrus, in patients with AD128.
Some evidence supports the therapeutic potential of
NAD+ supplementation in rescuing neurogenesis in AD
SASP inhibitors
Immunotherapeutics mice107. A GLP1 hormone analogue, liraglutide, has been
Apoptosis inducers shown to prevent decline in hippocampal neurogene-
Reactivation modulators sis in an AD mouse model129. Exercise, omega-3 fatty
acid utilization, curcumin and flavanols have also been
Fig. 5 | Cellular senescence and neurodegeneration. reported to improve neurogenesis in AD models128.
Cellular senescence is increased in Alzheimer disease (AD), Altered intercellular communication is observed
Parkinson disease (PD), amyotrophic lateral sclerosis (ALS) in brain tissue from patients with AD125. This finding
and Huntington disease (HD). A number of cellular is consistent with the insulin resistance and increased
senescence markers, including senescent microglia, insulin receptor expression that is seen in brain tissue
senescent astrocytes, senescent neurons, senescence-​ from AD mice126,130,131. Peroxisome proliferator-​activated
associated secretory phenotype (SASP) factors, telomere receptor γ (PPARγ) agonists are well studied for their
shortening, persistently activated DNA damage response,
role in peripheral metabolism, and also have therapeu-
increased SA-​β-gal and increased p16INK4A, have been
reported in age-​related neurodegenerative disease.
tic potential in patients with AD via anti-​inflammatory,
Several therapeutic strategies aim to target these anti-​amyloidogenic and insulin-​sensitizing effects132.
senescence markers. For example, some drugs, including The anti-​diabetic drug metformin also has beneficial
rapamycin, melatonin, resveratrol, metformin and effects on AD-​related pathology133. As mentioned ear-
oestrogen, can suppress the SASP in senescent cells. This lier, immune dysfunction and neuroinflammation are
process does not induce cell death or alter cell numbers. features of AD pathology. Compared with brain tissue
Another strategy is to induce cell death, which includes from age-​matched control individuals, brain tissue from
apoptosis and immunotherapy. Senolytics (drugs that patients with AD shows an increased number of acti-
preferentially kill senescent cells), FOXO4 peptide and vated microglia134. Aggregated Aβ fibrils in AD can
inhibitors of BCL-​W and BCL-​X induce apoptosis in
induce IL-1β secretion via NLRP3 inflammasome acti-
senescent cells. Immunotherapy is another promising
strategy for removing senescent cells from the brain and
vation69. The identification of potential small-​molecule
other tissues, and rejuvenation of senescent cells is compounds that reduce neuroinflammation might be an
currently under investigation as another treatment for effective strategy to combat AD.
age-​related pathologies. DDR , DNA damage responses. Together, these findings suggest that ageing hall-
marks are all closely related to AD, and targeting of
one or more of these hallmarks might have therapeutic
senescent cells from AD mice helps reduce neuroinflam- potential.
mation and Aβ accumulation and improves cognitive
deficits119. Tau pathology can also induce neuronal and Parkinson disease
glial senescence120. Interestingly, treatments that remove PD is characterized by neuronal loss in the substantia
senescent cells in the brain can be neuroprotective, as nigra, causing striatal dopamine deficiency, and by intra-
they can prevent tau-​dependent pathology and cognitive cellular inclusions containing aggregates of α-​synuclein.
decline117,118. Diagnostic features of PD include neuromuscular dys-
Nutritional and metabolic factors might also be risk function that affects movement amplitude and speed,
factors for AD. For example, calorie restriction can rigidity and/or rest tremor. The molecular pathogene-
have neuroprotective effects56, and insulin and IGF1 sis of PD involves multiple pathways and mechanisms,
resistance in the brain is an early and common feature including α-​synuclein proteostasis, oxidative stress,
of AD121. mTOR signalling is upregulated in AD, and mitochondrial function, calcium homeostasis, axonal
mTOR inhibitors, such as rapamycin, can improve AD transport and neuroinflammation10.

www.nature.com/nrneurol
Reviews

Aggregates of α-​synuclein are found in neurons of which encodes a master regulator of the DDR signal-
all patients with PD. Soluble α-​synuclein monomers ini- ling pathway. Ataxia telangiectasia is characterized by
tially form oligomers, and then combine to form small progressive cerebellar degeneration, immunodeficiency,
and eventually large insoluble α-​synuclein fibrils, which radiation sensitivity, diabetes, progressive dysarthria,
are neurotoxic and associated with cytoplasmic inclu- choreoathetosis, cancer predisposition, and features of
sions known as Lewy bodies135. α-​Synuclein aggregation premature ageing146. Cerebellar atrophy, especially the
and mitochondrial dysfunction seem to be synergistic loss of Purkinje cells and granule cells, is thought to
features of PD, and mitochondrial dysfunction has been lead directly to neurodegeneration in this disorder147.
suggested to promote α-​synuclein aggregation in degen- Although this process is not well understood, high
erating neurons of patients with PD136. Reducing the levels of DNA damage and ROS and/or mitochon-
activity of mitochondrial complex 1 (part of the electron drial dysfunction are proposed to have key roles in the
transport chain) and PPARγ co-​activator 1α (PGC1α) pathophysiology of ataxia telangiectasia148,149. Given
leads to mitochondrial dysfunction and oxidative stress the importance of ATM in DDR signalling, the patho-
in PD136,137. Post-​mortem brain imaging and biomarker genesis of ataxia telangiectasia illustrates how accelerated
studies strongly suggest that neuroinflammation is accumulation of DNA damage might result in ageing
prominent in brain tissue from patients with PD138. and neurodegeneration. Thus, this disorder serves as a
DNA repair defects can impair function of the dopa- good model to dissect the mechanisms of ageing and
minergic axis, thus increasing the risk of PD139. Several neurodegeneration.
studies have shown that telomere shortening is also ALS is characterized by deposition of TAR DNA-​
involved in the pathogenesis of PD25. Epigenetic alter- binding protein 43-positive protein inclusions and by
ations, including increased methylation of promoter defects in several pathways, including protein homeo­
regions and histone modifications, have also been stasis, nucleocytoplasmic and endosomal transport,
reported in PD140. Protein ubiquitination occurs at a endosomal and vesicular transport, axon structure
higher level in cells from PD patients than in cells from and function, DNA repair, oligodendrocyte function,
normal controls28. neuroinflammation and mitochondrial function150.
Mutations in PINK1 or PRKN (the gene encoding the HD pathology is linked with cytotoxic forms of hun-
protein Parkin) are also associated with PD32. A mouse tingtin protein, which lead to global cellular impair-
study has suggested that mitophagy, which is at least in ments including synaptic dysfunction, mitochondrial
part mediated by the PINK1–Parkin pathway, mitigates toxicity and decreased rates of axonal transport151.
PD33. Genetic and pharmacological induction of NIX-​ Cockayne syndrome is characterized by severe neuro­
dependent mitophagy might have therapeutic potential degeneration, accelerated ageing, DNA repair defects,
for PD35. transcription defects and mitochondrial dysfunction152.
With respect to cellular senescence, deposition of Some studies have shown an increased frequency of
α-​s ynuclein in PD is linked with increased senes- mitochondrial dysfunction and oxidative DNA dam-
cence, and increased abundance of senescent cells and age in motor neurons from patients with ALS or ataxia
increased expression of SA-​β-gal are observed in brain telangiectasia146,153. Premature ageing diseases associated
tissue from PD patients114. with DNA repair defects, including ataxia telangiecta-
Regarding deregulated nutrient sensing, targeting of sia and xeroderma pigmentosum groups A and F, are
mTOR and AMPK also has a protective role in PD141. accompanied by neurodegeneration, suggesting a corre-
Brain cells from AD, PD and HD models show reduced lation between DNA damage and neuronal dysfunction.
glucose and oxygen metabolic rates124. Brain cells from Telomere shortening is also associated with neurodegen-
patients with PD also show dysglycaemia126. Mutations erative diseases154–156. The HD locus maps to within 325 kb
in GBA1, SMPD1 or SREBF1 increase lipid accumulation of the end of chromosome 4p, suggesting that ageing-​
and are associated with a high risk of PD142. related telomere attrition could affect its transcription
Neurogenesis decreases in the early stages of PD143. or function. Shorter telomeres have also been linked
Transplantation of iPSC-​derived dopaminergic neurons with earlier onset of pathology in a mouse model of
has shown promising results in a primate model of PD144. ALS155. In HD, transcriptional dysregulation is thought
Considering neuroinflammation, activation of microglia to precede onset of massive neuronal cell death 108.
and sustained elevation of pro-​inflammatory mediators Increased protein ubiquitination has also been observed
are associated with PD145. The mutant α-​synuclein in PD in cells from patients with HD or ALS28.
is also capable of inducing IL-1β secretion via NLRP3 Antagonistic hallmarks of ageing, such as mitochon-
inflammasome activation70. drial dysfunction and deregulated nutrient sensing,
are also involved in these other neurodegenerative dis-
Other neurodegenerative diseases eases. For example, mitophagy mitigates inflammation,
This Review focuses mainly on AD and PD because thereby decreasing the risk of neurodegenerative dis-
they are the most prevalent neurodegenerative diseases. eases. In addition, patients with HD present with low
Other neurodegenerative diseases that have important cholesterol levels and are also at increased risk of type 2
links with ageing, including ataxia telangiectasia, ALS, diabetes;126,157 mutant huntingtin accumulates in the
HD and Cockayne syndrome (Table 1), are discussed pancreas and can decrease insulin secretion.
briefly here. Integrative hallmarks of ageing are also risk factors
Ataxia telangiectasia is a rare, complex genetic neuro­ for other neurodegenerative diseases. Neurogenesis in
degenerative disorder caused by mutations in ATM, the dentate gyrus is reduced in patients with HD and

Nature Reviews | Neurology


Reviews

can be rescued by exercise, omega-3 fatty acid utiliza- DNA repair-​deficient AD mouse model 3×TgAD/Polβ+/−
tion, curcumin and flavanols128. The ATM protein is (ref.46). Nicotinamide riboside also improves learning
essential during adult neurogenesis158; however, stem and memory in APP/PS1 mice23. NMN has similar
cell therapy for ataxia telangiectasia may be associated effects in mouse and rat models of AD23. Nicotinamide
with poor outcomes, as a patient developed multifocal riboside, NMN and other NAD+ precursors also reduce
brain tumours159. Nevertheless, targeting of aged stem inflammation in animal models of ataxia telangiectasia
cell regeneration might alleviate some age-​associated and AD106,107,167. Nicotinamide riboside and P7C3, an
neurogenerative diseases in the future. allosteric activator of NAMPT, have been reported to
A correlation between neuroinflammation and neuro­ have neuroprotective ability in AD and PD mouse mod-
degenerative disease has been shown many times. els, respectively168. It has been proposed that endogenous
Chronic inflammation, persistent activation of micro- NAD+ protects against ageing-​related neurodegenerative
glia, sustained elevation of pro-​inflammatory mediators, disease by promoting efficient DNA repair, autophagy,
and increased oxidative stress are seen in brain tissue mitophagy and mitochondrial health23. Nicotinamide
from patients with HD160 and spinal cord tissue from riboside and NMN are considered to show promise as
patients with ALS161. Studies have shown that the mutant potential interventions for treating AD, PD, ALS, ataxia
huntingtin in HD and mutant SOD1G93A in familial ALS telangiectasia and other neurodegenerative diseases23;
can induce IL-1β secretion through NLRP3 inflamma­ however, their potential has yet to be confirmed in
some activation71–73. Inhibition of inflammation has been sufficiently powered clinical studies in humans. One
shown to prevent the progression of ataxia telangiectasia small study (in 30 patients) showed no statistically
symptoms in ATM-​deficient mice162. Metabolic repro- significant benefit of nicotinamide on cognitive func-
gramming is sometimes initiated when cells need to sup- tions in patients with mild to moderate AD169. Active
port and respond to defective signalling networks; for phase II and III trials of treatments targeting age-​related
example, glutamine metabolic rewiring and glutamine disease mechanisms in patients with AD and PD are
supplementation might have roles in ataxia telangiec- summarized in Table 3.
tasia163,164. Chronic inflammation contributes to tissue
reprogramming, which is itself the result of metabolic Induction of mitophagy
reprogramming. These findings suggest that regulation Defects in mitophagy have been suggested to have a role
of the immune system could have therapeutic benefits in the pathogenesis of AD and other neurodegenerative
in neurodegenerative diseases. diseases170. Agents that stimulate or rescue mitophagy
might have therapeutic potential in these diseases, a
Therapeutic approaches possibility that has shown promise in various disease
Potential treatment options for neurodegenerative dis- models170.
eases related to ageing and hallmarks of ageing include Two classes of mitophagy-​stimulating molecules
NAD+ supplementation, induction of mitophagy, inhi- are known. One type of agent promotes mitophagy by
bition of cellular senescence, and targeting of protein inducing mitochondrial damage, making these agents
aggregation, metabolism or inflammation. unsuitable as human therapeutics because of their tox-
icity. Such molecules include mitochondrial uncouplers
NAD+ supplementation (such as FCCP and CCCP, which induce mitochondrial
NAD+ is a key cofactor in the cellular redox reactions depolarization), respiration-​damaging reagents (such
involved in cellular energy metabolism and other bio- as antimycin A), oligomycin and superoxide reagents
logical processes23. An adequate cellular level of NAD+ is (such as diquat and rotenone). A second group of agents,
required for mitochondrial health, energy homeostasis, which include resveratrol and NAD+ precursors, pro-
stem cell renewal, DNA repair, neurological function mote mitophagy without interfering with mitochondria.
and resistance to cellular stress23. NAD+ is a cofactor for The pleiotropic effects of these compounds include acti-
sirtuin deacetylases and cyclic ADP-​ribose hydrolases, vation of sirtuin 1 (ref.171). This second group of mito-
and is the ADP-​ribose donor for the DNA repair protein phagy inducers could be developed as potential drugs
PARP1 (ref.23). for neurodegenerative diseases. In C. elegans, NAD+
NAD+ levels decrease with age in many different precursors activate the DCT1 pathway, increase mito-
species including yeast, D. melanogaster, mice and phagy and improve the lifespan and healthspan of atm-​
humans23. NAD+ precursors (such as nicotinamide ribo- null worms106. Resveratrol reduces β-​secretase activity
side and nicotinamide mononucleotide (NMN)), NAD+ and Aβ peptide aggregation in AD models and has
biosynthetic enzymes (such as NAMPT activators) and shown neuroprotective effects in models of AD, PD and
NAD+ degradation inhibitors (such as CD38 inhibitors HD172. Another more specific mitophagy-​stimulating
and PARP inhibitors) can increase the level of NAD+ compound, urolithin A, a metabolite compound trans-
(ref.165). Exogenous dosing with compounds that increase formed from ellagitannins by gut bacteria, has shown
the endogenous pool of NAD+ (for example, nicotina- promising results in C. elegans models of ageing-​related
mide riboside and NMN) might improve lifespan and neurodegeneration, where it normalized defects in
healthspan and delay the onset or ameliorate patholog- mobility and pharyngeal pumping170. Urolithin A has
ical features of some ageing-​related diseases, including also shown beneficial effects on learning and memory in
AD and PD23,166. As an example, nicotinamide riboside the APP/PS1 mouse model111. In rats, urolithin A lacked
decreases tau phosphorylation and improves synaptic toxic adverse effects after oral dosing for 90 days173,
and cognitive functions in 3×TgAD mice and in the which has increased interest in its therapeutic use.

www.nature.com/nrneurol
Reviews

Table 3 | Active phase II/III clinical trials targeting age-​related disease mechanisms in AD and PD
Target Mechanisms of Drug Disease estimated Clinicaltrials.
action (n) termination gov
date identifier
Metabolism Cell signalling and cell Insulin AD (30) September 2019 NCT02462161
growth
AD (90) May 2019b NCT02503501
Glucagon-​like peptide-1 AD (206) December 2019 NCT01843075
agonists (liraglutide,
semaglutide) PD (57) December 2020 NCT02953665
PD (120)a December 2024 NCT03659682
Mitochondrial Antioxidant; autophagy Nicotinamide (vitamin B3) AD (48) June 2020 NCT03061474
function induction; DNA repair
induction; histone PD (200)a October 2021 NCT03568968
deacetylase inhibition N-acetylcysteine, l-carnitine AD (60), September 2020 NCT04044131
(inhibition of tau tartrate, nicotinamide PD (60)a
phosphorylation); riboside and serine
anti-​inflammatory
S-equol AD (40) October 2019 NCT03101085
Niacin PD (80) September 2019 NCT03462680
Resveratrol AD (48) October 2019 NCT02502253
Grape powder AD (32) May 2019 NCT03361410
Tyrosine kinase Nilotinib PD (135) October 2020 NCT03205488
inhibitor
AD (42) February 2020 NCT02947893
Inflammation Angiotensin-​II Telmisartan AD (240) March 2021 NCT02085265
receptor blocker
Candesartan AD (72) September 2021 NCT02646982
Anti-​viral Valacyclovir AD (130) August 2022 NCT03282916
Anti-​inflammatory Salsalate (salicylic acid) AD (40) October 2019 NCT03277573
Mast cell and ALZT-​OP1 (cromolyn, AD (600) November 2019 NCT02547818
macrophage inhibition ibuprofen)
Trials targeting protein aggregation as the primary mechanism and trials only evaluating bioavailability , biomarkers and safety are
not mentioned here. Reference: www.clinicaltrials.gov August 2019. AD, Alzheimer disease; PD, Parkinson disease. aNot yet
recruiting. bActive, not recruiting, results not yet published.

Lifestyle modifications that protect against ageing-​ Elimination strategies using FOXO4 peptide treatment,
related neurodegeneration, including exercise and cal- inhibition of BCL-​W and BCL-​X, and immunotherapy
orie restriction, also increase mitophagy; however, this all had beneficial effects on age-​related dysfunction
effect is likely to be indirect, instead reflecting a complex in animal models180. From these findings, inhibition
multifaceted organismal response174. of SASP, elimination of senescent cells and reversal of
cell senescence might all represent effective anti-​
Inhibition of cellular senescence ageing treatments and innovative strategies for treating
Studies have shown that mTOR is a crucial biochemical neurodegenerative diseases.
modulator of ageing and promotes SASP175. Inhibition
of prosenescent mTOR signalling with rapamycin decel- Targeting protein aggregation
erates cellular senescence in the ageing brain, perhaps Protein aggregation is a prominent feature of AD
by combining its multiple neuroprotective effects with and other neurodegenerative diseases, as described
suppression of cellular senescence176. above181,182. Therefore, several therapeutic approaches
Metformin has been shown to suppress cellular for AD aim to reduce the abundance of Aβ aggregates
senescence via activation of microRNA-​processing via immunotherapy or by inhibiting γ-​s ecretase or
proteins and to prevent amyloid plaque deposition and β-​secretase84. Multiple therapies of this type, investigated
α-​synuclein phosphorylation in AD and PD, respec- over 10–20 years, have produced disappointing results.
tively177,178. In a tau-​overexpression neurodegeneration Other strategies targeting tau aggregates and NFTs183,
mouse model, increased numbers of senescent astro- which are associated with several neurodegene­rative
cytes and microglia were associated with an increase in disorders, including AD, frontotemporal lobar degene­
the deposition of tau NFTs, whereas clearance of these ration, progressive supranuclear palsy, corticobasal
cells prevented tau-​dependent pathology and cognitive degeneration and Pick disease, are under investiga-
decline118. Senescent astrocytes or microglia accumulate tion93,184. Very few phase III clinical trials testing tau as a
with ageing and have been detected in patients with AD, therapeutic target for AD are currently in progress, but
PD and ALS114,115, so removal of senescent cells might many phase II trials targeting tau pathology in AD are
provide a novel therapeutic approach to the treatment moving through the pipeline185. As these studies pro-
of age-​related neurodegenerative diseases179 (Fig.  5). gress, it will become clearer whether strategies targeting

Nature Reviews | Neurology


Reviews

tau aggregates have therapeutic benefit for any of the adaptor protein STING. A 2018 study suggested that
neurodegenerative diseases. genomic instability and DNA damage activate cGAS,
linking DNA damage with inflammation, cellular senes-
Targeting metabolism and inflammation cence and cancer193, but a link between cGAS and AD
Human and animal studies have shown that dysregu- has not yet been investigated. The therapeutic potential
lation of insulin signalling contributes to ageing and of targeting the STING pathway in neurodegeneration
increases susceptibility to neurodegenerative diseases. is currently under investigation33.
Consistent with this idea, some antidiabetic compounds
are neuroprotective and improve cognitive function133,186. Conclusions
So far, the most promising treatments have involved Efforts to develop evidence-​based treatment strategies
administration of thiazolidinediones, especially pioglita- for neurodegenerative diseases are ongoing, but nei-
zone186 in AD, but ongoing clinical trials are also testing ther highly effective treatments nor potent protective
the efficacy of insulin and glucagon-​like peptide-1 in AD approaches have yet been identified. In the future, more
and PD (Table 3). studies on neurodegenerative diseases should focus
Neuroinflammation is a major pathophysiolog- on the nine hallmarks of ageing, most of which show
ical feature of neurodegenerative disorders. Anti-​ associations with neurodegenerative diseases. Among
inflammatory interventions have, however, been largely these hallmarks, those relating to DNA damage and
ineffective, although ibuprofen might reduce the risk mitochondrial dysfunction are central, and studies
of developing PD187,188. In a recently completed trial investigating communication between the nucleus and
(NCT02588677), the tyrosine kinase inhibitor mas- mitochondria will contribute to a mechanistic under-
itinib showed promising effects in patients with ALS189. standing of ageing and the pathogenesis of neuro­
Masitinib targets mast cells and macrophages by inhib- degenerative diseases. Although in the present Review
iting a number of protein kinases and also targets micro- we narrow our focus to aspects of DNA damage, mito-
glia and inhibits inflammatory processes within the chondrial dysfunction, cellular senescence and inflam-
CNS. Trials investigating other anti-​inflammatory drugs mation, which are frequently considered to be causes of
as therapies for ALS have shown negative results190. neurodegeneration, other ageing-​related hallmarks are
also important in neurodegenerative disease and the dif-
Other therapeutic approaches ferent hallmarks are highly interconnected. For example,
Other pharmacological interventions for neurological the loss of proteostasis owing to defects in proteasome or
diseases target various cellular processes or molecules autophagy observed in AD and PD is strongly related to
that promote or delay ageing, including calorie restric- inflammation and senescence13,194. Metabolic dysfunc-
tion, oxidative stress, telomerase, autophagy, stem cell tion observed in neurodegeneration is associated with
renewal and epigenetic mechanisms. alterations in NAD+ levels, mitochondrial dysfunction
Calorie restriction increases lifespan in multiple and oxidative stress57. Loss of functional stem cells is
species by modulating AMPK, IGF1 and mTOR signal- related to almost all of the ageing hallmarks, includ-
ling pathways and the processes they regulate including ing DNA damage, epigenetic deregulation, mitochon-
nutrient sensing, autophagy, mitochondrial function and drial dysfunction, telomerase inactivation and cell
cell proliferation. Some compounds, including 2-deoxy-​ senescence58.
d-glucose and resveratrol, mimic the beneficial effects of Given the complex nature of neurodegenerative
calorie restriction. diseases, and the fact that genetic and environmental
Many antioxidants are thought to be neuroprotective, factors can determine disease progression, single-​drug
including vitamin E, quercetin, N-​acetyl-l-​c ysteine, or single-​pathway-targeted approaches might be inad-
curcumin, carotenoids, flavonoids, isothiocyanates, equate — more holistic approaches or combined treat-
terpenoids, proanthocyanidins, omega-3 fatty acids, ment strategies might be necessary and more likely to
melatonin and coenzyme Q10 (CoQ10)191. Low CoQ10 succeed. The association between neurodegenerative
levels have been observed in patients with PD, HD diseases and ageing hallmarks could bring new hope for
or Friedreich ataxia (another debilitating neurolog- the treatment of such diseases. Multi-​targeted pharma-
ical movement disorder)192. However, clinical trials of ceutical evidence-​based approaches might need to be
CoQ10 supplementation have shown limited efficacy combined with non-​pharmacological approaches and/or
in patients with AD, PD or ALS192. Cyclic GMP–AMP lifestyle modification in order to slow the epidemic of
synthase (cGAS) is a DNA sensor that triggers innate neurological disease in elderly individuals.
immune responses mediated by the secondary messen-
ger cyclic GMP–AMP, which binds and activates the Published online xx xx xxxx

1. Rose, M. R. Adaptation, aging, and genomic 6. Dean, D. C., 3rd et al. Brain differences in infants 9. Hy, L. X. & Keller, D. M. Prevalence of AD among
information. Aging 1, 444–450 (2009). at differential genetic risk for late-​onset Alzheimer whites: a summary by levels of severity. Neurology 55,
2. Carmona, J. J. & Michan, S. Biology of healthy aging disease: a cross-​sectional imaging study. JAMA 198–204 (2000).
and longevity. Rev. Invest. Clin. 68, 7–16 (2016). Neurol. 71, 11–22 (2014). 10. Poewe, W. et al. Parkinson disease. Nat. Rev. Dis.
3. Alzheimer’s Association. 2018 Alzheimer’s disease facts 7. Schaefers, A. T. & Teuchert-​Noodt, G. Developmental Primers 3, 17013 (2017).
and figures. Alzheimers Dement. 14, 367–429 (2018). neuroplasticity and the origin of neurodegenerative 11. Mehta, P. et al. Prevalence of amyotrophic lateral
4. Wyss-​Coray, T. Ageing, neurodegeneration and brain diseases. World J. Biol. Psychiatry 17, 587–599 sclerosis – United States, 2014. MMWR Morb.
rejuvenation. Nature 539, 180–186 (2016). (2016). Mortal. Wkly. Rep. 67, 216–218 (2018).
5. Elobeid, A., Libard, S., Leino, M., Popova, S. N. 8. Nussbaum, R. L. & Ellis, C. E. Alzheimer’s disease and 12. Robinson, J. L. et al. Neurodegenerative disease
& Alafuzoff, I. Altered proteins in the aging brain. Parkinson’s disease. N. Engl. J. Med. 348, concomitant proteinopathies are prevalent, age-​related
J. Neuropathol. Exp. Neurol. 75, 316–325 (2016). 1356–1364 (2003). and APOE4-associated. Brain 141, 2181–2193 (2018).

www.nature.com/nrneurol
Reviews

13. Lopez-​Otin, C., Blasco, M. A., Partridge, L., Serrano, 41. Kuilman, T., Michaloglou, C., Mooi, W. J. & Peeper, D. S. 69. Halle, A. et al. The NALP3 inflammasome is involved
M. & Kroemer, G. The hallmarks of aging. Cell 153, The essence of senescence. Genes Dev. 24, 2463–2479 in the innate immune response to amyloid-​beta.
1194–1217 (2013). (2010). Nat. Immunol. 9, 857–865 (2008).
14. Chow, H. M. & Herrup, K. Genomic integrity and the 42. Loaiza, N. & Demaria, M. Cellular senescence and 70. Codolo, G. et al. Triggering of inflammasome by
ageing brain. Nat. Rev. Neurosci. 16, 672–684 tumor promotion: is aging the key? Biochim. Biophys. aggregated alpha-​synuclein, an inflammatory response
(2015). Acta 1865, 155–167 (2016). in synucleinopathies. PLoS One 8, e55375 (2013).
15. Madabhushi, R., Pan, L. & Tsai, L. H. DNA damage 43. Coppe, J. P., Desprez, P. Y., Krtolica, A. & Campisi, J. 71. Ona, V. O. et al. Inhibition of caspase-1 slows disease
and its links to neurodegeneration. Neuron 83, The senescence-​associated secretory phenotype: the progression in a mouse model of Huntington’s
266–282 (2014). dark side of tumor suppression. Annu. Rev. Pathol. 5, disease. Nature 399, 263–267 (1999).
16. Jeppesen, D. K., Bohr, V. A. & Stevnsner, T. DNA 99–118 (2010). 72. Johann, S. et al. NLRP3 inflammasome is expressed
repair deficiency in neurodegeneration. Prog. 44. Wiley, C. D. et al. Mitochondrial dysfunction induces by astrocytes in the SOD1 mouse model of ALS and in
Neurobiol. 94, 166–200 (2011). senescence with a distinct secretory phenotype. human sporadic ALS patients. Glia 63, 2260–2273
17. Thanan, R. et al. Oxidative stress and its significant Cell Metab. 23, 303–314 (2016). (2015).
roles in neurodegenerative diseases and cancer. 45. Nacarelli, T. et al. NAD(+) metabolism governs the 73. Meissner, F., Molawi, K. & Zychlinsky, A. Mutant
Int. J. Mol. Sci. 16, 193–217 (2015). proinflammatory senescence-​associated secretome. superoxide dismutase 1-induced IL-1β accelerates ALS
18. McKinnon, P. J. Maintaining genome stability in the Nat. Cell Biol. 21, 397–407 (2019). pathogenesis. Proc. Natl Acad. Sci. USA 107,
nervous system. Nat. Neurosci. 16, 1523–1529 46. Maynard, S., Fang, E. F., Scheibye-​Knudsen, M., 13046–13050 (2010).
(2013). Croteau, D. L. & Bohr, V. A. DNA damage, DNA repair, 74. Wang, W. Y., Tan, M. S., Yu, J. T. & Tan, L. Role of
19. Maynard, S., Schurman, S. H., Harboe, C., de Souza-​ aging, and neurodegeneration. Cold Spring Harb. pro-​inflammatory cytokines released from microglia in
Pinto, N. C. & Bohr, V. A. Base excision repair of Perspect. Med. https://doi.org/10.1101/cshperspect. Alzheimer’s disease. Ann. Transl Med. 3, 136 (2015).
oxidative DNA damage and association with cancer a025130 (2015). 75. Wang, W. et al. Caspase-1 causes truncation and
and aging. Carcinogenesis 30, 2–10 (2009). 47. Barrio-​Alonso, E., Hernandez-​Vivanco, A., Walton, C. C., aggregation of the Parkinson’s disease-​associated
20. Tell, G. & Demple, B. Base excision DNA repair and Perea, G. & Frade, J. M. Cell cycle reentry triggers protein α-​synuclein. Proc. Natl Acad. Sci. USA 113,
cancer. Oncotarget 6, 584–585 (2015). hyperploidization and synaptic dysfunction followed by 9587–9592 (2016).
21. Leandro, G. S., Sykora, P. & Bohr, V. A. The impact delayed cell death in differentiated cortical neurons. 76. Uchoa, M. F., Moser, V. A. & Pike, C. J. Interactions
of base excision DNA repair in age-​related Sci. Rep. 8, 14316 (2018). between inflammation, sex steroids, and Alzheimer’s
neurodegenerative diseases. Mutat. Res. 776, 31–39 48. Fielder, E., von Zglinicki, T. & Jurk, D. The DNA disease risk factors. Front. Neuroendocrinol. 43,
(2015). damage response in neurons: die by apoptosis or 60–82 (2016).
22. Akbari, M., Morevati, M., Croteau, D. & Bohr, V. A. survive in a senescence-​like state? J. Alzheimers Dis. 77. Grune, T., Jung, T., Merker, K. & Davies, K. J.
The role of DNA base excision repair in brain 60, S107–S131 (2017). Decreased proteolysis caused by protein aggregates,
homeostasis and disease. DNA Repair 32, 172–179 49. Jurk, D. et al. Postmitotic neurons develop a p21- inclusion bodies, plaques, lipofuscin, ceroid,
(2015). dependent senescence-​like phenotype driven by a and ‘aggresomes’ during oxidative stress, aging, and
23. Fang, E. F. et al. NAD(+) in aging: molecular mechanisms DNA damage response. Aging Cell 11, 996–1004 disease. Int. J. Biochem. Cell Biol. 36, 2519–2530
and translational implications. Trends Mol. Med. 23, (2012). (2004).
899–916 (2017). 50. Vaidya, A. et al. Knock-​in reporter mice demonstrate 78. Nakahira, K. et al. Autophagy proteins regulate innate
24. Herrmann, M., Pusceddu, I., Marz, W. & Herrmann, W. that DNA repair by non-​homologous end joining immune responses by inhibiting the release of
Telomere biology and age-​related diseases. Clin. Chem. declines with age. PLoS Genet. 10, e1004511 (2014). mitochondrial DNA mediated by the NALP3
Lab. Med. 56, 1210–1222 (2018). 51. Narita, M. et al. Spatial coupling of mTOR and inflammasome. Nat. Immunol. 12, 222–230 (2011).
25. Eitan, E., Hutchison, E. R. & Mattson, M. P. Telomere autophagy augments secretory phenotypes. Science 79. Baker, D. J. & Petersen, R. C. Cellular senescence in brain
shortening in neurological disorders: an abundance of 332, 966–970 (2011). aging and neurodegenerative diseases: evidence and
unanswered questions. Trends Neurosci. 37, 52. Gewirtz, D. A. Autophagy and senescence: perspectives. J. Clin. Invest. 128, 1208–1216 (2018).
256–263 (2014). a partnership in search of definition. Autophagy 9, 80. Pan, M. R., Li, K., Lin, S. Y. & Hung, W. C. Connecting
26. Bradley-​Whitman, M. A. & Lovell, M. A. Epigenetic 808–812 (2013). the dots: from DNA damage and repair to aging. Int. J.
changes in the progression of Alzheimer’s disease. 53. Kang, H. T., Lee, K. B., Kim, S. Y., Choi, H. R. & Park, S. C. Mol. Sci. 17, 685 (2016).
Mech. Ageing Dev. 134, 486–495 (2013). Autophagy impairment induces premature senescence 81. Frasca, D. & Blomberg, B. B. Inflammaging decreases
27. Hwang, J. Y., Aromolaran, K. A. & Zukin, R. S. The in primary human fibroblasts. PLoS One 6, e23367 adaptive and innate immune responses in mice and
emerging field of epigenetics in neurodegeneration (2011). humans. Biogerontology 17, 7–19 (2016).
and neuroprotection. Nat. Rev. Neurosci. 18, 54. Kang, C. et al. The DNA damage response induces 82. Bektas, A., Schurman, S. H., Sen, R. & Ferrucci, L.
347–361 (2017). inflammation and senescence by inhibiting autophagy Aging, inflammation and the environment.
28. Tanaka, K. & Matsuda, N. Proteostasis and of GATA4. Science 349, aaa5612 (2015). Exp. Gerontol. 105, 10–18 (2018).
neurodegeneration: the roles of proteasomal 55. Bhatia-​Dey, N., Kanherkar, R. R., Stair, S. E., 83. Valera, E. et al. Combination of alpha-​synuclein
degradation and autophagy. Biochim. Biophys. Acta Makarev, E. O. & Csoka, A. B. Cellular senescence immunotherapy with anti-​inflammatory treatment in
1843, 197–204 (2014). as the causal nexus of aging. Front. Genet. 7, 13 a transgenic mouse model of multiple system atrophy.
29. Johri, A. & Beal, M. F. Mitochondrial dysfunction in (2016). Acta Neuropathol. Commun. 5, 2 (2017).
neurodegenerative diseases. J. Pharmacol. Exp. Ther. 56. Fontana, L., Partridge, L. & Longo, V. D. Extending 84. Sevigny, J. et al. The antibody aducanumab reduces
342, 619–630 (2012). healthy life span – from yeast to humans. Science Aβ plaques in Alzheimer’s disease. Nature 537,
30. Keogh, M. J. & Chinnery, P. F. Mitochondrial DNA 328, 321–326 (2010). 50–56 (2016).
mutations in neurodegeneration. Biochim. Biophys. 57. Babbar, M. & Sheikh, M. S. Metabolic stress and 85. Van Cauwenberghe, C., Vandendriessche, C., Libert, C.
Acta 1847, 1401–1411 (2015). disorders related to alterations in mitochondrial & Vandenbroucke, R. E. Caloric restriction: beneficial
31. Greene, J. C. et al. Mitochondrial pathology and fission or fusion. Mol. Cell. Pharmacol. 5, 109–133 effects on brain aging and Alzheimer’s disease.
apoptotic muscle degeneration in Drosophila parkin (2013). Mamm. Genome 27, 300–319 (2016).
mutants. Proc. Natl Acad. Sci. USA 100, 4078–4083 58. Oh, J., Lee, Y. D. & Wagers, A. J. Stem cell aging: 86. Spielman, L. J., Little, J. P. & Klegeris, A. Physical
(2003). mechanisms, regulators and therapeutic opportunities. activity and exercise attenuate neuroinflammation in
32. Pickrell, A. M. & Youle, R. J. The roles of PINK1, Nat. Med. 20, 870–880 (2014). neurological diseases. Brain Res. Bull. 125, 19–29
parkin, and mitochondrial fidelity in Parkinson’s 59. Villeda, S. A. et al. Young blood reverses age-​related (2016).
disease. Neuron 85, 257–273 (2015). impairments in cognitive function and synaptic 87. Bekris, L. M., Yu, C. E., Bird, T. D. & Tsuang, D. W.
33. Sliter, D. A. et al. Parkin and PINK1 mitigate plasticity in mice. Nat. Med. 20, 659–663 (2014). Genetics of Alzheimer disease. J. Geriatr. Psychiatry
STING-​induced inflammation. Nature 561, 258–262 60. Amor, S. & Woodroofe, M. N. Innate and adaptive Neurol. 23, 213–227 (2010).
(2018). immune responses in neurodegeneration and repair. 88. Liu, C. C., Liu, C. C., Kanekiyo, T., Xu, H. & Bu, G.
34. Fivenson, E. M. et al. Mitophagy in neurodegeneration Immunology 141, 287–291 (2014). Apolipoprotein E and Alzheimer disease: risk,
and aging. Neurochem. Int. 109, 202–209 (2017). 61. He, F. & Balling, R. The role of regulatory T cells in mechanisms and therapy. Nat. Rev. Neurol. 9,
35. Koentjoro, B., Park, J. S. & Sue, C. M. Nix restores neurodegenerative diseases. Wiley Interdiscip. Rev. 106–118 (2013).
mitophagy and mitochondrial function to protect Syst. Biol. Med. 5, 153–180 (2013). 89. Bloom, G. S. Amyloid-​β and tau: the trigger and bullet
against PINK1/parkin-​related Parkinson’s disease. 62. Cao, W. & Zheng, H. Peripheral immune system in in Alzheimer disease pathogenesis. JAMA Neurol. 71,
Sci. Rep. 7, 44373 (2017). aging and Alzheimer’s disease. Mol. Neurodegener. 505–508 (2014).
36. Di Rita, A. et al. AMBRA1-mediated mitophagy 13, 51 (2018). 90. Wang, J., Gu, B. J., Masters, C. L. & Wang, Y. J.
counteracts oxidative stress and apoptosis induced by 63. Giunta, S. Is inflammaging an auto[innate]immunity A systemic view of Alzheimer disease – insights from
neurotoxicity in human neuroblastoma SH-​SY5Y cells. subclinical syndrome? Immun. Ageing 3, 12 (2006). amyloid-​β metabolism beyond the brain. Nat. Rev.
Front. Cell. Neurosci. 12, 92 (2018). 64. Currais, A. Ageing and inflammation – a central role Neurol. 13, 612–623 (2017).
37. Yun, J. et al. MUL1 acts in parallel to the PINK1/ for mitochondria in brain health and disease. Ageing 91. Lane, C. A., Hardy, J. & Schott, J. M. Alzheimer’s
parkin pathway in regulating mitofusin and Res. Rev. 21, 30–42 (2015). disease. Eur. J. Neurol. 25, 59–70 (2018).
compensates for loss of PINK1/parkin. Elife 3, 65. Lu, T. et al. Gene regulation and DNA damage in the 92. Selkoe, D. J. & Hardy, J. The amyloid hypothesis of
e01958 (2014). ageing human brain. Nature 429, 883–891 (2004). Alzheimer’s disease at 25 years. EMBO Mol. Med. 8,
38. Haynes, C. M. & Ron, D. The mitochondrial UPR – 66. Hickman, S., Izzy, S., Sen, P., Morsett, L. & El Khoury, J. 595–608 (2016).
protecting organelle protein homeostasis. J. Cell Sci. Microglia in neurodegeneration. Nat. Neurosci. 21, 93. Spillantini, M. G. & Goedert, M. Tau pathology and
123, 3849–3855 (2010). 1359–1369 (2018). neurodegeneration. Lancet Neurol. 12, 609–622
39. Hayflick, L. The limited in vitro lifetime of human 67. Cunningham, C. Microglia and neurodegeneration: (2013).
diploid cell strains. Exp. Cell Res. 37, 614–636 the role of systemic inflammation. Glia 61, 71–90 94. Fu, W. Y., Wang, X. & Ip, N. Y. Targeting
(1965). (2013). neuroinflammation as a therapeutic strategy for
40. Kultz, D. Molecular and evolutionary basis of the 68. Youm, Y. H. et al. Canonical Nlrp3 inflammasome links Alzheimer’s disease: mechanisms, drug candidates,
cellular stress response. Annu. Rev. Physiol. 67, systemic low-​grade inflammation to functional decline and new opportunities. ACS Chem. Neurosci. 10,
225–257 (2005). in aging. Cell Metab. 18, 519–532 (2013). 872–879 (2019).

Nature Reviews | Neurology


Reviews

95. Hardy, J. The amyloid hypothesis for Alzheimer’s IGF-1 resistance, IRS-1 dysregulation, and cognitive 148. Valentin-​Vega, Y. A. et al. Mitochondrial dysfunction in
disease: a critical reappraisal. J. Neurochem. 110, decline. J. Clin. Invest. 122, 1316–1338 (2012). ataxia-​telangiectasia. Blood 119, 1490–1500
1129–1134 (2009). 122. Oddo, S. The role of mTOR signaling in Alzheimer (2012).
96. Hall, A. M. & Roberson, E. D. Mouse models of disease. Front. Biosci. (Schol Ed) 4, 941–952 149. Guo, Z., Kozlov, S., Lavin, M. F., Person, M. D. & Paull,
Alzheimer’s disease. Brain Res. Bull. 88, 3–12 (2012). T. T. ATM activation by oxidative stress. Science 330,
(2012). 123. Salminen, A., Kaarniranta, K., Haapasalo, A., 517–521 (2010).
97. Ewald, C. Y. & Li, C. Understanding the molecular Soininen, H. & Hiltunen, M. AMP-​activated protein 150. Hardiman, O. et al. Amyotrophic lateral sclerosis.
basis of Alzheimer’s disease using a Caenorhabditis kinase: a potential player in Alzheimer’s disease. Nat. Rev. Dis. Primers 3, 17071 (2017).
elegans model system. Brain Struct. Funct. 214, J. Neurochem. 118, 460–474 (2011). 151. Bates, G. P. et al. Huntington disease. Nat. Rev.
263–283 (2010). 124. Camandola, S. & Mattson, M. P. Brain metabolism in Dis. Primers 1, 15005 (2015).
98. Prussing, K., Voigt, A. & Schulz, J. B. Drosophila health, aging, and neurodegeneration. EMBO J. 36, 152. Karikkineth, A. C., Scheibye-​Knudsen, M., Fivenson, E.,
melanogaster as a model organism for Alzheimer’s 1474–1492 (2017). Croteau, D. L. & Bohr, V. A. Cockayne syndrome:
disease. Mol. Neurodegener. 8, 35 (2013). 125. De Felice, F. G. & Lourenco, M. V. Brain metabolic clinical features, model systems and pathways. Ageing
99. Tan, F. H. P. & Azzam, G. Drosophila melanogaster: stress and neuroinflammation at the basis of cognitive Res. Rev. 33, 3–17 (2017).
deciphering Alzheimer’s disease. Malays. J. Med. Sci. impairment in Alzheimer’s disease. Front. Aging 153. Penndorf, D., Witte, O. W. & Kretz, A. DNA plasticity
24, 6–20 (2017). Neurosci. 7, 94 (2015). and damage in amyotrophic lateral sclerosis. Neural
100. Arber, C., Lovejoy, C. & Wray, S. Stem cell models 126. Cai, H. et al. Metabolic dysfunction in Alzheimer’s Regen. Res. 13, 173–180 (2018).
of Alzheimer’s disease: progress and challenges. disease and related neurodegenerative disorders. 154. Cai, Z., Yan, L. J. & Ratka, A. Telomere shortening and
Alzheimers Res. Ther. 9, 42 (2017). Curr. Alzheimer Res. 9, 5–17 (2012). Alzheimer’s disease. Neuromol. Med. 15, 25–48
101. Teng, E. et al. Dietary DHA supplementation in an 127. Simons, M. et al. Cholesterol depletion inhibits the (2013).
APP/PS1 transgenic rat model of AD reduces generation of β-​amyloid in hippocampal neurons. 155. Linkus, B. et al. Telomere shortening leads to earlier
behavioral and Aβ pathology and modulates Aβ Proc. Natl Acad. Sci. USA 95, 6460–6464 (1998). age of onset in ALS mice. Aging 8, 382–393 (2016).
oligomerization. Neurobiol. Dis. 82, 552–560 (2015). 128. Vivar, C. Adult hippocampal neurogenesis, aging and 156. Kota, L. N. et al. Reduced telomere length in
102. Lovell, M. A., Gabbita, S. P. & Markesbery, W. R. neurodegenerative diseases: possible strategies to neurodegenerative disorders may suggest shared
Increased DNA oxidation and decreased levels of prevent cognitive impairment. Curr. Top. Med. Chem. biology. J. Neuropsychiatry Clin. Neurosci. 27,
repair products in Alzheimer’s disease ventricular CSF. 15, 2175–2192 (2015). e92–e96 (2015).
J. Neurochem. 72, 771–776 (1999). 129. McClean, P. L., Parthsarathy, V., Faivre, E. & Holscher, C. 157. Block, R. C., Dorsey, E. R., Beck, C. A., Brenna, J. T.
103. Weissman, L. et al. Defective DNA base excision repair The diabetes drug liraglutide prevents degenerative & Shoulson, I. Altered cholesterol and fatty acid
in brain from individuals with Alzheimer’s disease and processes in a mouse model of Alzheimer’s disease. metabolism in Huntington disease. J. Clin. Lipidol. 4,
amnestic mild cognitive impairment. Nucleic Acids J. Neurosci. 31, 6587–6594 (2011). 17–23 (2010).
Res. 35, 5545–5555 (2007). 130. Ho, L. et al. Diet-​induced insulin resistance promotes 158. Allen, D. M. et al. Ataxia telangiectasia mutated is
104. Sykora, P. et al. DNA polymerase β deficiency leads to amyloidosis in a transgenic mouse model of essential during adult neurogenesis. Genes Dev. 15,
neurodegeneration and exacerbates Alzheimer Alzheimer’s disease. FASEB J. 18, 902–904 (2004). 554–566 (2001).
disease phenotypes. Nucleic Acids Res. 43, 943–959 131. Takamatsu, Y. et al. Combined immunotherapy with 159. Amariglio, N. et al. Donor-​derived brain tumor
(2015). “anti-​insulin resistance” therapy as a novel therapeutic following neural stem cell transplantation in an ataxia
105. Wang, H. Z. et al. Validating GWAS-​identified risk loci strategy against neurodegenerative diseases. telangiectasia patient. PLoS Med. 6, e1000029
for Alzheimer’s disease in Han Chinese populations. NPJ Parkinsons Dis. 3, 4 (2017). (2009).
Mol. Neurobiol. 53, 379–390 (2016). 132. Heneka, M. T., Reyes-​Irisarri, E., Hull, M. & 160. Sapp, E. et al. Early and progressive accumulation
106. Fang, E. F. et al. NAD(+) replenishment improves Kummer, M. P. Impact and therapeutic potential of of reactive microglia in the Huntington disease brain.
lifespan and healthspan in ataxia telangiectasia PPARs in Alzheimer’s disease. Curr. Neuropharmacol. J. Neuropathol. Exp. Neurol. 60, 161–172 (2001).
models via mitophagy and DNA repair. Cell Metab. 9, 643–650 (2011). 161. Henkel, J. S. et al. Presence of dendritic cells, MCP-1,
24, 566–581 (2016). 133. Rotermund, C., Machetanz, G. & Fitzgerald, J. C. and activated microglia/macrophages in amyotrophic
107. Hou, Y. et al. NAD(+) supplementation normalizes The therapeutic potential of metformin in lateral sclerosis spinal cord tissue. Ann. Neurol. 55,
key Alzheimer’s features and DNA damage responses neurodegenerative diseases. Front. Endocrinol. 9, 221–235 (2004).
in a new AD mouse model with introduced DNA 400 (2018). 162. Hui, C. W., Song, X., Ma, F., Shen, X. & Herrup, K.
repair deficiency. Proc. Natl Acad. Sci. USA 115, 134. Sarlus, H. & Heneka, M. T. Microglia in Alzheimer’s Ibuprofen prevents progression of ataxia
E1876–E1885 (2018). disease. J. Clin. Invest. 127, 3240–3249 (2017). telangiectasia symptoms in ATM-​deficient mice.
108. Kwon, M. J., Kim, S., Han, M. H. & Lee, S. B. 135. Melki, R. Role of different alpha-​synuclein strains J. Neuroinflammation 15, 308 (2018).
Epigenetic changes in neurodegenerative diseases. in synucleinopathies, similarities with other 163. Chow, H. M. et al. ATM is activated by ATP depletion
Mol. Cells 39, 783–789 (2016). neurodegenerative diseases. J. Parkinsons Dis. 5, and modulates mitochondrial function through NRF1.
109. Ow, S. Y. & Dunstan, D. E. A brief overview of amyloids 217–227 (2015). J. Cell Biol. 218, 909–928 (2019).
and Alzheimer’s disease. Protein Sci. 23, 1315–1331 136. Rocha, E. M., De Miranda, B. & Sanders, L. H. Alpha-​ 164. Chen, J. et al. The impact of glutamine supplementation
(2014). synuclein: pathology, mitochondrial dysfunction and on the symptoms of ataxia-​telangiectasia: a preclinical
110. Kerr, J. S. et al. Mitophagy and Alzheimer’s disease: neuroinflammation in Parkinson’s disease. Neurobiol. assessment. Mol. Neurodegener. 11, 60 (2016).
cellular and molecular mechanisms. Trends Neurosci. Dis. 109, 249–257 (2018). 165. Trammell, S. A. et al. Nicotinamide riboside is uniquely
40, 151–166 (2017). 137. Dias, V., Junn, E. & Mouradian, M. M. The role of and orally bioavailable in mice and humans. Nat.
111. Fang, E. F. et al. Mitophagy inhibits amyloid-​β and oxidative stress in Parkinson’s disease. J. Parkinsons Commun. 7, 12948 (2016).
tau pathology and reverses cognitive deficits in models Dis. 3, 461–491 (2013). 166. Zhang, H. et al. NAD(+) repletion improves
of Alzheimer’s disease. Nat. Neurosci. 22, 401–412 138. Hirsch, E. C. & Hunot, S. Neuroinflammation in mitochondrial and stem cell function and enhances life
(2019). Parkinson’s disease: a target for neuroprotection? span in mice. Science 352, 1436–1443 (2016).
112. Bhat, R. et al. Astrocyte senescence as a component Lancet Neurol. 8, 382–397 (2009). 167. Gomes, A. P. et al. Declining NAD(+) induces a
of Alzheimer’s disease. PLoS One 7, e45069 (2012). 139. Sepe, S. et al. Inefficient DNA repair is an aging-​ pseudohypoxic state disrupting nuclear-​mitochondrial
113. Boccardi, V., Pelini, L., Ercolani, S., Ruggiero, C. & related modifier of Parkinson’s disease. Cell Rep. 15, communication during aging. Cell 155, 1624–1638
Mecocci, P. From cellular senescence to Alzheimer’s 1866–1875 (2016). (2013).
disease: the role of telomere shortening. Ageing Res. 140. Labbe, C., Lorenzo-​Betancor, O. & Ross, O. A. 168. De Jesus-​Cortes, H. et al. Neuroprotective efficacy
Rev. 22, 1–8 (2015). Epigenetic regulation in Parkinson’s disease. Acta of aminopropyl carbazoles in a mouse model of
114. Chinta, S. J. et al. Cellular senescence is induced Neuropathol. 132, 515–530 (2016). Parkinson disease. Proc. Natl Acad. Sci. USA 109,
by the environmental neurotoxin paraquat and 141. Curry, D. W., Stutz, B., Andrews, Z. B. & Elsworth, J. D. 17010–17015 (2012).
contributes to neuropathology linked to Parkinson’s Targeting AMPK signaling as a neuroprotective 169. Phelan, M. J., Mulnard, R. A., Gillen, D. L. &
disease. Cell Rep 22, 930–940 (2018). strategy in Parkinson’s disease. J. Parkinsons Dis. 8, Schreiber, S. S. Phase II clinical trial of nicotinamide
115. Turnquist, C. et al. p53 isoforms regulate astrocyte-​ 161–181 (2018). for the treatment of mild to moderate Alzheimer’s
mediated neuroprotection and neurodegeneration. 142. Alecu, I. & Bennett, S. A. L. Dysregulated lipid disease. J. Geriatr. Med. Gerontol. 3, 021 (2017).
Cell Death Differ. 23, 1515–1528 (2016). metabolism and its role in α-​synucleinopathy in 170. Ryu, D. et al. Urolithin A induces mitophagy and
116. He, N. et al. Amyloid-​β(1-42) oligomer accelerates Parkinson’s disease. Front. Neurosci. 13, 328 prolongs lifespan in C. elegans and increases muscle
senescence in adult hippocampal neural stem/ (2019). function in rodents. Nat. Med. 22, 879–888
progenitor cells via formylpeptide receptor 2. 143. Regensburger, M., Prots, I. & Winner, B. Adult (2016).
Cell Death Dis. 4, e924 (2013). hippocampal neurogenesis in Parkinson’s disease: 171. Georgakopoulos, N. D., Wells, G. & Campanella, M.
117. Kirkland, J. L. & Tchkonia, T. Cellular senescence: impact on neuronal survival and plasticity. Neural The pharmacological regulation of cellular mitophagy.
a translational perspective. EBioMedicine 21, 21–28 Plast. 2014, 454696 (2014). Nat. Chem. Biol. 13, 136–146 (2017).
(2017). 144. Kikuchi, T. et al. Human iPS cell-​derived dopaminergic 172. Albani, D., Polito, L., Signorini, A. & Forloni, G.
118. Bussian, T. J. et al. Clearance of senescent glial cells neurons function in a primate Parkinson’s disease Neuroprotective properties of resveratrol in different
prevents tau-​dependent pathology and cognitive model. Nature 548, 592–596 (2017). neurodegenerative disorders. Biofactors 36,
decline. Nature 562, 578–582 (2018). 145. McGeer, P. L., Itagaki, S., Boyes, B. E. & McGeer, E. G. 370–376 (2010).
119. Zhang, P. et al. Senolytic therapy alleviates Reactive microglia are positive for HLA-​DR in the 173. Heilman, J., Andreux, P., Tran, N., Rinsch, C. &
Aβ-​associated oligodendrocyte progenitor cell substantia nigra of Parkinson’s and Alzheimer’s Blanco-​Bose, W. Safety assessment of urolithin A,
senescence and cognitive deficits in an Alzheimer’s disease brains. Neurology 38, 1285–1291 (1988). a metabolite produced by the human gut microbiota
disease model. Nat. Neurosci. 22, 719–728 (2019). 146. McKinnon, P. J. ATM and ataxia telangiectasia. EMBO upon dietary intake of plant derived ellagitannins
120. Musi, N. et al. Tau protein aggregation is associated Rep. 5, 772–776 (2004). andellagic acid. Food Chem. Toxicol. 108, 289–297
with cellular senescence in the brain. Aging Cell 17, 147. Walter, J. T., Alvina, K., Womack, M. D., Chevez, C. & (2017).
e12840 (2018). Khodakhah, K. Decreases in the precision of Purkinje 174. Moreira, O. C. et al. Mitochondrial function and
121. Talbot, K. et al. Demonstrated brain insulin resistance cell pacemaking cause cerebellar dysfunction and mitophagy in the elderly: effects of exercise.
in Alzheimer’s disease patients is associated with ataxia. Nat. Neurosci. 9, 389–397 (2006). Oxid. Med. Cell. Longev. 2017, 2012798 (2017).

www.nature.com/nrneurol
Reviews

175. Herranz, N. et al. mTOR regulates MAPKAPK2 188. Rees, K. et al. Non-​steroidal anti-​inflammatory drugs 201. Mak, E. et al. Cognitive deficits in mild Parkinson’s
translation to control the senescence-​associated as disease-​modifying agents for Parkinson’s disease: disease are associated with distinct areas of grey
secretory phenotype. Nat. Cell Biol. 17, 1205–1217 evidence from observational studies. Cochrane matter atrophy. J. Neurol. Neurosurg. Psychiatry 85,
(2015). Database of Systematic Reviews 9, CD008454 576–580 (2014).
176. Walters, H. E., Deneka-​Hannemann, S. & Cox, L. S. (2011). 202. Talbott, E. O., Malek, A. M. & Lacomis, D.
Reversal of phenotypes of cellular senescence by 189. US National Library of Medicine. ClinicalTrials.gov The epidemiology of amyotrophic lateral sclerosis.
pan-​mTOR inhibition. Aging 8, 231–244 (2016). https://clinicaltrials.gov/ct2/show/NCT02588677 Handb. Clin. Neurol. 138, 225–238 (2016).
177. Katila, N. et al. Metformin lowers α-​synuclein (2018). 203. Walker, F. O. Huntington’s disease. Lancet 369,
phosphorylation and upregulates neurotrophic factor 190. Petrov, D., Mansfield, C., Moussy, A. & Hermine, O. 218–228 (2007).
in the MPTP mouse model of Parkinson’s disease. ALS clinical trials review: 20 years of failure. are we 204. Subramaniam, S., Sixt, K. M., Barrow, R. & Snyder, S. H.
Neuropharmacology 125, 396–407 (2017). any closer to registering a new treatment? Front. Rhes, a striatal specific protein, mediates mutant-​
178. Ou, Z. et al. Metformin treatment prevents amyloid Aging Neurosci. 9, 68 (2017). huntingtin cytotoxicity. Science 324, 1327–1330
plaque deposition and memory impairment in APP/ 191. Vaiserman, A. M., Lushchak, O. V. & Koliada, A. K. (2009).
PS1 mice. Brain. Behav. Immun. 69, 351–363 (2018). Anti-​aging pharmacology: promises and pitfalls. 205. Duff, K. et al. Psychiatric symptoms in Huntington’s
179. Naylor, R. M., Baker, D. J. & van Deursen, J. M. Ageing Res. Rev. 31, 9–35 (2016). disease before diagnosis: the Predict-​HD study.
Senescent cells: a novel therapeutic target for aging 192. Hernandez-​Camacho, J. D., Bernier, M., Lopez-​Lluch, G. Biol. Psychiatry 62, 1341–1346 (2007).
and age-​related diseases. Clin. Pharmacol. Ther. 93, & Navas, P. Coenzyme Q10 supplementation in aging 206. Zweig, Y. R. & Galvin, J. E. Lewy body dementia:
105–116 (2013). and disease. Front. Physiol. 9, 44 (2018). the impact on patients and caregivers. Alzheimers
180. Krimpenfort, P. & Berns, A. Rejuvenation by 193. Li, T. & Chen, Z. J. The cGAS-​cGAMP-STING Res. Ther. 6, 21 (2014).
therapeutic elimination of senescent cells. Cell 169, pathway connects DNA damage to inflammation, 207. Rothblum-​Oviatt, C. et al. Ataxia telangiectasia:
3–5 (2017). senescence, and cancer. J. Exp. Med. 215, a review. Orphanet J. Rare Dis. 11, 159 (2016).
181. Chiti, F. & Dobson, C. M. Protein misfolding, amyloid 1287–1299 (2018). 208. Kleijer, W. J. et al. Incidence of DNA repair deficiency
formation, and human disease: a summary of progress 194. Giraldez-​Perez, R., Antolin-​Vallespin, M., Munoz, M. & disorders in western Europe: xeroderma pigmentosum,
over the last decade. Annu. Rev. Biochem 86, 27–68 Sanchez-​Capelo, A. Models of α-​synuclein aggregation Cockayne syndrome and trichothiodystrophy.
(2017). in Parkinson’s disease. Acta Neuropathol. Commun. 2, DNA Repair 7, 744–750 (2008).
182. Jack, C. R. Jr. et al. NIA-​AA research framework: 176 (2014).
toward a biological definition of Alzheimer’s disease. 195. Reitz, C. & Mayeux, R. Alzheimer disease: Acknowledgements
Alzheimers Dement. 14, 535–562 (2018). epidemiology, diagnostic criteria, risk factors and The authors’ research is supported by the Intramural
183. Medina, M. An overview on the clinical development biomarkers. Biochem. Pharmacol. 88, 640–651 Research Program of the NIH National Institute on Aging. The
of tau-​based therapeutics. Int. J. Mol. Sci. 19, 1160 (2014). authors thank B. Yang and N. B. Fakouri for critical reading of
(2018). 196. Vina, J. & Lloret, A. Why women have more the manuscript. The Bohr laboratory receives nicotinamide
184. Braak, H. & Braak, E. Evolution of the neuropathology Alzheimer’s disease than men: gender and riboside as a gift from ChromaDex.
of Alzheimer’s disease. Acta Neurol. Scand. Suppl. mitochondrial toxicity of amyloid-​β peptide.
165, 3–12 (1996). J. Alzheimers Dis. 20, S527–S533 (2010). Author contributions
185. Cummings, J., Lee, G., Ritter, A. & Zhong, K. 197. Luchsinger, J. A. et al. Aggregation of vascular risk All authors researched data for the article, Y.H., X.D., M.B.,
Alzheimer’s disease drug development pipeline: 2018. factors and risk of incident Alzheimer disease. Y. W., D.L.C. and V.A.B. contributed substantially to the dis-
Alzheimers Dement. 4, 195–214 (2018). Neurology 65, 545–551 (2005). cussion of content, Y.H., X.D., M.B., Y.W., S.G.H., D.L.C. and
186. Cao, B. et al. Comparative efficacy and acceptability 198. Grant, W. B., Campbell, A., Itzhaki, R. F. & Savory, J. V.A.B. wrote the article, and Y.H. and V.A.B reviewed
of antidiabetic agents for Alzheimer’s disease and mild The significance of environmental factors in the and edited the manuscript before submission.
cognitive impairment: a systematic review and etiology of Alzheimer’s disease. J. Alzheimers Dis. 4,
network meta-​analysis. Diabetes Obes. Metab. 20, 179–189 (2002). Competing interests
2467–2471 (2018). 199. Kalia, L. V. & Lang, A. E. Parkinson’s disease. Lancet The authors declare no competing interests.
187. Miguel-​Alvarez, M. et al. Non-​steroidal anti-​ 386, 896–912 (2015).
inflammatory drugs as a treatment for Alzheimer’s 200. Weintraub, D. & Stern, M. B. Psychiatric complications Publisher’s note
disease: a systematic review and meta-​analysis of in Parkinson disease. Am. J. Geriatr. Psychiatry 13, Springer Nature remains neutral with regard to jurisdictional
treatment effect. Drugs Aging 32, 139–147 (2015). 844–851 (2005). claims in published maps and institutional affiliations.

Nature Reviews | Neurology

You might also like