Skip to main content
. 2017 Sep 28;6:1779. [Version 1] doi: 10.12688/f1000research.12110.1

Table 1. Hallmarks of aging in comparison with Werner syndrome.

Aging hallmarks Brief description Werner syndrome (WS) Reference for WS
Genome
instability
Alteration to the genetic information over time
due to DNA damage and defective DNA repair
mechanisms. Genomic instability affects overall
functions of the cell.
Patient cells show gross genomic
instability. WRN-deficient cells display
large deletions.
Salk et al. 8
Chen et al. 9
Telomere
attrition
Progressive decrease in telomere length over
multiple cell divisions. Telomere attrition mainly
occurs owing to the end-replication problem and
the lack of telomerase enzyme.
WRN interacts with Pot1 and TRF2
components of the shelterin complex
to promote telomere maintenance.
Telomere length in older patients with WS
(40–60 years) is markedly shorter than
in younger patients with WS (~30 years)
and age-matched non-WS individuals.
Opresko et al. 11
Ishikawa et al. 10
Tokita et al. 72
Epigenetic
alterations
Involves alterations in the DNA methylation patterns,
post-translational modification of histones, and
chromatin remodeling
Patients with WS show an increased
DNA methylation age with an average
of 6.4 years. WRN interacts with
methylation complex consisting of
SUV39H1, HP1α, and LAP2β, which is
responsible for the epigenetic histone
mark H3K9 trimethylation (H3K9me3). In
response to DNA damage, WRN recruits
chromatin assembly factor 1 (CAF-1) to
alter chromatin structure.
Maierhofer et al. 12
Jiao et al. 78
Zhang et al. 14
Loss of
proteostasis
Impairment of protein homeostasis due to
accumulation of misfolded proteins and
deregulation of proteolytic system. Chronic
expression of misfolded, unfolded, or aggregation
of proteins contributes to the development of age-
related pathologies such as Alzheimer’s disease
and cataracts.
Cataracts are one of the most common
features observed in patients with WS.
WRN expression is severely affected
by promoter hypermethylation in age-
related cataract lens cells.
Zhu et al. 15
Mitochondrial
dysfunction
Reduction in the biogenesis of mitochondria and
mitophagy. Reduced ATP production coupled
with increased electron leakage. Oxidation of
mitochondrial proteins.
WS cells show increased reactive
oxygen species (ROS) production.
Hepatocytes of Wrn (Δhel/Δhel) mice
have decreased mitochondria and show
altered mitochondrial functions.
Cogger et al. 16
Cellular
senescence
Stable arrest of the cell cycle coupled with
stereotyped phenotypic changes such as
the accumulation of persistent DNA damage,
senescence-associated β-galactosidase, p16 INK4A,
and/or telomere shortening
Cellular senescence is a striking feature
of WS patient cells. WRN deficiency
increased the accumulation of persistent
DNA damage, p16, and senescence-
associated β-galactosidase.
Norwood et al. 73
Lu et al. 13
Deregulated
nutrient sensing
Somatotropic axis essentially consisting of growth
hormone, insulin-like growth factors (IGF-1 and
II), and their carrier proteins and receptors
regulates metabolism in mammals. In addition
to insulin–IGF-1 (IIS) signaling pathway, which
senses glucose, three interconnected nutrient
sensing systems are associated with aging. The
mechanistic target of rapamycin (mTOR) senses
high amino acid concentrations, AMPK (5′-
adenosine monophosphate [AMP]-activated protein
kinase) senses low-energy states by detecting
high AMP levels, and sirtuins sense low-energy
states by detecting high NAD + levels. With aging,
IIS pathway decreases, mTOR activity increases,
AMPK upregulates in skeletal muscles, and sirtuins
are downregulated.
WRN protects against starvation-induced
autophagy. Further research is required
to elaborate the role of WRN in regulating
nutrient-sensing mechanisms.
Maity et al. 17
Stem cell
exhaustion
A decline in the proliferation of stem and progenitor
cells, which are required for tissue regeneration
WRN-deficient mesenchymal stem cells
showed progressive disorganization
of heterochromatin and premature
senescence.
Zhang et al. 14
Altered
intercellular
communication
Enhanced activation of nuclear factor kappa
B (NF-κB) and increased production of tumor
necrosis factor (TNF), interleukin-1 beta (IL-1β), and
cytokines resulting in age-associated alteration
in intercellular communication. Accumulation of
pro-inflammatory tissue damage, failure of immune
system to clear pathogens and dysfunctional host
cells, and occurrence of defective autophagy
response. Bystander effect in which senescent cells
induce senescence in neighboring cells via gap
junction–mediated cell-cell contacts and ROS.
Patients with WS have elevated
levels of inflammation-driven aging-
associated cytokines (IL-4, IL-6, IL-10,
granulocyte macrophage colony-
stimulating factor [GM-CSF], IL-2, TNF-
α, interferon gamma [IFNγ], monocyte
chemoattractant protein-1 [MCP-1], and
granulocyte colony-stimulating factor [G-
CSF]) compared with normal individuals.
Goto et al. 18