Anda di halaman 1dari 9

REVIEWS

Ageing and the pathogenesis


of osteoarthritis
Richard F. Loeser1, John A. Collins1 and Brian O. Diekman2
Abstract | Ageing-associated changes that affect articular tissues promote the development of
osteoarthritis (OA). Although ageing and OA are closely linked, they are independent processes.
Several potential mechanisms by which ageing contributes to OA have been elucidated.
This Review focuses on the contributions of the following factors: age-related inflammation
(also referred to as ‘inflammaging’); cellular senescence (including the senescence-associated
secretory phenotype (SASP)); mitochondrial dysfunction and oxidative stress; dysfunction in
energy metabolism due to reduced activity of 5ʹ‑AMP-activated protein kinase (AMPK), which is
associated with reduced autophagy; and alterations in cell signalling due to age-related changes
in the extracellular matrix. These various processes contribute to the development of OA by
promoting a proinflammatory, catabolic state accompanied by increased susceptibility to cell
death that together lead to increased joint tissue destruction and defective repair of damaged
matrix. The majority of studies to date have focused on articular cartilage, and it will be important
to determine whether similar mechanisms occur in other joint tissues. Improved understanding
of ageing-related mechanisms that promote OA could lead to the discovery of new targets for
therapies that aim to slow or stop the progression of this chronic and disabling condition.

Telomere attrition
A number of risk factors for the development of osteo­ replacement. Improved understanding of how ageing
The shortening and arthritis (OA) exist, including prior joint injury, obesity, contributes to the development of OA could lead to new
deterioration of the genetics, sex, and anatomical factors related to joint therapies that slow or stop the progression of the disease,
protective caps on the ends shape and alignment; however, the most prominent risk which would have a major impact on public health.
of chromosomes that is
factor is increasing age1. A Spanish study published in OA that occurs in young adults is most often
associated with ageing.
2014, which included more than 3 million individuals, caused by a prior joint injury, a process known as post-­
examined the incidence of clinically diagnosed OA traumatic OA1, whereas in older adults a number of
and reported that incident hand OA in women peaked factors related to ageing can contribute to the develop­
between the ages of 60 and 64 years, whereas that of ment of OA (BOX 1). These ‘ageing factors’ probably
1
Thurston Arthritis Research the hip and knee continued to increase with increasing work in concert with other OA risk factors. Important
Center, Division of
age2. A US study using data from the National Health differences between joint ageing and OA demonstrate
Rheumatology, Allergy, and
Immunology, 3300 Thurston Interview Survey reported that incident symptomatic that they are distinct processes (BOX 2). Although OA is
Building, Campus Box 7280, knee OA peaked between the ages of 55 and 64, whereas a condition that affects the entire joint 7 and results in
University of North Carolina prevalent disease increased with age such that at age joint failure, the majority of research to date has focused
School of Medicine, 85 years and older, prevalence ranged from ~13% in on ageing-associated changes in the articular cartilage.
Chapel Hill, North Carolina
27599–7280, USA.
nonobese men to 32% in obese women3. However, studies on the meniscus8, anterior cruciate
2
Lineberger Comprehensive Musculoskeletal conditions, including OA, are a ligament 9 and bone10 have shown age-related changes
Cancer Center, University major cause of disability worldwide4 and have a substan­ similar to those observed in articular cartilage — includ­
of North Carolina School of tial contribution to healthcare costs, accounting for an ing loss of cellularity, and disruption and degeneration
Medicine, 450 West Drive,
estimated 1.0–2.5% of the gross domestic product in the of the extracellular matrix — suggesting that common
Campus Box 7295,
Chapel Hill, North Carolina USA, Canada, the UK, France and Australia5. Advanced processes could be involved.
27599–7295, USA. OA often requires joint replacement to reduce pain and In 2013, nine cellular and molecular hallmarks of
Correspondence to R.F.L. disability, and the number of knee replacement surgeries ageing were proposed11 to highlight the underlying causes
richard_loeser@med.unc.edu has substantially increased over the past 20 years6. The of age-related dysfunction and assist with research into
doi:10.1038/nrrheum.2016.65 ageing of our population will compound the number potential therapeutic interventions. These hallmarks
Published online 19 May 2016 of older adults disabled by OA and in need of joint include genomic instability, telomere attrition, epigenetic

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points published elsewhere22,23 have detailed the potential


mechanisms by which obesity leads to OA, and so here
• Ageing-associated changes promote the development of osteoarthritis (OA), we will only summarize the key findings most relevant
but ageing and OA are independent processes to age-related OA. The increase in fat mass with ageing
• Several hallmarks of ageing could contribute to OA: genomic instability, telomere is associated with increased numbers of adipocytes
attrition, epigenetic alterations, loss of proteostasis, dysregulated nutrient sensing, and proinflammatory macrophages in adipose tissue,
mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered and these cells produce a number of cytokines and adi­
intercellular communication
pokines that could contribute to OA24,25, although the
• The increase in fat mass and related metabolic changes that occur with ageing can importance of these mediators in OA remains unclear.
result in ageing-related inflammation (referred to as ‘inflammaging’), a chronic
Obesity and increased fat mass can also contribute to
low-grade systemic proinflammatory state
OA through metabolic alterations that are sometimes
• Elevated levels of reactive oxygen species can contribute to OA by causing oxidative
referred to as ‘meta-inflammation’ (REFS 22,23). In addi­
damage and disrupting normal cell signalling, leading to imbalanced anabolic and
catabolic activity and ultimately cell death
tion to increased levels of cytokines and adipokines,
meta-inflammation is associated with increased levels
• Chondrocytes can undergo cellular senescence with age and OA in response to
growth signals released as a result of underlying cellular damage
of circulating free fatty acids, hyperglycaemia and oxi­
dative stress, which can all negatively influence joint
• The non-enzymatic crosslinking of collagen that occurs with ageing alters the
tissues to promote matrix destruction22,23. Furthermore,
mechanical properties of cartilage, and the resulting changes to mechanotransduction
pathways reduce extracellular matrix synthesis by chondrocytes obesity associated with low muscle mass (that is, sarco­
penic obesity) has been associated with knee OA26 and
could contribute to joint instability and an increased risk
of falls in older adults, particularly in women27.
alterations, loss of proteostasis, dysregulated nutrient In addition to an age-related increase in visceral fat,
sensing, mitochondrial dysfunction, cellular senescence, local fat depots such as the infrapatellar fat pad increase
stem cell exhaustion and altered intercellular communica­ with age28. Given that this fat pad is proximal to the knee
tion. This Review covers selected aspects of ageing that joint and produces the adipokines adiponectin and lep­
are relevant to OA and, where possible, relates these tin, as well as basic fibroblast growth factor (bFGF; also
studies to the hallmarks of ageing. known as FGF2), vascular endothelial growth factor,
TNF and IL‑6 (REF. 29), it is plausible that an age-related
Proteostasis Ageing, inflammation and OA increase in fat pad volume could contribute to OA.
A state of protein homeostasis Mounting evidence suggests that OA is associated with However, in vitro studies of conditioned media from
in which properly folded low-grade systemic and local inflammation12,13. Ageing infrapatellar fat pads removed from patients with end-
proteins are stabilized
and damaged proteins
has likewise been associated with chronic low-grade stage knee OA found a protective rather than catabolic
are degraded. inflammation, sometimes referred to as ‘inflammaging’ effect on bovine cartilage explants30.
(REF. 14), which could promote OA, although studies to
Cellular senescence date have not identified the precise mechanisms. Several Cellular senescence and the SASP
A phenotypic state that can
of the hallmarks of ageing could also have a role in OA, Cellular senescence is one of the hallmarks of ageing,
emerge in response to in vitro
or in vivo cellular stress. including epigenetic alterations, dysregulated nutrient and chondrocytes have many features that are character­
Senescent cells do not divide sensing, mitochondrial dysfunction, cellular senescence istic of senescent cells during ageing and during OA31,32
upon stimulation and often and altered intercellular communication. As discussed (FIG. 1). The observation that OA catalyses the develop­
display an altered secretory further below, increased production of proinflamma­ ment of senescence indicates that cellular stresses prob­
profile along with increased
lysosomal activity.
tory mediators is a feature of the senescence-associated ably have a key role in establishing this phenotypic state.
secretory phenotype (SASP) and could be an important A key challenge has been deciding how to best define
Stem cell exhaustion mechanism in OA. chondrocyte senescence and determine the underlying
The inability of stem cells to A key cytokine associated with ageing and age-related mechanisms that might be targeted therapeutically.
provide a sufficient number
disease is IL‑6. Levels of IL‑6 in the systemic circulation Morphological changes and permanent replicative arrest
of differentiated cells to
maintain normal tissue increase with age15,16 and are strongly associated with after monolayer expansion have defined cellular senes­
function. Premature stem cell the risk of OA progression17,18. IL‑6 is important in the cence for in vitro settings, but defining cellular senescence
exhaustion can occur under pathogenesis of rheumatoid arthritis, and IL‑6 inhibition in vivo has proved more difficult33.
conditions of high demand for is an effective therapy approved for clinical use in this Two distinguishing characteristics of senescent cells
progenitors or when aberrant
signalling interrupts the
setting 19. However, a causal role for IL‑6 in age-related are stress-induced permanent proliferative arrest and
maintenance of quiescence. OA has not been established, as mice with deletion of resistance to both mitogenic and oncogenic stimuli33,
the Il6 gene have more severe (rather than less severe) both of which are challenging to assess in chondro­
Senescence-associated age-related OA20, suggesting that other mediators could cytes. Chondrocytes exhibit low proliferation rates,
secretory phenotype
be involved. In addition, the levels of multiple proinflam­ which hinders the measurement and interpretation of
(SASP). A cellular phenotype
that is characterized by the matory and anti-inflammatory mediators change with the functional impact of a further reduction in prolif­
production of high levels of age16, and so it is unlikely that the association between eration34. Lineage-tracing experiments in mice indicate
certain inflammatory cytokines age and OA is driven by a single factor. that the proliferation of particular chondrocyte sub­
and matrix metalloproteinases. Inflammaging is probably attributable, at least in part, populations could help to maintain cartilage tissue35,
The SASP represents a
mechanism by which senescent
to the age-related increase in visceral fat mass that is asso­ suggesting, therefore, that the loss of proliferation in
cells may function in a ciated with a decline in muscle mass21. Obesity is a well- these cells due to senescence could contribute to OA
non-autonomous fashion. accepted risk factor for OA in all age-groups3. Reviews progression. The other defining feature of senescence

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 1 | Age-related factors that contribute to osteoarthritis development other than chondrocytes, such as cells in the meniscus,
synovium or bone7. Future work is needed to determine
• Reduced muscle mass and increased fat mass alter joint loading and are associated whether any of these cell types display other features
with an increase in adipokine and cytokine production, resulting in low-grade of senescence.
systemic inflammation109.
• Changes in the extracellular matrix — including the accumulation of advanced The induction of chondrocyte senescence
glycation end-products, reduced aggrecan size, reduced hydration and increased A detailed understanding of how chondrocytes enter
collagen cleavage — alter the mechanical properties of cartilage and make it more
the senescent state might enable the development of
susceptible to degeneration31.
therapies designed to prevent such a phenotypic switch
• Extracellular matrix disruption and reduced cell density in the meniscus and
during OA. One model proposes that senescence occurs
ligaments promote degeneration and can potentially alter joint mechanics8,9.
during conditions characterized by simultaneous signals
• The function of subchondral bone is impaired due to reduced numbers of osteocytes
for cell cycle arrest and cell growth7,42, and aged cartilage
and altered mineral composition10.
tissue has the potential to provide both these cues.
• Mitochondrial dysfunction, oxidative stress and reduced autophagy in
The cumulative DNA damage and oxidative stress
chondrocytes alters their function, promoting catabolic processes and cell death
over anabolic processes31.
that occurs with ageing and in the OA tissue micro­
environment alters gene expression patterns in chon­
drocytes7,43 and could cause telomere attrition due to
is a beneficial function of preventing the division of targeted DNA damage41,44. Notably, stem cells and post­
transformed cells36. Chondrosarcomas typically derive mitotic cells such as neurons and chondrocytes can be
from the growth plate and perichondrium but not from particularly susceptible to the accumulation of cellular
articular cartilage37, suggesting that articular cartilage damage during the long interval between replicative
might be inherently unlikely to undergo uncontrolled events45,46. One common consequence of accumulating
growth. This characteristic presents a challenge for the DNA damage is the upregulation of cell cycle inhibitors,
identification of senescent chondrocytes on the basis of such as p16INK4A and p21, which mediate the stable arrest
an increased resistance to oncogenic transformation. that is associated with senescence33. Chondrocytes from
Cellular senescence could have other functional roles patients with OA have increased p16INK4A expression
aside from inhibition of proliferation, as the SASP is compared with those from age-matched controls, pos­
defined by the production of high levels of proinflamma­ sibly due to a reduction in expression of the microRNA
tory cytokines and matrix-degrading enzymes38. Indeed, miR‑24 (REF. 47). Indeed, knocking down miR-24 led to
the SASP seems to be regulated independently from cell increased expression of p16INK4A in an in vitro pellet cul­
cycle arrest 39,40. Some of the most highly upregulated ture model47. These observations in chondrocytes seem
SASP-related factors, such as IL‑1α, IL‑6 and monocyte to be aligned with a model explored in mouse muscle
chemoattractant protein 1 (MCP1; also known as CC stem cells, in which increased p16INK4A expression with
chemokine ligand 2 (CCL2)) are found in OA cartilage41, age made cells more likely to undergo senescence during
hindering efforts to determine whether the level of a subsequent proliferation after injury 48.
particular mediator is due to senescence or a result of OA. The second signal required for establishing senescence,
The cell type responsible for producing these cytokines is a strong growth signal, can be provided in the form of
also difficult to determine. As these factors are also found growth factors that are either directly released from dam­
in the synovial fluid, they could be produced by cells aged cartilage or synthesized at high levels by chondrocytes
during OA. For example, bFGF is released from damaged
cartilage tissue49 and can be detected in focal clusters of
Box 2 | Differences between normal joint ageing and osteoarthritis chondrocyte proliferation that develop near areas of
• With normal joint ageing, articular cartilage remains intact but loses thickness and cartilage damage50. As a downstream result of signalling
has a reduced glycosaminoglycan (GAG) content. With osteoarthritis (OA), fibrillation by inflammatory cytokines associated with OA, expression
of the cartilage surface occurs in focal areas and can be associated with a complete of bone morphogenetic protein 2 (BMP2) is increased51,
loss of staining for GAGs31,110. thereby stimulating enhanced turnover of extracellular
• Non-enzymatic crosslinking of collagen by advanced glycation end-products matrix 52. Although their competence for proliferation
(AGEs)  increases in cartilage with age101. A mouse model of injury-induced OA indicates that these chondrocytes are not senescent, cells
demonstrated that collagen crosslinking occurs through a distinct mechanism that enter the cell cycle in the context of potentially dam­
involving lysyl oxidase104. aging stimuli have an increased likelihood of entering
• The density of chondrocytes in cartilage decreases with age, but chondrocyte senescence as opposed to returning to quiescence53,54.
‘clusters’ emerge during the development of OA near sites of tissue damage and Observations that senescence occurs after monolayer
may indicate attempted repair or altered cellular signals31,110. expansion of chondrocytes (reviewed elsewhere55) provide
• Aged chondrocytes have reduced levels of extracellular matrix gene expression and further evidence that contexts of increased proliferation
synthesis, whereas during OA chondrocytes become highly active with increases correlate with emergence of the senescent phenotype.
in both anabolic processes (for example, matrix synthesis) and catabolic pathways
(for example, those induced by inflammatory cytokines)31,110.
The role of circadian signals
• Synovial inflammation and hypertrophy occur in OA but have not been described Also of potential interest is the finding that chondrocytes
in normal joint ageing7.
have an intrinsic circadian clock which is disrupted in
• Bone mass and density decrease with ageing, whereas subchondral bone thickening cartilage from aged mice56. A decline in expression of the
is seen in patients with OA7.
circadian clock gene BMAL1 (also known as ARNTL) has

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Growth signals: Arrest signals: that ERCC1‑mediated DNA repair helped to restrain
• Growth factors released from the • DNA damage and oxidative stress senescence in human articular chondrocytes63. A sepa­
extracellular matrix • Inflammatory joint environment rate study further supports the strategy of inhibiting
• Altered mechanical stimulation
anti-apoptotic pathways by identifying navitoclax as an
inhibitor of B‑cell lymphoma family proteins that spe­
cifically targets senescent human fibroblasts in vitro and
mouse stem cells in vivo64.
Features of senescent chondrocytes: Another therapeutic approach could be to interfere
• p16INK4A expression with inflammatory pathways that are active in senes­
• Senescence-associated β-galactosidase
• SASP cent chondrocytes. Rapamycin inhibits the translation
of SASP proteins in human fibroblasts through decreas­
ing the rate of IL‑1α translation, which is regulated by
serine/threonine-protein kinase mTOR (also known
Functional effects:
• Secretion of cytokines and MMPs as mammalian target of rapamycin)65. With increased
• Permanent cell cycle exit progress in the understanding of how senescence is reg­
in chondrocytes ulated in chondrocytes, particular cellular features could
Figure 1 | Chondrocytes exhibit features of cellular senescence in the contexts of provide other novel drug targets for OA.
ageing and osteoarthritis. Senescence is often caused by the combination of growth
and arrest signals, conditions that can occur in cartilage during
Nature ageing.
ReviewsFeatures of
| Rheumatology Oxidative stress and the mitochondria
senescence include expression of p16INK4A and positive staining for senescence-­ Mitochondrial dysfunction is a hallmark of ageing that
associated β‑galactosidase. The functional effects of chondrocyte senescence are has attracted particular attention in the context of OA.
challenging to measure because of the low proliferation rate of chondrocytes and the The free radical theory proposes that cellular damage
overlap between features of the senescence-associated secretory phenotype (SASP) and occurring as a result of excessive levels of reactive oxygen
those associated with the development of osteoarthritis (OA). Understanding the species (ROS) substantially contributes to the develop­
mechanism of senescence could yield therapeutic interventions to prevent development
ment of the ageing phenotype and to the progression of
of the SASP or specifically eliminate senescent cells from joint tissues. MMPs, matrix
metalloproteinases.
age-related diseases66. However, in addition to cellular
damage, elevated levels of ROS produced as a result of
age-associated oxidative stress also promote disease by
disturbing homeostatic physiological cell signalling 66–68.
been associated with stress-induced senescence in fibro­ Mitochondrial function has long been recognized
blasts57 and has been noted to occur in human OA chon­ to decline during ageing, and a causative link between
drocytes and in the cartilage of aged mice58. Mice with mitochondrial dysfunction, oxidative stress and the age­
cartilage-specific deletion of Bmal1 developed premature ing phenotype has been proposed66,69. Evidence from a
knee cartilage lesions that appeared at 2 months of age 2015 study 70 demonstrates that, compared with normal
and became progressively more severe in adult animals58. chondrocytes, chondrocytes from patients with OA
However, no other OA‑associated changes in the bone, have reduced mitochondrial mass and mitochondrial
ligaments or synovium were evident in these mice, and DNA content along with reduced levels of electron
the finding that the cartilage changes were detectable transport chain proteins and proteins involved in mito­
before the mice were skeletally mature suggests that the chondrial biogenesis. The researchers confirmed that
phenotype is the result of a developmental defect rather some of these changes occurred during ageing and were
than OA. Further work is needed to determine the role of independent of OA by analysing mouse cartilage tissue
BMAL1 and the circadian clock in age-related human OA. sections. Interestingly, protein levels of nuclear receptor
erythroid 2‑related factors 1 and 2 (also known as NRF1
Therapeutic targeting of senescence and NRF2, respectively), which confer cellular protection
One emerging therapeutic strategy for age-related dis­ through regulation of antioxidant gene expression, were
eases is to specifically kill senescent cells in order to also decreased in chondrocytes from patients with OA
prevent the detrimental secretion of SASP-related fac­ compared with those from healthy controls70.
tors. Although this approach has not yet been explored As mitochondria are an important source of ROS,
in OA, the concept builds on studies that eliminated age-related mitochondrial dysfunction that leads to
p16INK4A-positive cells in mouse models of premature59 an imbalance between the production of ROS and the
and natural ageing 60. Drugs that eliminate senescent cells antioxidant capacity of the cell has been identified as
in aged tissue would ideally mimic the efficient clearance a contributing factor in the development of OA68,71,72
of senescent cells that occurs after wound repair 61. The (FIG. 2). Mice with post-traumatic OA showed elevated
‘senolytic’ compounds dasatanib and quercetin target intracellular and mitochondrial superoxide generation,
senescent cells by inhibiting the anti-apoptotic pathways which was associated with downregulation of mito­
that are upregulated in senescent cells62. One anti-ageing chondrial superoxide dismutase 2 (SOD2) expression73.
effect of this treatment was protection from proteogly­ Furthermore, in this same model, SOD2 loss resulted
can loss in the intervertebral disc of Ercc1‑mutant mice62 in greatly increased age-related cartilage degeneration73.
(a model of accelerated ageing based on compromised In vitro studies using paraquat to induce mitochondrial
DNA damage repair). Although articular cartilage was superoxide generation in primary mouse articular chon­
not investigated in this study, other work has shown drocytes led to substantial mitochondrial dysfunction,

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Mitochondrion
Consistent with the concept that age-related oxidative
stress alters cell signalling, studies have demonstrated
an age-related disruption ofinsulin-like growth factor 1
Peroxiredoxin (IGF1) signalling in human chondrocytes that results
↓ Mitochondrial
↓ SOD2 hyperoxidation mass and number in reduced extracellular matrix gene expression and
ETC O2• H2O2 ↓ mtDNA protein synthesis77,78. In human chondrocytes from older
↓ Mitochondrial adults, this effect was associated with an increased sen­
biogenesis
↓ ETC function sitivity to oxidative stress, which resulted in inhibition
of IGF1‑mediated activation of RACα serine/threonine-
protein kinase (AKT) and increased activation of cata­
bolic mitogen-­activated protein kinase (MAPK) signalling
pathways78.
A key mechanism by which ROS regulate cell signal­
↑ ROS production ling is through oxidative post-translational modifications
↓ Antioxidant levels
of specific thiol groups in proteins that contain reactive
cysteines79. Cysteine oxidation initially results in the for­
↑ Oxidative stress mation of a cysteine sulfenic acid (Cys-SOH) in a process
known as S‑sulfenylation. In a 2016 study, chondrocytes
from patients with OA had an increased basal level of
Disturbances in signalling: S‑sulfenylation compared with those from healthy con­
↓ IGF1 signalling (via oxidative inhibition trols80. ROS induced sulfenylation of multiple chondro­
↑ Damage to lipids, of IRS1 and AKT) ↑ Chondrocyte
proteins and DNA ↑ MAPK signalling (via phosphorylation catabolism cyte proteins including the tyrosine kinase SRC, the
of p38 MAPKs, ERK1 and ERK2) activity of which promoted an increase in the production
↑ Protein thiol oxidation of matrix metalloproteinase 13 (MMP13)80. These data
suggest that ROS-induced sulfenylation of chondrocyte
proteins can alter signalling pathways that can promote
• Cartilage matrix loss cartilage degradation.
• Chondrocyte death
• Ageing phenotype In the presence of excessive levels of ROS, cysteine
oxidation can proceed from cysteine sulfenic acid to
sulfinic (Cys-SO2H) or sulfonic (Cys-SO3H) acid; this
so‑called hyperoxidation can lead to the inactivation of
Figure 2 | Mitochondrial dysfunction, oxidative stress and changes in normal cell redox-sensitive proteins. Hyperoxidation of the peroxi­
signalling in ageing and osteoarthritis. Mitochondrial dysfunction in both ageing and redoxin family of antioxidant enzymes has been dem­
Natureintegrity
osteoarthritis (OA) is characterized by reduced mitochondrial Reviews(mass,
| Rheumatology
number onstrated in cartilage samples from older adult humans
and DNA content) and impaired electron transport chain (ETC) function. These features
and patients with OA81. Conditions of oxidative stress
contribute to increased production of reactive oxygen species (ROS). Concomitant
reductions in mitochondrial antioxidant capacity — including reduced mitochondrial
(induced in vitro with the ROS generator menadione)
superoxide dismutase 2 (SOD2) levels and peroxiredoxin hyperoxidation — lead to led to an age-related increase in peroxiredoxin hyper­
enhanced oxidative stress and ROS-mediated damage in the mitochondria (indicated by oxidation, which was associated with both inhibition of
red lightning bolts). In ageing and OA, extra-mitochondrial antioxidant systems (namely, pro-survival cell signalling and p38 MAPK-induced chon­
SOD1, catalase and glutathione synthetase; not shown) are also considerably less active, drocyte cell death81. Importantly, reduction of ROS levels
which exacerbates oxidative stress. ROS cause damage to proteins, lipids and DNA, by mitochondrion-specific overexpression of the antioxi­
increase chondrocyte catabolism and, importantly, lead to disturbances in normal cell dant enzyme catalase prevented peroxiredoxin hyperoxi­
signalling. These disturbances can include inhibition of pro-survival insulin-like growth dation in vitro and reduced the severity of age-related OA
factor 1 (IGF1) signalling and increased catabolic mitogen-activated protein kinase in mice in vivo81. Collectively, these studies highlight ROS
(MAPK) signalling. Increased levels of ROS can also result in increased protein thiol
as crucial secondary signalling molecules in chondrocytes
oxidation, resulting in S‑sulfenylation or, when excessive, hyperoxidation of reactive
protein cysteines, which might contribute to altered cell signalling. mtDNA, mitochondrial
that warrant further study in the context of ageing and OA.
DNA; ERK, extracellular signal-regulated kinase; IGFR, IGF receptor; IRS1, insulin
receptor substrate 1. Dysfunctional energy metabolism
Another hallmark of ageing is dysregulated nutrient
sensing. 5ʹ‑AMP activated protein kinase (AMPK) is a key
regulator of cellular metabolism and energy balance that is
along with reduced expression of both antioxidant genes activated by stressors that enhance the cellular AMP:ATP
(including Sod2) and anabolic genes in cartilage, whereas ratio82. The activity of AMPK and its regulatory upstream
catabolic gene expression was upregulated73. These data kinase, serine/threonine-protein kinase STK11 (also
are in accordance with other studies demonstrating known as liver kinase B1 (LKB1)), is reduced in cartilage
downregulation of SOD2 expression at the mRNA and from aged mice and mice with OA, as well as in bovine
protein levels in cartilage from both humans74–76 and chondrocytes after dynamic compression-induced bio­
animals76 with OA, and support the hypothesis that mechanical injury83. Similarly, OA‑associated reductions in
mitochondrial dysfunction and the associated redox AMPK activity have been observed in human chondrocytes
imbalance is a key mechanism contributing to cartilage and cartilage84, and this reduced activity has been associated
degeneration and the pathogenesis of OA. with substantially reduced mitochondrial biogenesis70.

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Importantly, AMPK might also modulate key homeo­ protein level, and lentivirus-induced overexpression of
static signalling pathways through regulation of autophagy, SIRT6 in the knee joint protects young mice from surgi­
a cellular process that removes damaged and dysfunctional cally induced OA99. These studies highlight a potential
organelles and proteins82,85. Autophagy is relevant to role for SIRT6 in cartilage homeostasis that warrants
another hallmark of ageing: loss of proteostasis. Transgenic further investigation.
overexpression of AMPK delays the onset of the ageing
phenotype through direct upregulation of autophagy in Changes in the extracellular matrix
Drosophila melanogaster, which ultimately increases the Articular cartilage is a load-bearing tissue, and the ability
lifespan of these organisms86. Consistent with this obser­ of chondrocytes to sense and respond to mechanical
vation, the expression of key autophagy proteins is consid­ signals is essential for maintaining joint homeostasis
erably reduced in cartilage from aged mice compared with (reviewed elsewhere100). Accumulation of advanced
that from young mice, and these changes are associated glycation end-products (AGEs) with ageing causes non-­
with increased levels of apoptosis and cartilage degener­ enzymatic collagen crosslinking that directly alters the
ation87. Similar results have also been found in a mouse mechanical properties of the extracellular matrix 101.
model of post-traumatic OA88, implicating dysfunctional However, a canine model using ribose and threose injec­
autophagy as a key mechanism in both ageing and OA. tions to increase AGE levels in young animals to match
Although AMPK levels were not measured in these those of aged controls was insufficient to initiate OA102.
post-traumatic OA studies, other work has demonstrated Nevertheless, chondrocytes did synthesize fewer proteo­
that AMPK signalling stimulates autophagy in chon­ glycans in the highly crosslinked tissue, which might
drocytes89. As inhibition of mTOR signalling is a widely indicate that compromised cellular function would
investigated strategy to prevent age-related disease90, the prevent sufficient tissue maintenance in the context of
fact that cartilage-specific loss of mTOR increased AMPK ageing or injury. This possibility is supported by a study
levels and autophagy, and was sufficient to protect mice in rabbits, in which forced exercise in combination with
from OA following surgery, is of particular interest 91. ribose injections did induce OA, through AGE-mediated
Taken together, a plausible interpretation of these findings suppression of protective chondrocyte signalling 103.
is that age-related and OA‑related reductions in AMPK Whether limiting AGE accumulation would be suf­
signalling reduce autophagy and contribute to the cellular ficient to prevent age-related OA remains unclear, but
dysfunction observed in OA cartilage, a connection that genetic deletion of lysyl oxidase (an enzymatic collagen
is worthy of future investigation. crosslinker that is upregulated by joint injury) protected
In addition to AMPK, many lines of evidence suggest mice from injury-induced OA104. The potential relevance
that the evolutionarily conserved sirtuin (SIRT) family of this finding to AGEs and thus ageing was suggested by
of NAD+-dependent deacetylase proteins also contrib­ the finding that collagen crosslinking induced by AGEs
utes to cellular homeostasis through regulation of energy and by lysyl oxidase had similar effects on chondrocyte
balance. This regulation is achieved via direct nutrient signalling in collagen gels and on the development of
sensing, which can contribute to increased lifespan in OA after injury.
model organisms, such as worms and flies92. However, Chondrocytes integrate signals from mechanotrans­
whether overexpression of sirtuins can have a direct duction pathways that sense changes in the extracellular
longevity-promoting effect remains unclear 93. SIRT6 has matrix and those initiated by soluble factors, and both
been identified as a crucial regulator of processes that are types of signal are affected by ageing. Mechanical com­
hypothesized to be directly involved in ageing, including pression of bovine cartilage stimulates increased expres­
metabolic homeostasis, genome stability and transcrip­ sion of the gene encoding transforming growth factor‑β
tion94. Sirt6‑knockout mice have reduced IGF1 levels (TGFβ) and the subsequent phosphorylation of SMAD2
and accelerated ageing-like degenerative processes that and SMAD3 through TGFβ receptor type 1 (TGFR-1),
ultimately lead to premature death, which suggests a role which increases matrix synthesis and limits catabolic
for SIRT6 in ageing 95. Similarly, male transgenic mice signalling 105. However, ageing causes a significant loss
overexpressing Sirt6 had an increased lifespan, in part in expression of TGFR1, resulting in a shift that favours
due to regulation of IGF1 signalling 96, which suggests TGFβ signalling through serine/threonine-protein
that age-related loss of SIRT6 activity could contribute kinase receptor R3 (SKR3; also known as TGFβ super­
to ageing by disturbing normal cell signalling pathways. family receptor type I). SKR3‑mediated signalling leads
Advanced glycation
In cartilage, research has predominantly focused on to phosphorylation of SMAD1, SMAD5 and SMAD8,
end-products the role of SIRT1 (reviewed elsewhere97). As such, there which is associated with an increase in catabolic signal­
(AGEs). A diverse set of is a paucity of research on how other sirtuins regulate ling and increased production of MMP13 (REF. 106). In a
molecules formed by chondrocyte function. However, depletion of SIRT6 in 2016 study of bovine cartilage explants subjected to
non-enzymatic reaction of
human chondrocytes (achieved using RNA interference) anabolic dynamic compression, tissues from aged animals
proteins with reducing sugars
such as glucose and ribose. resulted in substantially increased DNA damage and had deficient phosphorylation of SMAD2 and SMAD3
telomere dysfunction, which was associated with pre­ in response to loading compared with explants from
Anabolic dynamic mature senescence98. SIRT6 depletion also increased young animals105. The exact mechanism responsible for
compression expression of MMP1 and MMP13, which are implicated this effect is difficult to deduce because, compared with
A process by which physical
compression of cells within a
in the pathogenesis of OA. Compared with cells from young explants subjected to the same load, aged tissue
tissue promotes an anabolic normal human tissue, human OA chondrocytes show showed less deformation, exhibited less upregulation of
response by the cells. reduced expression of SIRT6 at both the mRNA and growth factors and also had lower levels of TGFR1 on

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 3 | Therapeutic targeting of ageing-related processes in osteoarthritis function. These changes promote catabolic activity over
anabolic activity and eventually result in cell death. We
• Interventions designed to extend ‘healthspan’ by targeting systemic ageing- can easily envision how these changes could result in
related processes could potentially delay or prevent various chronic diseases, joint tissue degeneration and matrix loss, but the precise
including osteoarthritis111,112. mechanisms need to be clarified. The concept of hall­
• Senolytics, such as dasatanib and quercetin, are being designed to specifically kill marks of ageing suggests that common mechanisms will
senescent cells; removal of these cells might alleviate their harmful effects on drive dysfunction in various tissues and organ systems
neighbouring cells, including the release of proinflammatory factors that characterize
that are most affected by ageing. These hallmarks repre­
the senescence-associated secretory phenotype (SASP)62.
sent areas for further research into elucidating the con­
• Agents that activate 5ʹ‑AMP-activated protein kinase (AMPK), such as metformin,
nections between ageing and OA. Although the majority
could restore physiological levels of autophagy and promote proteostasis112.
of research into the effects of ageing continues to focus
• Sirtuin activators could restore normal nutrient sensing and energy metabolism112.
on articular chondrocytes, further studies are needed to
• Antioxidants targeted to the mitochondria or designed to restore homeostatic determine whether similar mechanisms are at play in
redox signalling could counteract mitochondrial dysfunction and altered
other cells of the joint that are affected by OA, in order
intracellular signalling113.
to find common targets for intervention.
A primary goal of ageing research is to better under­
stand common mechanisms that could be targeted with
the cell surface. However, previous work using a hydrogel the intent of delaying loss of function in more than a
system with varied stiffness revealed that mouse chondro­ single system, which would result in improved ‘healthspan’
cytes show a maximal response to exogenous TGFβ at a rather than simply extending lifespan. Heterochronic
physiologically relevant stiffness of 500 kPa107, indicating parabiosis experiments, in which the circulatory systems
that the age-related increase in matrix stiffness probably of young and old animals are joined, have revealed sys­
contributes to the reduced phosphorylation of SMAD2 temic factors that alter the ageing phenotype, including
and SMAD3 in aged tissue. growth and differentiation factor 11 (GDF11), oxytocin
and IL‑15, which might be able to reduce ageing in mul­
Conclusions tiple organ systems108. An important aim of future studies
Researchers are becoming increasingly interested in will be to determine whether these ageing-associated
elucidating the mechanisms by which ageing promotes factors influence the development of OA.
the development and progression of OA. Studies in the Another emerging approach is the development of
early 2000s detailed age-related changes in the extracell­ senolytic agents that would target and remove senescent
ular matrix, such as the accumulation of AGEs, which cells62,64. These and other therapies that target ageing
could promote OA by altering the mechanical proper­ processes could be developed into novel treatments for
ties of joint tissues. Subsequent research has focused OA (BOX 3). Given the prevalence of conditions such as OA
on age-related cellular changes that include not only an in older adults and the importance of the musculoskeletal
accumulation of damaged proteins, lipids and DNA, but system in physical function, achieving improvements in
also alterations in mitochondrial function, levels of ROS healthspan will certainly require interventions that benefit
and energy metabolism that disrupt cell signalling and the musculoskeletal system.

1. Johnson, V. L. & Hunter, D. J. The epidemiology of 10. Busse, B. et al. Decrease in the osteocyte lacunar 19. Smolen, J. S. et al. Effect of interleukin‑6 receptor
osteoarthritis. Best Pract. Res. Clin. Rheumatol. 28, density accompanied by hypermineralized lacunar inhibition with tocilizumab in patients with rheumatoid
5–15 (2014). occlusion reveals failure and delay of remodeling arthritis (OPTION study): a double-blind, placebo-
2. Prieto-Alhambra, D. et al. Incidence and risk factors in aged human bone. Aging Cell 9, 1065–1075 controlled, randomised trial. Lancet 371, 987–997
for clinically diagnosed knee, hip and hand (2010). (2008).
osteoarthritis: influences of age, gender and 11. Lopez-Otin, C., Blasco, M. A., Partridge, L., 20. de Hooge, A. S. et al. Male IL‑6 gene knock out mice
osteoarthritis affecting other joints. Ann. Rheum. Dis. Serrano, M. & Kroemer, G. The hallmarks of aging. developed more advanced osteoarthritis upon aging.
73, 1659–1664 (2014). Cell 153, 1194–1217 (2013). Osteoarthritis Cartilage 13, 66–73 (2005).
3. Losina, E. et al. Lifetime risk and age at diagnosis of 12. Attur, M. et al. Low-grade inflammation in 21. Delmonico, M. J. et al. Longitudinal study of muscle
symptomatic knee osteoarthritis in the US. Arthritis symptomatic knee osteoarthritis: prognostic value strength, quality, and adipose tissue infiltration.
Care Res. (Hoboken) 65, 703–711 (2013). of inflammatory plasma lipids and peripheral blood Am. J. Clin. Nutr. 90, 1579–1585 (2009).
4. Vos, T. et al. Years lived with disability (YLDs) for 1160 leukocyte biomarkers. Arthritis Rheumatol. 67, 22. Wang, X., Hunter, D., Xu, J. & Ding, C. Metabolic
sequelae of 289 diseases and injuries 1990–2010: 2905–2915 (2015). triggered inflammation in osteoarthritis. Osteoarthritis
a systematic analysis for the Global Burden of Disease 13. Scanzello, C. R. & Loeser, R. F. Inflammatory activity in Cartilage 23, 22–30 (2015).
Study 2010. Lancet 380, 2163–2196 (2012). symptomatic knee osteoarthritis: not all inflammation 23. Courties, A., Gualillo, O., Berenbaum, F. & Sellam, J.
5. March, L. M. & Bachmeier, C. J. Economics of is local. Arthritis Rheumatol. 67, 2797–2800 (2015). Metabolic stress-induced joint inflammation
osteoarthritis: a global perspective. Baillieres Clin. 14. Franceschi, C. et al. Inflamm-aging: an evolutionary and osteoarthritis. Osteoarthritis Cartilage 23,
Rheumatol. 11, 817–834 (1997). perspective on immunosenescence. Ann. NY Acad. Sci. 1955–1965 (2015).
6. Cram, P. et al. Total knee arthroplasty volume, 908, 244–254 (2000). 24. Sellam, J. & Berenbaum, F. Is osteoarthritis a
utilization, and outcomes among Medicare 15. Ershler, W. B. Interleukin‑6: a cytokine for metabolic disease? Joint Bone Spine 80, 568–573
beneficiaries, 1991–2010. JAMA 308, 1227–1236 gerontologists. J. Am. Geriatr. Soc. 41, 176–181 (2013).
(2012). (1993). 25. Guilak, F. Biomechanical factors in osteoarthritis.
7. Loeser, R. F., Goldring, S. R., Scanzello, C. R. & 16. Morrisette-Thomas, V. et al. Inflamm-aging does not Best Pract. Res. Clin. Rheumatol. 25, 815–823 (2011).
Goldring, M. B. Osteoarthritis: a disease of the joint as simply reflect increases in pro-inflammatory markers. 26. Lee, S., Kim, T. N. & Kim, S. H. Sarcopenic obesity is
an organ. Arthritis Rheum. 64, 1697–1707 (2012). Mech. Ageing Dev. 139, 49–57 (2014). more closely associated with knee osteoarthritis than
8. Pauli, C. et al. Macroscopic and histopathologic 17. Spector, T. D. et al. Low-level increases in serum is nonsarcopenic obesity: a cross-sectional study.
analysis of human knee menisci in aging and C‑reactive protein are present in early osteoarthritis Arthritis Rheum. 64, 3947–3954 (2012).
osteoarthritis. Osteoarthritis Cartilage 19, of the knee and predict progressive disease. 27. Scott, D., Blizzard, L., Fell, J. & Jones, G. Prospective
1132–1141 (2011). Arthritis Rheum. 40, 723–727 (1997). study of self-reported pain, radiographic
9. Hasegawa, A. et al. Anterior cruciate ligament 18. Livshits, G. et al. Interleukin‑6 is a significant predictor osteoarthritis, sarcopenia progression, and falls risk in
changes in the human knee joint in aging and of radiographic knee osteoarthritis: the Chingford community-dwelling older adults. Arthritis Care Res.
osteoarthritis. Arthritis Rheum. 64, 696–704 (2012). study. Arthritis Rheum. 60, 2037–2045 (2009). (Hoboken) 64, 30–37 (2012).

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

28. Chuckpaiwong, B., Charles, H. C., Kraus, V. B., 52. Blaney Davidson, E. N. et al. Elevated extracellular 78. Loeser, R. F., Gandhi, U., Long, D. L., Yin, W. &
Guilak, F. & Nunley, J. A. Age-associated increases matrix production and degradation upon bone Chubinskaya, S. Aging and oxidative stress reduce
in the size of the infrapatellar fat pad in knee morphogenetic protein‑2 (BMP‑2) stimulation point the response of human articular chondrocytes to
osteoarthritis as measured by 3T MRI. J. Orthop. Res. toward a role for BMP‑2 in cartilage repair and insulin-like growth factor 1 and osteogenic protein 1.
28, 1149–1154 (2010). remodeling. Arthritis Res. Ther. 9, R102 (2007). Arthritis Rheumatol. 66, 2201–2209 (2014).
29. Ushiyama, T., Chano, T., Inoue, K. & Matsusue, Y. 53. Johmura, Y. et al. Necessary and sufficient role for a 79. Klomsiri, C., Karplus, P. A. & Poole, L. B.
Cytokine production in the infrapatellar fat pad: mitosis skip in senescence induction. Mol. Cell 55, Cysteine-based redox switches in enzymes.
another source of cytokines in knee synovial fluids. 73–84 (2014). Antioxid. Redox Signal. 14, 1065–1077 (2011).
Ann. Rheum. Dis. 62, 108–112 (2003). 54. Krenning, L., Feringa, F. M., Shaltiel, I. A., van den 80. Wood, S. T. et al. Cysteine-mediated redox regulation
30. Bastiaansen-Jenniskens, Y. M. et al. Infrapatellar fat Berg, J. & Medema, R. H. Transient activation of p53 in of cell signaling in chondrocytes stimulated with
pad of patients with end-stage osteoarthritis inhibits G2 phase is sufficient to induce senescence. Mol. Cell fibronectin fragments. Arthritis Rheumatol. 68,
catabolic mediators in cartilage. Ann. Rheum. Dis. 55, 59–72 (2014). 117–126 (2016).
71, 288–294 (2012). 55. Ashraf, S. et al. Regulation of senescence associated 81. Collins, J. A. et al. Oxidative stress promotes
31. Lotz, M. & Loeser, R. F. Effects of aging on articular signaling mechanisms in chondrocytes for cartilage peroxiredoxin hyperoxidation and attenuates
cartilage homeostasis. Bone 51, 241–248 (2012). tissue regeneration. Osteoarthritis Cartilage 24, pro-survival signalling in aging chondrocytes.
32. Mobasheri, A., Matta, C., Zakany, R. & Musumeci, G. 196–205 (2016). J. Biol. Chem. 291, 6641–6654 (2016).
Chondrosenescence: definition, hallmarks and 56. Gossan, N. et al. The circadian clock in murine 82. Hardie, D. G. & Ashford, M. L. AMPK: regulating
potential role in the pathogenesis of osteoarthritis. chondrocytes regulates genes controlling key aspects energy balance at the cellular and whole body levels.
Maturitas 80, 237–244 (2015). of cartilage homeostasis. Arthritis Rheum. 65, Physiology (Bethesda) 29, 99–107 (2014).
33. Sharpless, N. E. & Sherr, C. J. Forging a signature of 2334–2345 (2013). 83. Petursson, F. et al. Linked decreases in liver kinase B1
in vivo senescence. Nat. Rev. Cancer 15, 397–408 57. Khapre, R. V., Kondratova, A. A., Susova, O. & and AMP-activated protein kinase activity modulate
(2015). Kondratov, R. V. Circadian clock protein BMAL1 matrix catabolic responses to biomechanical injury in
34. Aigner, T. et al. Apoptotic cell death is not a regulates cellular senescence in vivo. Cell Cycle 10, chondrocytes. Arthritis Res. Ther. 15, R77 (2013).
widespread phenomenon in normal aging and 4162–4169 (2011). 84. Terkeltaub, R., Yang, B., Lotz, M. & Liu-Bryan, R.
osteoarthritis human articular knee cartilage: a study 58. Dudek, M. et al. The chondrocyte clock gene Bmal1 Chondrocyte AMP-activated protein kinase activity
of proliferation, programmed cell death (apoptosis), controls cartilage homeostasis and integrity. suppresses matrix degradation responses to
and viability of chondrocytes in normal and J. Clin. Invest. 126, 365–376 (2016). proinflammatory cytokines interleukin‑1β and tumor
osteoarthritic human knee cartilage. Arthritis Rheum. 59. Baker, D. J. et al. Clearance of p16Ink4a-positive necrosis factor α. Arthritis Rheum. 63, 1928–1937
44, 1304–1312 (2001). senescent cells delays ageing-associated disorders. (2011).
35. Kozhemyakina, E. et al. Identification of a Prg4‑positive Nature 479, 232–236 (2011). 85. Lotz, M. K. & Carames, B. Autophagy and cartilage
articular cartilage progenitor cell population. Arthritis 60. Baker, D. J. et al. Naturally occurring p16ink4a‑positive homeostasis mechanisms in joint health, aging and
Rheumatol. 67, 1261–1273 (2015). cells shorten healthy lifespan. Nature 530, 184–189 OA. Nat. Rev. Rheumatol. 7, 579–587 (2011).
36. Munoz-Espin, D. & Serrano, M. Cellular senescence: (2016). 86. Ulgherait, M., Rana, A., Rera, M., Graniel, J. &
from physiology to pathology. Nat. Rev. Mol. Cell Biol. 61. Demaria, M. et al. An essential role for senescent cells Walker, D. W. AMPK modulates tissue and organismal
15, 482–496 (2014). in optimal wound healing through secretion of aging in a non-cell-autonomous manner. Cell Rep. 8,
37. Bovee, J. V., Hogendoorn, P. C., Wunder, J. S. & PDGF‑AA. Dev. Cell 31, 722–733 (2014). 1767–1780 (2014).
Alman, B. A. Cartilage tumours and bone development: 62. Zhu, Y. et al. The Achilles’ heel of senescent cells: 87. Carames, B., Olmer, M., Kiosses, W. B. & Lotz, M. K.
molecular pathology and possible therapeutic targets. from transcriptome to senolytic drugs. Aging Cell 14, The relationship of autophagy defects to cartilage
Nat. Rev. Cancer 10, 481–488 (2010). 644–658 (2015). damage during joint aging in a mouse model.
38. Freund, A., Orjalo, A. V., Desprez, P. Y. & Campisi, J. 63. Takayama, K. et al. Involvement of ERCC1 in the Arthritis Rheumatol. 67, 1568–1576 (2015).
Inflammatory networks during cellular senescence: pathogenesis of osteoarthritis through the modulation 88. Carames, B., Taniguchi, N., Otsuki, S., Blanco, F. J. &
causes and consequences. Trends Mol. Med. 16, of apoptosis and cellular senescence. J. Orthop. Res. Lotz, M. Autophagy is a protective mechanism in
238–246 (2010). 32, 1326–1332 (2014). normal cartilage, and its aging-related loss is linked
39. Coppe, J. P. et al. Tumor suppressor and aging 64. Chang, J. et al. Clearance of senescent cells by ABT263 with cell death and osteoarthritis. Arthritis Rheum.
biomarker p16INK4a induces cellular senescence without rejuvenates aged hematopoietic stem cells in mice. 62, 791–801 (2010).
the associated inflammatory secretory phenotype. Nat. Med. 22, 78–83 (2016). 89. Bohensky, J., Leshinsky, S. Srinivas, V. & Shapiro, I. M.
J. Biol. Chem. 286, 36396–36403 (2011). 65. Laberge, R. M. et al. MTOR regulates the Chondrocyte autophagy is stimulated by HIF‑1
40. Freund, A. & Patil, C. K. & Campisi, J. p38MAPK is a pro-tumorigenic senescence-associated secretory dependent AMPK activation and mTOR suppression.
novel DNA damage response-independent regulator phenotype by promoting IL1A translation. Pediatr. Nephrol. 25, 633–642 (2010).
of the senescence-associated secretory phenotype. Nat. Cell Biol. 17, 1049–1061 (2015). 90. Johnson, S. C. & Rabinovitch, P. S. & Kaeberlein, M.
EMBO J. 30, 1536–1548 (2011). 66. Jones, D. P. Redox theory of aging. Redox Biol. 5, mTOR is a key modulator of ageing and age-related
41. Tsuchida, A. I. et al. Cytokine profiles in the joint 71–79 (2015). disease. Nature 493, 338–345 (2013).
depend on pathology, but are different between 67. Finkel, T. Signal transduction by reactive oxygen species. 91. Zhang, Y. et al. Cartilage-specific deletion of mTOR
synovial fluid, cartilage tissue and cultured J. Cell Biol. 194, 7–15 (2011). upregulates autophagy and protects mice from
chondrocytes. Arthritis Res. Ther. 16, 441 (2014). 68. Hui, W. et al. Oxidative changes and signalling pathways osteoarthritis. Ann. Rheum. Dis. 74, 1432–1440
42. Demidenko, Z. N. & Blagosklonny, M. V. Growth are pivotal in initiating age-related changes in articular (2015).
stimulation leads to cellular senescence when the cell cartilage. Ann. Rheum. Dis. 75, 449–458 (2016). 92. Michan, S. & Sinclair, D. Sirtuins in mammals: insights
cycle is blocked. Cell Cycle 7, 3355–3361 (2008). 69. Finkel, T. Signal transduction by mitochondrial oxidants. into their biological function. Biochem. J. 404, 1–13
43. Rose, J. et al. DNA damage, discoordinated gene J. Biol. Chem. 287, 4434–4440 (2012). (2007).
expression and cellular senescence in osteoarthritic 70. Wang, Y., Zhao, X., Lotz, M., Terkeltaub, R. & 93. Burnett, C. et al. Absence of effects of Sir2
chondrocytes. Osteoarthritis Cartilage 20, Liu-Bryan, R. Mitochondrial biogenesis is impaired overexpression on lifespan in C. elegans and
1020–1028 (2012). in osteoarthritis chondrocytes but reversible via Drosophila. Nature 477, 482–485 (2011).
44. Harbo, M. et al. The relationship between ultra-short peroxisome proliferator-activated receptor γ coactivator 94. Kugel, S. & Mostoslavsky, R. Chromatin and beyond:
telomeres, aging of articular cartilage and 1α. Arthritis Rheumatol. 67, 2141–2153 (2015). the multitasking roles for SIRT6. Trends Biochem. Sci.
the development of human hip osteoarthritis. 71. Blanco, F. J., Rego, I. & Ruiz-Romero, C. The role of 39, 72–81 (2014).
Mech. Ageing Dev. 134, 367–372 (2013). mitochondria in osteoarthritis. Nat. Rev. Rheumatol. 95. Mostoslavsky, R. et al. Genomic instability and aging-
45. Jurk, D. et al. Postmitotic neurons develop a 7, 161–169 (2011). like phenotype in the absence of mammalian SIRT6.
p21‑dependent senescence-like phenotype driven by 72. Loeser, R. F. Aging and osteoarthritis. Curr. Opin. Cell 124, 315–329 (2006).
a DNA damage response. Aging Cell 11, 996–1004 Rheumatol. 23, 492–496 (2011). 96. Kanfi, Y. et al. The sirtuin SIRT6 regulates lifespan
(2012). 73. Koike, M. et al. Mechanical overloading causes in male mice. Nature 483, 218–221 (2012).
46. Liu, L. & Rando, T. A. Manifestations and mechanisms mitochondrial superoxide and SOD2 imbalance in 97. Liu-Bryan, R. & Terkeltaub, R. Emerging regulators
of stem cell aging. J. Cell Biol. 193, 257–266 (2011). chondrocytes resulting in cartilage degeneration. of the inflammatory process in osteoarthritis.
47. Philipot, D. et al. p16INK4a and its regulator miR‑24 link Sci. Rep. 5, 11722 (2015). Nat. Rev. Rheumatol. 11, 35–44 (2015).
senescence and chondrocyte terminal differentiation- 74. Aigner, T. et al. Large-scale gene expression profiling 98. Nagai, K. et al. Depletion of SIRT6 causes cellular
associated matrix remodelling in osteoarthritis. reveals major pathogenetic pathways of cartilage senescence, DNA damage, and telomere dysfunction
Arthritis Res. Ther. 16, R58 (2014). degeneration in osteoarthritis. Arthritis Rheum. 54, in human chondrocytes. Osteoarthritis Cartilage 23,
48. Sousa-Victor, P. et al. Geriatric muscle stem cells switch 3533–3544 (2006). 1412–1420 (2015).
reversible quiescence into senescence. Nature 506, 75. Ruiz-Romero, C. et al. Mitochondrial dysregulation of 99. Wu, Y. et al. Overexpression of Sirtuin 6 suppresses
316–321 (2014). osteoarthritic human articular chondrocytes analyzed cellular senescence and NF‑κB mediated inflammatory
49. Vincent, T., Hermansson, M., Bolton, M., Wait, R. & by proteomics: a decrease in mitochondrial superoxide responses in osteoarthritis development. Sci. Rep.
Saklatvala, J. Basic FGF mediates an immediate dismutase points to a redox imbalance. Mol. Cell. 5, 17602 (2015).
response of articular cartilage to mechanical injury. Proteomics 8, 179–189 (2008). 100. Sanchez-Adams, J., Leddy, H. A., McNulty, A. L.,
Proc. Natl Acad. Sci. USA 99, 8259–8264 (2002). 76. Scott, J. L. et al. Superoxide dismutase downregulation O’Conor, C. J. & Guilak, F. The mechanobiology of
50. Hoshiyama, Y. et al. Chondrocyte clusters adjacent to in osteoarthritis progression and end-stage disease. articular cartilage: bearing the burden of
sites of cartilage degeneration have characteristics of Ann. Rheum. Dis. 69, 1502–1510 (2010). osteoarthritis. Curr. Rheumatol. Rep. 16, 451 (2014).
progenitor cells. J. Orthop. Res. 33, 548–555 (2015). 77. Yin, W., Park, J. I. & Loeser, R. F. Oxidative stress 101. Verzijl, N. et al. Crosslinking by advanced glycation
51. Fukui, N., Zhu, Y., Maloney, W. J., Clohisy, J. & inhibits insulin-like growth factor‑I induction of end products increases the stiffness of the collagen
Sandell, L. J. Stimulation of BMP‑2 expression by chondrocyte proteoglycan synthesis through differential network in human articular cartilage: a possible
pro-inflammatory cytokines IL‑1 and TNF-α in normal regulation of phosphatidylinositol 3‑Kinase-Akt and mechanism through which age is a risk factor
and osteoarthritic chondrocytes. J. Bone Joint Surg. Am. MEK–ERK MAPK signaling pathways. J. Biol. Chem. for osteoarthritis. Arthritis Rheum. 46, 114–123
85‑A (Suppl. 3), 59–66 (2003). 284, 31972–31981 (2009). (2002).

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrrheum


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

102. Vos, P. A. et al. Elevation of cartilage AGEs does 107. Allen, J. L., Cooke, M. E. & Alliston, T. ECM stiffness Acknowledgements
not accelerate initiation of canine experimental primes the TGFβ pathway to promote chondrocyte The authors’ research work is funded by grants from the
osteoarthritis upon mild surgical damage. differentiation. Mol. Biol. Cell 23, 3731–3742 (2012). National Institute on Aging (RO1 AG044034 to R.F.L. and
J. Orthop. Res. 30, 1398–1404 (2012). 108. Conboy, M. J., Conboy, I. M. & Rando, T. A. F32 AG050399 to B.O.D.).
103. Li, Y. et al. Establishment of a rabbit model to study Heterochronic parabiosis: historical perspective and
the influence of advanced glycation end products methodological considerations for studies of aging Author contributions
accumulation on osteoarthritis and the protective and longevity. Aging Cell 12, 525–530 (2013). All authors researched data for the article and contributed to
effect of pioglitazone. Osteoarthritis Cartilage 24, 109. Greene, M. A. & Loeser, R. F. Aging-related discussion of content, writing the article, and reviewing and
307–314 (2016). inflammation in osteoarthritis. Osteoarthritis Cartilage editing the manuscript before submission.
104. Kim, J. H. et al. Matrix cross-linking-mediated 23, 1966–1971 (2015).
Competing interests statement
mechanotransduction promotes posttraumatic 110. Loeser, R. F. Aging processes and the development of
The authors declare no competing interests.
osteoarthritis. Proc. Natl Acad. Sci. USA 112, osteoarthritis. Curr. Opin. Rheumatol. 25, 108–113
9424–9429 (2015). (2013). Review criteria
105. Madej, W. et al. Ageing is associated with reduction of 111. Kaeberlein, M., Rabinovitch, P. S. & Martin, G. M. The authors searched the PubMed database for articles using
mechanically-induced activation of Smad2/3P Healthy aging: the ultimate preventative medicine. the search terms “ageing”, “cellular senescence”, “oxidative
signaling in articular cartilage. Osteoarthritis Cartilage Science 350, 1191–1193 (2015). stress”, “AMPK”, “autophagy” and “sirtuin”, combined with
24, 146–157 (2016). 112. Longo, V. D. et al. Interventions to slow aging in “chondrocyte”, “cartilage” or “osteoarthritis”, and years start-
106. Blaney Davidson, E. N. et al. Increase in ALK1/ALK5 humans: are we ready? Aging Cell 14, 497–510 (2015). ing at 2010. Full-length original manuscripts published in
ratio as a cause for elevated MMP‑13 expression in 113. Dai, D. F., Chiao, Y. A., Marcinek, D. J., Szeto, H. H. & English were selected for further review. Key references from
osteoarthritis in humans and mice. J. Immunol. 182, Rabinovitch, P. S. Mitochondrial oxidative stress in identified manuscripts that reported important earlier studies
7937–7945 (2009). aging and healthspan. Longev. Healthspan 3, 6 (2014). of interest were also reviewed.

NATURE REVIEWS | RHEUMATOLOGY ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

Anda mungkin juga menyukai