You are on page 1of 13

REVIEWS

Inflammatory mechanisms in
tendinopathy – towards translation
Neal L. Millar1, George A. C. Murrell2 and Iain B. McInnes1
Abstract | Tendinopathy is a multifactorial spectrum of tendon disorders that affects different
anatomical sites and is characterized by activity-related tendon pain. These disorders are
common, account for a high proportion (~30%) of referrals to musculoskeletal practitioners and
confer a large socioeconomic burden of disease. Our incomplete understanding of the
mechanisms underpinning tendon pathophysiology continues to hamper the development of
targeted therapies, which have been successful in other areas of musculoskeletal medicine.
Debate remains among clinicians about the role of an inflammatory process in tendinopathy
owing to a lack of clinical correlation. The advent of modern molecular techniques has
highlighted the presence of immune cells and inflammatory mechanisms throughout the
spectrum of tendinopathy in both animal and human models of disease. Key inflammatory
mediators — such as cytokines, nitric oxide, prostaglandins and lipoxins — play crucial parts in
modulating changes in the extracellular matrix within tendinopathy. Understanding the links
between inflammatory mechanisms, tendon homeostasis and resolution of tendon damage will
be crucial in developing novel therapeutics for human tendon disease.

Enthesopathy Tendinopathy is a term used to describe a complex mul- production of proinflammatory cytokines9. Similar to
Injury to the enthesis, the ti-faceted pathology of the tendon characterised by pain, tendinopathy, mechanical overload in OA remains a
portion of tendon at the decline in function and reduced exercise tolerance1,2. key driver of pathology but the evidence now suggests
bone–tendon junction site. Tendinopathies pose an important clinical problem — it is mediated, at least in part, through elements of the
TNFΔARE mice
particularly in musculoskeletal and sports-related med- inflammatory response10.
Mouse model in which icine3 — and account for up to 30% of general practice Furthermore, recent musculoskeletal scientific atten-
systemic overexpression of musculoskeletal consultations. Historically, there has tion has focused of the immunobiology of the enthesis11,
TNF leads to the development been considerable disagreement with regard to classifi- the connective tissue between tendon or ligament and
of inflammation.
cation and terminology related to tendon disorders. The bone. Enthesopathy has been considered analogous to
term ‘tendonitis’ was traditionally used by rheumatolo- tendinopathy as both the enthesis and tendon are sites
gists to describe painful symptoms attributed to inflam- of high mechanical stress12. Interestingly, the enthesis
mation of the tendon4 whereas ‘tendinosis’ was used to contains a unique population of resident T cells, which,
1
Institute of Infection,
Immunity and Inflammation, reflect degenerative changes at the microscopic level5. when activated by the cytokine IL‑23, can promote
College of Medicine, Much controversy has stemmed from sports-­related pathogenesis that is characteristic of spondyloarthritis13.
Veterinary and Life Sciences tendon disease studies that concluded that there is no Additionally, TNFΔARE mice (which provide a translational
University of Glasgow, clinical evidence (redness, tenderness, skin changes or spondyloarthritic model)14 show that enthesitis (inflam-
120 University Avenue,
Glasgow G12 8TA, UK.
raised systemic inflammatory markers in blood) of a mation of the enthesis) is driven by mechanical strain
2
Orthopaedic Research classic inflammatory event in tendino­pathy and that and resident stromal cells, two facets that are dysregu-
Institute, Department of a chronic degenerative disorder devoid of an inflam- lated in tendinopathic lesions15. Similarly, advances in
Orthopaedic Surgery, matory process prevails. A similar view was taken in tendon biology have increasingly recognized that a lack
St George Hospital Campus,
relation to the pathophysiology of osteo­arthritis (OA), of observation of an acute inflammatory infiltrate does
4–10 South Street Kogarah,
Sydney, NSW 2217, which was attributed mainly to excessive wear and tear 6 not exclude a role for inflammation in the pathogenesis of
Australia. and devoid of an inflammatory phenotype7. However, tendinopathy throughout the spectrum or kinetics of the
Correspondence to N.L.M.  recent evidence suggests that OA is an active process disease16. Inflammation in immunological terms may be
neal.millar@glasgow.ac.uk caused not simply by ageing and cartilage loading but defined as a signal-mediated response to cellular insult 17.
doi:10.1038/nrrheum.2016.213 also by infiltration of inflammatory cells8 (includ- The inflammatory response protects the body against
Published online 25 Jan 2017 ing CD4+ T cells and natural killer (NK) cells) and injury and can promote healing but can itself become

110 | FEBRUARY 2017 | VOLUME 13 www.nature.com/nrrheum


©
2
0
1
7
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
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points I trials showing no efficacy compared with saline26,27.


Stem-­­­cell-­based therapy has improved tendon healing
• Tendinopathy is a complex multi-faceted tendon pathology commonly associated in equine tendinopathy 28 but effective translation to
with overuse humans has yet to materialise. Importantly, in an era
• Most current treatments for tendinopathy are neither effective nor evidence-based of increased regulatory oversight of medical devices
• Many recent studies have highlighted inflammatory cell infiltrates in both animal and and pharmaceuticals29 none of the recent ‘novel’ tendi-
human tendon disease nopathy treatments (such as PRP or stem-cell-based
• The potential roles of inflammatory mediators acting on the resident tenocytes are therapy) have provided mechanistic insight; thus, their
the source of some controversy and require in‑depth investigation using in vitro and wholehearted adoption in the clinical community is not
in vivo models yet apparent. Surgical options including debridement
• Understanding the key inflammatory pathways affecting extracellular matrix and microcautery remain the last option owing to their
regulation and homeostasis are critical in designing future targeted therapies morbidity and inconsistent outcomes30.
for tendinopathy
Tendinopathy pathophysiology
The primary function of a tendon is to enable transduc-
dysregulated with deleterious consequences. It is now tion of mechanical forces from the muscle to bone31. The
increasingly evident that inflammatory mechanisms and high tensile strength of tendons is derived from a strictly
the innate immune system are activated within the ten- linear arrangement of collagen fibrils formed as a result
don matrix microenvironment during tissue injury and of covalent cross-linking of collagen molecules. The
probably contribute to dysregulated homeostasis18. This main component of tendons is type I collagen, which
Review aims to consolidate and interpret the inflam- accounts for approximately 60–85% of the dry weight
matory molecular events involved in tendinopathy, and of the tendons, with the remainder made up of proteo­
analyse how this information might best be utilised to glycans, glycosaminoglycans (GAGs), glycoproteins
transform the management of human tendon disease. and other collagen types (III, XII, V)2,32. Tenocytes are
fibroblast-like cells that make up the basic cellular com-
Clinical features of tendinopathy ponent of tendon tissue and are found uniformly aligned
The absence of pain and functional limitation makes between collagen fibrils33. Tenocytes react to external
the diagnosis of early tendinopathy difficult (FIG. 1). stimuli to regulate the synthesis and turnover of extracel-
Tendinopathy is clinically diagnosed after gradual onset lular matrix (ECM) components facilitating functional
of activity-related pain, decreased function and some- adaptation in response to altered mechanical load34,35.
Mid substance times localised swelling, and clinical examination reveals The pathological features of tendinopathy have
Midportion of a tendon.
pain with stretching and palpation of the pathological been described in many studies over the past 30 years36.
Eccentric contraction area15,19. Ultrasonography and MRI are extremely helpful Changes in the ECM in tendinopathy are character-
Phase of contraction that in formulating a diagnosis while additionally differen- ised by a loss of collagen organization, fibrocartilagenous
occurs as the muscle tiating between mid substance versus enthesial tendi- change with deposition of additional ECM protein (such
lengthens.
nopathy and assisting surgical planning. Treatments as GAGs)37,38. Importantly, type III collagen is produced
Isometric contraction commonly commence with physical therapy, which in the initial phases of tendon damage39 as a conserved
Type of contraction during itself has moved from the use of pure ‘eccentric’ training mechanism to provide a rapid ‘patch’ to the area of
which muscle length does not programmes20 towards ‘isometric’ exercise, which has damage. Type III collagen is laid down in a haphazard
change (as opposed to been shown to reduce initial pain followed by eccen- fashion, contributing to the irregular alignment seen
concentric or eccentric
contractions).
tric retraining of tendon21. Robust systematic evidence microscopically and translating to inferior biomechani-
continues to support first-line physiotherapy treatment cal strength in damaged tendon40. In the normal tendon,
Extracorpeal shockwave with between 53–86% improvement in the symptoms22. type III collagen is gradually replaced by type I collagen,
therapy Additionally, physiotherapy modalities such as ultra- which resumes the linear structured arrangement with
Use of high-amplitude pulses
sound, laser therapy, hyperthermia and extracorpeal eventual resolution to normal tendon ultrastructure41.
of mechanical energy, similar
to soundwaves, to treat shockwave therapy (ESWT) have been advocated; how- Microscopically, tenocytes become rounder (enlarged
tendinopathic lesions. ever, evidence to support their widespread use in all lysosomes), more proliferative and apoptotic before
tendinopathies remains inconsistent 23. Oral NSAIDs the development of tendinopathy 42–44. At the electron-­
Debridement have been used extensively for decades to treat pain asso- microscopic level, collagen fibres in tendinopathic ten-
Surgical removal of
degenerative tendon tissue.
ciated with tendon overuse. Whereas evidence suggests dons are angulated, vary in diameter, and have buckling
that both oral and locally administered NSAIDS are of the ECM45 consistent with poor structural integrity.
Microcautery effective in relieving the pain associated with tendino­ Additionally, increased neovascularisation has been
Micro-debridement of diseased pathy in the short term (7–14 days), their efficacy in the previously thought to represent a classic pathological
areas of tendon tissue using a
treatment of chronic tendinopathy is unclear 24. Platelet- feature of tendinopathy 23,46; however, evidence now sug-
high-temperature fine-tipped
instrument. rich plasma (PRP) is a blood derivative that contains gests that this is probably part of a complex interaction
high levels of growth factors (platelet-derived growth with neoinnervation and may represent homeostatic
Fibrocartilaginous change factor, vascular endothelial growth factor and cytokines remodelling 47,48.
A process in which such as IL‑6, IL‑8 and IL‑1β) that can promote tissue Theories about the pathophysiology of tendino­pathy
chondrogenesis occurs in an
area of tendon and the
healing in vitro 25 and has quickly been adopted into postulated thus far are varied, probably not mutually
structure of the cells changes the clinically arena. However, the benefits of PRP for exclusive and largely unproven15. The mechanisms
to chondrocytes. tendinopathy remain controversial, with several level include dysregulated apoptosis49, mechanical overload50,

NATURE REVIEWS | RHEUMATOLOGY VOLUME 13 | FEBRUARY 2017 | 111


©
2
0
1
7
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
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Paratenon Epitenon Endotenon Collagen Tenocyte


fibre

Collagen fibril

Fascicle

Pathology
• Mucoid degeneration
• Neovascularization
• ↑ Type III/type I collagen ratio
• Apoptosis

Normal Tendinopathic

Clinical features
• Pain
• Swelling
• Loss of function

Treatments
• Physiotherapy
(isometric, eccentric)
• Oral analgesia, NSAIDs
• Orthotics
• Injections
• Shockwave therapy
• Surgery

Figure 1 | Pathological and clinical features of tendinopathy. Schematic representation of the microstructure of normal
tendon showing collagen fibril structure and tenocytes in homeostatic tendon. The main Nature pathological features
Reviews of
| Rheumatology
tendinopathy include mucoid degeneration, loss of parallel collagen fibril structure, increased neovessels, changes in
type III to type I collagen ratio and cell death (apoptosis). Importantly, there is a ‘switch’ in collagen production from the
predominant type I collagen (95% in normal tendon) to the biomechanically inferior type III collagen (5% in normal
tendon), which is increased by ~30% in tendinopathic tissues. Cardinal clinical signs include pain, fusiform swelling and
loss of function, particularly in sporting individuals. Treatments range from physiotherapy (isometric and eccentric
exercises) aiming to reduce pain by loading the tendon back to a homeostatic state, to pain relief with NSAIDs and
local corticosteroid injections. Surgery (tendon release and debridement, microcautery) remains possible for
recalcitrant situations with more-recent therapies including shockwave therapy and platelet-rich plasma injections
whose evidence base remains limited.

imbalance in the equilibrium between the activity of One of the major limitations of human studies on tendin-
matrix metalloproteinases (MMPs) and tissue inhibitors of opathy is that tendon biopsy samples are usually obtained
metalloproteinases (TIMPs)1, genetic factors (such as pol- when patients are sufficiently symptomatic to require sur-
ymorphisms in COL5A1 (encoding type V collagen) and gery and therefore such biopsy­obtained material is likely
MMP8)51, neuronal proliferation52 and inflammation53,54. to represent chronic rather than early-phase disease55.

112 | FEBRUARY 2017 | VOLUME 13 www.nature.com/nrrheum


©
2
0
1
7
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
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

techniques, a clear signature defining inflammatory


Blood vessel mechanisms has become apparent in the spectrum of
tendinopathic disease in both animals and humans61–64.
One of the most clinically and functionally relevant
Infiltrating compartment classification systems for tendinopathy remains that
• Immune cell differentiation of Martin Blazina through his original work on patel-
M1 • Cytokine and chemokine
Mϑ lar tendinopathy62. This classification system divides the
production
Infiltrating chronology of symptoms into acute (when symptoms
mast cell Macrophage Proinflammatory Pro-resolving are present for 0–6 weeks), sub-acute (6–12 weeks)
macrophage M2
Infiltrating T cell macrophage and classic chronic tendinopathy (when symptoms are
present for more than 3 months). Molecular evidence
suggests that many of the key inflammatory interac-
Collagen fibres
tions occur in the earlier (acute and subacute) stages of
repetitive tendon microtrauma when patients may be
Resident Stromal compartment asymptomatic, which could account for the disparate
macrophage • Matrix regulation, views between healthcare professionals and the scien-
Tenocytes inflammatory crosstalk tific community 63. At these early stages, changes in tissue
• Cytokine and chemokine microenvironment and activation of the innate immune
Resident mast cell production
system interact at a crossroads between reparative ver-
sus degenerative ‘inflammatory’ healing. Additional
evidence suggests that repetitive biomechanical stress
and its associated damage in stromal tissues plays a key
Immune-sensing compartment part in the immune system response to regeneration64.
• DAMP and PAMP recognition Therefore, it seems that inflammation in tendinopathy
• Cytokine and chemokine production encompasses three distinct cellular compartments (stro-
mal, immune-sensing and infiltrating compartments),
Figure 2 | Immunobiology of tendinopathy. Inflammation in tendinopathy
encompasses three distinct compartments involved in a NaturecomplexReviews
network| Rheumatology
of
each contributing to a complex milieu of inflammatory
downstream effects with consequences for tendon homeostasis. The infiltrating mechanisms effecting tendon homeostasis (FIG. 2).
compartment includes influxing immune cells: T cells, mast cells and macrophages
(proinflammatory (M1) versus pro-resolving (M2)) probably recruited through stromal The stromal compartment
and resident immune cell activation that, in normal circumstances, represents a The most abundant cells of the stroma are tenocytes,
homeostatic inflammatory response but is aberrant in tendinopathic disease. The which are responsible for tissue remodelling and repair.
immune-sensing compartment comprises tendon-resident innate cells (mast cells and Literature supporting a critical role for tissue stroma in
macrophages) that act as sentinels to respond to initial tissue insult through further defining the phenotype and outcome of inflam-
damage-associated or pathogen-associated molecular patterns (DAMPs and PAMPs,
matory responses is rapidly increasing 65. Given the rel-
respectively) and become activated via downstream cytokine signalling. Finally, the
centrally placed resident tenocytes sit in the influential stromal compartment, which is
ative paucity of primary tenocyte studies, we consider
responsible for tissue remodelling and repair. Tenocytes secrete cytokines and that a comparative approach with another musculo­
chemokines in either an autocrine or paracrine way due to the presence of cell-surface skeletal disease, such as rheumatoid arthritis (RA),
immune receptors and can be driven toward an activated inflammatory phenotype. may be informative. RA synovial fibroblasts (RASFs)
These three compartments contribute to interactions between the inflammatory exhibit anchorage independence, semi-autonomous
response and the extracellular matrix in diseased tendon, which in turn comprise a proliferation and altered ECM synthesis, and can pro-
balance between reparation versus further degeneration within the tendon. duce inflammatory cytokines and chemokines66 (such
as IL‑8, CCL5, and growth-regulated alpha protein
(CXCL1)) that recruit and activate leukocytes in the
Additionally, purported theories on the pathophysiological synovial compartment. Thus, RASF activation results
mechanisms involved in tendinopathy have changed over in the accumulation, survival and retention of leuko-
time. For example, the ‘continuum’ model that describes cytes at sites of disease, mimicking the microenviron-
reactive tendinopathy, tendon disrepair and degenerative ment seen in lymphoid tissues, and thereby promoting
tendinopathy reports no inflammation-meditated change chronic inflammation. Moreover, RASFs that are iden-
throughout the course of these injuries56, whereas the tified by expression of the markers podoplanin (PDPN)
‘failed healing’ theory defines inflammatory responses and transmembrane receptor CD248 have been shown
throughout the course of tendinopathy (injury, failed to drive aberrant tissue damage67. This work has been
healing, and clinical presentation)57. reproduced in tendinopathy whereby biopsy-obtained
samples of diseased tendon showed increased CD248
Inflammation in tendinopathy and PDPN expression and cytokine (IL‑1β, IL‑6, IL‑8)
Disparate views surrounding the presence of an inflam- stimulation drove the resident stromal tenocyte toward
matory phenotype in tendinopathy have persisted pos- an activated PDPN+CD248+ inflammatory phenotype68.
sibly as a result of several historical studies highlighting We therefore propose that resident tenocytes possess the
Patellar tendinopathy
A common overuse injury,
no inflammatory cells or acute inflammatory infiltrates capacity to behave similarly, perhaps in response to tissue
caused by repeated stress on (from polymorphonuclear cells) in various human tissue damage components recognized via damage-associated
the patellar (kneecap) tendon. specimens58–60. With the advent of modern molecular molecular patterns (DAMPs) and pathogen-associated

NATURE REVIEWS | RHEUMATOLOGY VOLUME 13 | FEBRUARY 2017 | 113


©
2
0
1
7
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
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

molecular patterns (PAMPs), or other alarmins. Thereby, tendon is stressed, providing a molecular signature
subsequent immune-stromal cell–matrix interactions for early human disease. Moreover, other studies have
could have an important role in the failed resolution revealed the presence of NK cells and T lymphocytes
of an inflammatory response in normal mechanically in tendinopathic and normal human tendon biopsy
loaded tendons leading to chronic persistent disease that samples81,82, which suggests that the infiltrating cell
would manifest as tendinopathy. population may be more immunologically active than
previously thought.
The immune-sensing compartment
In tissues, macrophages and mast cells act as immune Molecular mechanisms of inflammation
sentinels in the front line of tissue defence, discretely It is increasingly clear that interactions between immune
positioned and transcriptionally programmed for cells (either resident or infiltrating) and resident stromal
detecting and responding to initial tissue damage. cells have important roles in turning a spontaneously
Increased accumulation of macrophages and mast cells resolving inflammatory response within tendon tissue
has been reported in a rodent model of upper extrem- into a chronic disease with ultimate tissue degeneration.
ity overuse69 and a calcaneal tendon overuse model70, As with other musculoskeletal pathologies, the ability
respectively. Frequently, macrophages have been func- to sample human tissue that truly reflects the molecu-
tionally grouped into two classes: M1 and M2. This con- lar mechanisms driving disease in terms of location and
cept has often been over-interpreted as a rigid functional time remains a challenge. Therefore, animal models of
classification of macrophages and not, as it was seem- tendinopathy may improve our understanding of human
ingly intended, as a simplified operational concept 71. tendinopathy and its underlying pathology. However,
M1‑polarized (classically activated) macrophages seem current animal models present advantages and limi-
to show a proinflammatory response pattern, whereas tations, and there remains no consensus regarding the
M2 (alternatively activated) macrophages dampen criteria of an universal tendinopathy animal model83.
inflammatory responses by producing immuno­ Notwithstanding the limitations of such models, we shall
suppressive cytokines such as IL‑1 receptor antagonist now consider the current range of inflammatory mole-
(IL‑1Ra), IL‑10, IL‑4 and IL‑13 (REF. 72). Per this model, cules in both animal and human models of disease (FIG. 3)
tissue-resident macrophages are classified as M2‑like, that may play a part in the stromal microenvironment
with fundamental roles in tissue homeostasis that relate leading to tendinopathic features. For clarity and subse-
to the role of macrophages during development, main- quent translational utility, we have taken a reductionist
tenance of homeostasis and resolution of inflamma- approach in describing individual candidate effector
tion73. Pan macrophage (CD68) and M2 macrophage moieties. It is vital to recall that inflammatory responses
(CD206) markers have been described in equine 74 comprise complex multi-cellular responses coordinated
and human75 tendinopathy in early and late stages of by groups of mediators operating in a coordinated system
the disease. Mast cells have been found throughout the rather than in isolation.
spectrum of normal and tendinopathic human ten-
don disease76. Mast-cell-derived immune mediators Cytokines
modulate collagen synthesis and MMP expression in Cytokines remain the most investigated inflammatory
human tenocytes77, reflecting their probably impor- mediators in tendinopathy, probably owing to their crit-
tant homeostatic role in matrix regulation within this ical interactions with the resident tenocytes, ECM and
compartment. immune cells, and the extent of suggestive cytokine effec-
tor biology gleaned from other musculoskeletal disease
The infiltrating compartment. This compartment pathologies (TABLE 1).
comprises an influx of immune cells, their subsequent
downstream cytokine production and the effect of IL‑1 family signalling. The IL‑1 superfamily of cytokines
inflammation and ECM crosstalk that determines rep- are important regulators of innate and adaptive immu-
aration versus degeneration. Surgically induced tendon nity, playing key parts in host defence against infection,
injury elicited a sequential pattern of inflammatory cell inflammation, injury and stress. IL‑1β is released by
infiltration, consisting of a rapid and transient accumu- tendon cells in response to mechanical stretching 84,
lation of neutrophils, followed by an increase in macro­ whereas the addition of IL‑1β to tendon cells in vitro
phage infiltration between 1 and 28 days post-injury results in the production of proinflammatory molecules,
in a rat model of Achilles tendon injury 78. A systemic such as prostaglandin G/H synthase 2 (also known as
Alarmins review of the presence of immune cells in human cyclooxygenase‑2 (COX‑2)), MMP1, MMP3 and EP4
Molecules released from a tendinopathy has shown increased macrophages (four receptor for prostaglandin E2 (PGE2)85. Importantly,
damaged or diseased cell that
studies) and mast cells (three studies) in tendinopathic IL‑1β downregulates considerably the expression of
stimulate an immune response.
versus healthy tissues79. We previously suggested non- type I collagen mRNA in human tenocytes thus linking
Full-thickness rotator torn subscapularis tendon in patients with full-thickness inflammation and ECM remodelling. In addition, IL‑1β
cuff tears rotator cuff tears as an ‘early model of tendinopathy’ on selectively stimulates the expression of PGE2 in human
Tearing of one or more of the the basis of histological appearance and substantially tenocytes, which may lead to ECM degradation in the
rotator cuff tendons whereby
the tendon no longer fully
increased levels of cytokines and apoptotic markers80. tendon86. Proteomic analysis of equine tendinopathy has
attaches to the head of the This observation, in turn, likely reflects the earliest shown ECM cleavage fragments in early stages of tendon
humerus. impact of altered biomechanical loading whereby the disease generated by an IL‑1β‑mediated mechanism87.

114 | FEBRUARY 2017 | VOLUME 13 www.nature.com/nrrheum


©
2
0
1
7
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
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

IL-33 IL-6
Type III collagen regulation Collagen synthesis
IL-4
IL-4R Reduced tendon strength
IL-33
IL-6 IL-6R
IL-1 IL-1RAcP ST2L IL-4
Reduced tendon IL-4Rα IL-13
strength IL-13R Tenocyte
IL-1β γC
proliferation
IL-1R1 IL-13
gp130 JAK1 IL-4Rα
miR29a IL-13Rα1 IL-21R
TNF MyD88 P JAK3
Tenocyte activation STAT6 Tenocyte
TYK1 JAK1 IL-13Rα2 activation
IRAK
TNF P
TRAF6 MAPK STAT6 IL-21R
STAT3
TNFR ERK, p38, JNK TYK2

TRAF2 • Inflammation STAT3


• Collagen remodelling
NF-κB COX2 • Cell proliferation Tenocyte
TRAF6 • Neoangiogenesis

IL-17R
EP2/4

IL-17A, IL-17F
FPR2
PGE2
LXA4
IL-17 15-epi-LXA4
Apoptosis
Cytokine induction LXA4 PGE2 Inflammatory/
Type III collagen Tenocyte activation Tenocyte proliferation matrix crosstalk
Figure 3 | Inflammatory mechanisms in tendinopathy. This figure highlights the potential targetable intracellular
signalling pathways that have functional consequences (such as regulation of extracellular matrix (ECM), immune cell
recruitment and cell proliferation) relevant to tendon pathology. For example, activation of mitogen-activated protein
Nature
kinase (MAPK) signalling by the cytokines IL‑33 (through myeloid differentiation primary Reviews
response | Rheumatology
protein MyD88, IL‑1
receptor-associated kinase (IRAK) and TNF receptor-associated factor 6 (TRAF6)), IL‑6 (through p130) and IL‑17
(through TRAF6); activation of nuclear factor κB (NF‑κB) signalling by TNF (through TRAF2) or 15‑epi-lipoxin A4
(15‑epi‑LXA4); and activation Janus kinase (JAK) and signal transducer and activator of transcription (STAT) by IL-13
result in downstream mediation of enhanced cytokine and chemokine production, ECM remodelling (collagen and
other proteins), proliferation and angiogenesis, all of which become dysregulated in tendon disease. Appreciation of
these pathways could help define the molecular checkpoints that modify a homeostatic inflammatory response toward
an aberrant inflammatory tendon microenvironment that leads to clinical tendinopathy. An ideal tendinopathy target
should modify the proinflammatory response and promote resolution by sparing inflammation-induced healing
moieties and encouraging robust and rapid matrix repair. COX, cyclooxygenase; EP, prostaglandin E2 receptor; FPR2,
N-formyl peptide receptor 2; PGE2, prostaglandin E2.

A further IL‑1 family member, IL‑18, has been identi- (IL‑R) signalling pathway. In an in vivo patellar tendon
fied at mRNA and protein levels in human supraspinatus injury model, intraperotineal injection of rhIL‑33 did
tendinopathy samples53. not affect synthesis of type I collagen but increased
Another IL‑1 homologue, IL‑33, is released following type III collagen synthesis considerably, particularly
cellular damage88 and biomechanical overload89, and has in injured tendons91. Moreover, rhIL33 administration
been implicated in a variety of inflammatory patholo- markedly reduced ultimate tendon strength at all the
gies90. We observed elevated IL‑33 expression in early time points post-injection, suggesting that such changes
human tendinopathy compared with both established were of functional impact. Conversely, in St2−/− mice
tendinopathy and intact control tendon91. Addition of (which lack the St2 gene (also known as Ll1rl1) encod-
recombinant human IL‑33 (rhIL‑33) to human tenocyte ing the IL‑33 receptor), rhIL‑33 administration did not
cultures in vitro resulted in increased expression of both affect collagen ECM synthesis or ultimate strength of
mRNA and protein levels of type I collagen and, par- the healing tendon, which confirmed that IL‑33 acted
ticularly, type III collagen, with an associated increase in via an ST2‑dependent pathway. Neutralizing antibodies
protein expression of IL‑6, IL‑8 and monocyte chemo­ directed against IL‑33 attenuated the type I collagen to
attractant protein‑1 (CCL2). These IL‑33‑induced type III collagen switch at days 1 and 3 post-injury in
changes in levels of expression of collagen and cytokines injured wild-type mice resulting in a substantial increase
were abrogated by NF‑κB inhibition, suggesting that in biomechanical strength of the tendons of wild-type
IL‑33 operates in tenocytes via a canonical IL‑1 receptor mice at day 1 post-injury. Targeting IL‑33 or ST2 the

NATURE REVIEWS | RHEUMATOLOGY VOLUME 13 | FEBRUARY 2017 | 115


©
2
0
1
7
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
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 | Cytokines implicated in tendinopathy proinflamatory and immunoregulatory cytokines102, and


culturing tendon explants in the presence of TNF leads
Cytokines Proposed function in tendinopathy to upregulation of the expression of Toll-like receptor 2
IL‑1β ECM remodelling, reduced type I collagen production84,85 (TLR2) mRNA103. Additionally, increased expression of
IL‑18 ECM remodelling and immune cell recruitment53
the TNF receptors in human Achilles tendon was noted
at mRNA and protein levels104.
IL‑33 ECM remodelling, increased type III collagen and cytokine production 91
IL‑6 Increased total collagen synthesis, cytokine feedback94,95 IL‑4 and IL‑13. Both IL‑4 and IL‑13 promote acute
TNF family Reduced type I collagen, increased proinflammatory cytokine
inflammatory processes, and their receptors are expressed
production53,104 on a number of cell types105 and are associated with ECM
homeostasis in many disease models. IL‑4 knockout
IL‑21R Proinflammatory cytokine induction111
(Il4−/−) mice display disorganised collagen orientation
IL‑17 ECM remodelling, increased type III collagen production, tenocyte and lower cross-sectional area and mechanical proper-
family apoptosis and cytokine production97,114 ties106 in a tendon injury model, whereas levels of IL‑10
IL‑4 • Reduced tendon strength in Il4−/− mice95 and IL‑13 in Il4−/− mice are comparable and even superior
IL‑13 • ECM remodelling107 to those of control mice95. Reverse transcriptase quantita-
IL‑15 • ECM remodelling and immune cell recruitment53 tive PCR (RT‑qPCR) experiments reveal that mRNAs for
ECM, extracellular matrix, IL‑21R, IL‑21 receptor. the IL‑4 receptor subunit alpha (IL‑4Rα), IL‑13Rα1 and
IL‑13Rα2, but not the common γ‑chain, are present in all
tested tendon tissues and cultured tenocytes107.
has been posited as a potential therapeutic approach for
autoimmune diseases such as RA90 with first‑in‑human The IL‑21–IL‑21R axis. The IL‑21 and IL‑21 receptor
dosing of an anti‑IL‑33 antibody (ANB020) enter- (IL‑21R) axis is a proinflammatory common‑γ chain
ing phase I clinical trials92. The safety and tolerability signalling cytokine system that has been associated with
observed in these trials will probably guide potential chronic inflammatory disease108,109. Tissue injury results
applications in patients with chronic tendinopathy. in upregulation of both IL21 and IL21R mRNA110, and its
downstream signalling pathway, STAT3, has been impli-
IL‑6 signalling. IL‑6 is a pleiotropic cytokine with a cated by tendinopathy array data97. We detected elevated
wide range of biological activities in immune regula- expression of IL‑21R mRNA and protein in human tendon
tion, haematopoiesis, inflammation and oncogenesis93. samples (in macrophages and tenocytes, in which these
Cyclic tensile strain increases expression of type I collagen levels were upregulated by proinflammatory cytokines
and IL‑6 in tendon fascicles from bovine foot extensors such TNF and IL‑1β in vitro) but found no convincing evi-
whereas cyclic loading increases IL‑6 expression in equine dence of the presence of IL‑21 at mRNA or protein level111.
superficial digital flexor tendons94. Patellar tendon injury
in Il6−/− mice show a trend towards lower angular devi- IL‑17 family. Experimental evidence points to the impor-
ation (more organized, normal tendon structure) and tance of the IL‑17 cytokine family in the pathogenesis of
a much higher modulus (greater overall strength)95. several immunoinflammatory diseases112,113. Recent tran-
Array data from human tendinopathic tissues from var- scriptomic analysis has highlighted increased expression
ious anatomical sites have shown that cyclical loading of of IL‑17F in human tendinopathy biopsy samples97. On
tenocytes96 increases IL‑6 production whereas damaged the basis of the aforementioned dataset and plausible bio-
tendons show aberrant regulation of the IL‑6 pathway, logical profile exhibited by IL‑17A, we hypothesized that
including decreased expression of IL‑6R and upregula- IL‑17A may play a part in tendinopathy. IL‑17A mRNA
tion of its intracellular regulator signal transducer and and protein expression levels were increased in early
activator of transcription 3 (STAT3)97. In humans, pro- human tendinopathic samples114. Furthermore, in human
longed running exercise results in concomitant increase tenocytes in vitro, IL‑17A regulated proinflammatory
of the concentration of IL‑6 and collagen synthesis in cytokines, key apoptotic mediators and ECM changes
the peritendinous tissue98. IL‑6 and IL‑8 concentrations toward a type III collagen phenotype. The availability
can increase immediately after ESWT and remain sig- of highly effective anti‑IL‑17A monoclonal antibodies
Cyclic tensile strain nificantly elevated for 4 h post-ESWT99. Thus, it seems such as secukinumab and ixekinumab, of proven effi-
The distribution of forces
that change over time in a
likely that IL‑6 delivers both proinflammatory and cacy in the treatment of enthesopathy associated to axial
repetitive fashion. anti-inflammatory effects in tendon disease similar to ankylosing spondylitis (SpA) and psoriatic arthritis115,116,
those seen in muscle pathology 100. renders IL‑17A an especially intriguing potential target
Cyclic loading in tendinopathy.
The application of repeated
TNF. Tendinopathy affecting the equine superficial digital
stresses, strains or stress
intensities. flexor tendon is associated with increases in both protein Neuronal inflammatory pathways
and mRNA levels of TNF and TNF receptor 1 (TNFR1) in Inflammatory responses are modulated by a bidirectional
Nociceptors tendon tissue101, whereas TNF mRNA is increased 11‑fold communication between the brain and the immune sys-
Sensory nerve cells that when torn supraspinatus tendon is compared with con- tem117. Inflammatory mediators are released from dam-
respond to damaging or
potentially damaging stimuli
trol tendon obtained during shoulder surgery 53. Cultured aged tissue and can stimulate nociceptors directly 118. PGE2
by sending signals to the human tenocytes treated with TNF reduce their type I derived from an arachidonic acid via the COX2 pathway
spinal cord and brain. collagen deposition and highly upregulate expression of is released from resident cells in damaged site tissue and

116 | FEBRUARY 2017 | VOLUME 13 www.nature.com/nrrheum


©
2
0
1
7
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
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

immune cells to promote inflammation and nociception119. macrophages134 and is central in controlling the duration
Both in vivo and in vitro evidence support the hypothesis and magnitude of the inflammatory response, providing
that PGE2 plays an important part in the development of endogenous stop signals for inflammation135. Resolution
tendinopathy18. Improved structural and material proper- pathways are programmed responses activated during
ties of the patellar tendon were found after PGE2 injection inflammation, resulting in a switch in lipid mediators
in vivo 120 and repetitive mechanical strain in human teno- from the dominance of prostaglandins to the lipoxins
cytes in vitro increases the production of PGE2 (REF. 121). to promote resolution of inflammation and the return
Inflammatory mediators have also been linked to the of tissues to their normal homeostatic state136. Studies
disturbance of glutaminergic pathways (glutamate is a in equine tendon have demonstrated increased expres-
major excitatory neurotransmitter in the nervous sys- sion of the pro-resolving receptor FPR274 and the switch
tem)122. Microarray analysis of a rodent running model from proinflammatory cytokines to the production of
of tendon overuse showed dysregulation of glutamate sig- pro-resolving lipids such as lipoxin A4 (LXA4) during
nalling in tendon tissue123. Glutamatergic and inflamma- the early stage of tendon injury. Importantly, expression
tory pathways are active in painful human tendinopathy, of FPR2 decreases with time after tendon injury and
despite there being no significant differences in basic ten- with ageing 137. This work was translated to a human
don histology between painful and pain-free tendons124. tendinopathy model system including examination of
This study showed that differences in specific glutamate supraspinatus tendinopathy samples from patients expe-
receptors — glutamate receptor ionotropic, kainate 4 riencing pain before and after surgical intervention in the
(GluK4, also known as KA1), metabotropic glutamate form of subacromial decompression138. Interestingly,
receptor 2 (mGluR2) and mGluR7 — and inflammatory the pro-resolving proteins FPR2 and chemokine-like
cell markers (CD45 and CD206) were associated with res- receptor 1 (CMKLR1, also known as ChemR23) were
olution of pain in rotator cuff tendinopathy 124. Moreover, increased in early-stage disease compared with inter-
a systematic review by the same group, analysing 27 stud- mediate and advanced tendinopathy samples. This work
ies, showed clear evidence of changes in the peripheral also suggests that tendon-derived stromal cells adopt a
neuronal phenotype in painful human tendinopathy 52. proinflammatory phenotype (expressing interferon reg-
The excitatory glutaminergic system was substantially ulatory factor 5 (IRF5), indoleamine 2,3‑dioxygenase 1
upregulated in seven studies, and four studies revealed (IDO1), p65 and nuclear factor NF‑κB) once exposed
a considerable increase in sensory neuropeptide expres- to an inflammatory cytokine milieu that primes the res-
sion. Thus, further mechanistic studies investigating the ident tenocytes to become hyper responsive to repeated
link between inflammatory and nociceptive mediators exposure to proinflammatory mediators.
are required to elucidate potential therapeutics that target
tendon pain and inflammatory–matrix interactions. Toward ‘translational tendinopathy’
Detailed mechanistic investigation of inflammatory
Nitric oxide pathways using the ‘molecule to clinical’ intervention
Nitric oxide (NO) is a signalling molecule that has a key paradigm has been remarkably successful in other
role in the pathogenesis of inflammation125. Following areas of musculoskeletal therapeutics, most notably in
injury to a tendon, NO is produced by all three isoforms of inflammatory arthropathies. In particular, the advent
nitric oxide synthase (NOS)126: NOS activity is upregulated of highly specific kinase immunomodulators has
in tendinopathy127. In a rat Achilles tendon model, the first increased the therapeutic armamentarium available
isoform to appear was inducible NOS (iNOS), followed by to the practising rheumatologist. Cytokine inhibition
endothelial NOS (eNOS) and then brain NOS (bNOS)128. has yielded transformative results for patients with RA
iNOS was expressed in macrophage-like cells and eNOS and axial spondyloarthropathies139. Despite increased
was found in endothelial cells, and all three isoforms were understanding of the molecular mediators involved in
expressed in tenocytes129. In an exercise-­induced overuse tendon disease, successful targeted mechanistic treat-
model of tendon degeneration, iNOS, eNOS and bNOS ments have so far eluded the field. Novel injectable
mRNAs were overexpressed in the supraspinatus ten- therapies, such as autologous stem cells and PRP, target
don of rats subjected to treadmill running for 14 days130. multiple downstream immune regulators with no clear
Expression of all isoforms was confirmed in human tendon definite pathway targeting 140,141. It is clear that further
disease from biopsy samples taken during shoulder sur- mechanistic work is required across the field to char-
gery131. Cultured human tenocytes exposed to exogenous acterise and subtype not only the key innate immune
NO increased total collagen synthesis132 whereas micro­ cells, together with their critical regulatory cytokine
array analysis showed substantial increases in type I colla- groups, but additionally the resident tenocytes in nor-
gen α1, type III collagen α1, type IV collagen α5, biglycan, mal versus diseased tendon pathologies. For example,
decorin, laminin and MMP10 (REF. 133). macrophage depletion in healing rodent Achilles ten-
don has been shown to increase tensile strength whereas
Pro-resolving mediators M2 macrophage recruitment was correlated with ten-
Specialised pro-resolving mediators, such as lipoxins, don regeneration in a validated ovine preclinical tendon
have an important mechanistic contribution to the regenerative model142, highlighting the critical role of
inflammation pathways involved in tendon disease. macrophages skewing towards an M2 phenotype dur-
N‑Formyl peptide receptor 2 (FPR2, also known as ing skeletal muscle regeneration143. Additionally, mast
ALX or ALX/FPR2) is expressed by monocytes and cells exert proinflammatory effects on human tenocytes

NATURE REVIEWS | RHEUMATOLOGY VOLUME 13 | FEBRUARY 2017 | 117


©
2
0
1
7
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
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 2 | Potential therapeutic agents targeting inflammatory pathways in tendinopathy


Potential targets Therapeutic agents used in musculoskeletal disease
Cytokines
IL‑1β • Anakinra (IL‑1Ra),
• Rilonacept (soluble IL‑1R that binds IL‑1β, IL‑1α and IL‑1Ra)
• Canakinumab (neutralizing anti‑IL‑1β IgG1 mAb)
TNF Infliximab, etanercept, adalimumab, certolizumab pegol and golimumab (anti-TNF mAbs)
IL‑6 • Tocilizumab, sarilumab, clazakizumab (anti‑IL‑6R mAbs)
• Sirukumab
IL‑17 Secukinumab, ixekizumab, brodalumab (anti‑IL‑17A mAbs)
IL‑33 ANB020 (anti‑IL‑33 antibody) in phase I trials
Small-molecule inhibitors
JAK–STAT signalling Tofacitinib and baricitinib (JAK inhibitors)
Resolution pathways
15‑epi‑LXA4 Aspirin
microRNA
miR‑29, miR‑210 MRG‑201 (synthetic miRNA‑29b mimic) and synthetic miR‑210 in preclinical studies
15‑epi‑LXA4, 15‑epi-lipoxin A4; IL‑1R, IL‑1 receptor; IL‑1Ra, IL‑1 receptor antagonist; IL‑6R, IL‑6 receptor; JAK, Janus kinase;
mAb, monoclonal antibody; STAT, signal transducer and activator of transcription.

whereas adipose-derived mesenchymal stromal cells describing the role for hypoxic tissue stress in human
co‑cultured with M1 macrophages successfully sup- tendinopathy showed that ERK and p38 inhibitors
pressed the effects of M1 macrophages on human teno- reduced proinflammatory cytokine/chemokine produc-
cytes by inducing a phenotypic switch from a M1 to M2 tion but did not modulate apoptosis in tenocytes148.
phenotype144. Thus, immune cell manipulation towards NAD-dependent protein deacetylase sirtuin‑1
a regenerative phenotype may provide promising ther- (SIRT1) is required for the inhibition of apoptosis and
apeutic options in tendinopathy. Several molecules inflammatory responses in human tenocytes acting via
(TABLE 2) have been highlighted as potential therapeu- NF‑kB signalling in vitro 149. Importantly, the SIRT1
tic targets in tendon disease. Moreover, appreciation activator resveratrol inhibited NF‑κB inflammatory
of the finely balanced ‘reparative’ versus ‘degenerative’ (COX‑2) and matrix degradation (MMP9) pathways,
inflammatory response in tendon damage is required demonstrating a potential role for resveratrol in the
to identify the molecular checkpoints that modify a treatment of tendinopathy. Curcumin (also known as
homeo­static inflammatory response toward aberrant diferuloylmethane), a naturally occurring polyphenol,
ECM emodelling and the chronic degenerative picture acts via NF‑κB inhibition. In vitro human tenocyte cul-
seen in clinical tendinopathy. tures revealed that curcumin suppressed IL‑1β‑induced
PI3K and p85/AKT activation and its association with
Pathway manipulation NF‑κB essential modulator IKK150. This study pro-
There is considerable interest in the development of posed a potential role for curcumin in treating tendon
highly specific molecules that can modulate intracellular inflammation through modulation of NF‑κB signal-
signal pathways known to confer the exquisite specific- ling, involving PI3K/Akt and the tendon-specific tran-
ity of cellular functions in the context of coordinated scription factor scleraxis in tenocytes. Pharmaceutical
inflammatory responses. Recently, JAK inhibitors have development of small-molecule MAPK inhibitors have
entered clinical use in RA, providing elegant proof of expanded rapidly over the past 5 years with numerous
concept for this approach. phase II trials in RA, all of which failed151,152; however,
Several animal models have highlighted the role of efforts in targeting the upstream JAK family of recep-
mitogen-activated protein kinase (MAPK) signalling tor tyrosine kinases have demonstrated efficacy and
within tenocyte ECM interactions. A rodent model of acceptable safety in RA153. Collectively, therefore, small-­
tendinopathy showed increased extracellular signal-­ molecule inhibitors targeting relevant signalling path-
regulated kinase (ERK) 1/2 signalling 145 whereas a way may provide promising avenues for development of
rodent model of plantaris tendon growth revealed inhi- novel tendinopathy therapies, particularly as inhibitors
bition of p38, which resulted in decreased IL‑6 expres- have several advantages over biologic agents in that they
sion, and had a modest effect on the expression of other can be administered orally and as synthetic compounds
ECM and cell proliferation genes146. ERK1/2 signalling they are comparatively inexpensive to manufacture.
was identified as having a critical role in the mechanism Moreover, by selectively blocking families of cytokine
whereby glucocorticoids inhibit proliferation and colla- effector functions they offer the capacity to modulate
gen synthesis in human tenocytes147. Our previous work integrated cellular regulatory systems in disease.

118 | FEBRUARY 2017 | VOLUME 13 www.nature.com/nrrheum


©
2
0
1
7
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
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Clinical cytokine manipulation IL‑6 significantly stimulated collagen synthesis in the


IL‑1 blockade. In a carrageenan-induced animal tendin- peritendinous tissue, highlighting the potential of tar-
opathy model the use of IL‑1Ra decreased the develop- geting crosstalk between inflammatory signalling and
ment of pathological changes154. It also showed marked ECM as a therapeutic approach.
reduction in key tendinopathic features including cell
infiltration, angiogenesis and cell density. The main Targeting pro-resolving molecules
limitation of this study was the use of an inflamma- Elucidating the cellular mechanisms by which resolu-
tory induced model system, which may not reflect tion of inflammation occurs and the key biochemical
the molecular signals of a chronic overuse lesion. A pathways associated with return to tissue homeostasis
follow‑up study using a stress-shielded rodent model may open new avenues for therapeutic interventions
showed that IL‑1Ra prevented morphological deterio- across a range of diseases associated with unresolved
ration and collagen metabolism of Achilles tendon after inflammation158. Investigation of resolution pathways
stress shielding 155 while IL‑1Ra effectively reduced the in human tendon disease in vitro has been prompted by
development of mechanical, chemical and histologic the observation of induced gene expression of CD206
changes seen in carrageenan-induced tendinopathy 154. and ALOX15 as a consequence of increased levels of
Anakinra is a recombinant, non-glycosylated form of STAT6 in tendon samples from patients with resolu-
human IL‑1Ra. In a human proof-of-concept study, 10 tion of symptoms after surgery 138. In the same study a
patients underwent ultrasonography guided, peritendi- stable lipoxin isoform, 15‑epi‑LXA4, induced by aspi-
nous injection of anakinra for chronic Achilles tendino­ rin, increased the expression of CD206, ALOX15 and
pathy 156. The investigators noted a substantial increase CCL22 mRNA in disease-derived tenocytes treated
in tendon thickness over time but no changes in walking with lipopolysaccharides. It is possible that low doses
pain, pain at rest or intratendinous blood flow in the of aspirin and its metabolites may act by triggering the
anakinra group while additionally reporting no tendon expression of anti-inflammatory and pro-resolving
ruptures. This pilot study comprised a non-randomized, mediators. Accordingly, aspirin-induced lipid mediators
non-blinded and non-controlled design, which limits offer potential therapeutics in tendinopathy and merit
its overall interpretation but demonstrates proof of con- future trials.
cept that targeting inflammatory cytokines is a testable
hypothesis in chronic human tendinopathy. Novel regulatory pathways in cellular activation
Emerging studies highlight microRNAs (mi­­­RNAs) as
TNF. On the basis of the role of TNF in tendinopathy, key epigenetic regulators of integrated mammalian cell
to date there has been one experimental medicine trial functions. Specific mi­­­RNAs have emerged that particu-
using a fully humanised anti-TNF monoclonal anti- larly regulate cytokine networks while orchestrating
body, adalimumab, which was administered to a group proliferation and differentiation of stromal lineages that
of 10 athletes with symptomatic unilateral tendinop- determine ECM composition159. mi­RNAs have provoked
athy for more than 6 months156. Conservative treat- considerable interest as regulators of musculoskeletal
ment (non-medical and non-surgical) had failed and diseases, although their precise contributions to com-
all the patients had ultrasonography‑determined spin- plex disease pathways remains uncertain160. miR‑210 has
dle-shaped thickening (1 mm) of the symptomatic ten- been reported as a crucial regulator of angiogenesis161, a
don in relation to the asymptomatic tendon. The patients key factor in tendon disease, and it accelerated healing of
were treated with ultrasonography‑guided peritendi- the tendon when injected into injured Achilles tendons
nous injections of adalimumab. Walking pain showed of rats162. miR‑210 also induced enhanced collagen qual-
a trend toward improvement at 7 days and 12 weeks ity, abundant vessels and upregulation of expression of
post-­treatment. Pain at rest was significantly reduced vascular endothelial growth factor, fibroblast growth fac-
after 7 days and again showed a trend (P = 0.07) towards tor 2 and type I collagen162. mi­­­RNAs designed and engi-
continued improvement at 12 weeks; all patients showed neered according to genetic sequences of transforming
significantly reduced blood flow in the tendon over the growth factor β1 (TGFβ1) and injected in the chicken
12‑week study period. Again, this pilot study comprised a tendon injury model achieved downregulation of TGFβ1
non-randomized, non-blinded and non-controlled design expression in vitro and in vivo 163. Some compounds can
but provides proof of principle of cytokine targeting in regulate endogenous miRNA expression, which may con-
human tendon disease. fer therapeutic value. One such study demonstrated that
miR‑29b mediated chitosan-induced prevention of ten-
IL‑6. In a small in vivo study assessing changes in the don adhesion after rodent Achilles tendon injury surgery
ECM as a result of recombinant cytokine therapy in by regulating the TGFβ1–Smad3 pathway 164.
human tendinopathy, levels of type I collagen turnover We investigated the role of miR29a in tendon disease
Carrageenan-induced
tendinopathy markers were measured in 14 healthy male volunteers, on the basis of our observations of IL‑33‑mediated ECM
Injection of a family of who had recombinant human IL‑6 (rhIL‑6) infused changes in human and rodent models91. We identified
carrageenans, linear sulfated into the peritendinous tissue of the Achilles tendon in reduced expression of miR‑29a in human tendino­
polysaccharides that are one leg, with the other leg serving as the control157. The pathy biopsy samples and demonstrated that its func-
extracted from red edible
seaweeds, directly into a
individuals were randomly assigned to either a resting tional reduction leads to development of tendinopathy.
mouse tendon to establish group or an exercise group performing a 1‑ h treadmill Importantly, in human tenocytes, miR‑29a was only capa-
tendinopathy. run (12 km/h, 2% uphill) before infusion. Infusion of ble of influencing the expression of type III collagen and

NATURE REVIEWS | RHEUMATOLOGY VOLUME 13 | FEBRUARY 2017 | 119


©
2
0
1
7
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
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

not type I. These data indicate that miR‑29a‑mediated at the tissue mechanistic level has delivered striking
regulation of type III collagen transcript stability is the transitional benefit to the patient. The historical dogma
predominant factor in the observed increase in type III of a lack of inflammation in tendinopathy has been
collagen production. Although it is likely that miR29a challenged over the past 10 years. Modern molecular
targets other genes in the tendon inflammatory–ECM techniques have clearly identified an inflammatory
mileu44,45, the reintroduction of miR‑29a to the tendon phenotype that is present throughout the spectrum of
injury could reverse the collagen switch that remains tendon disease despite the absence of obvious classical
a core pathological feature of tendinopathy. mi­­­RNAs clinical ‘inflammation’ signs. We contend that the cur-
have several important advantages in that they are bio­ rent lack of targeted therapeutics represents a failure to
developable and comprise known sequences that are properly embrace the potential in molecular and cellu-
often highly conserved among species, both attractive lar translational immune biology to deliver for tendino­
features from a drug development standpoint. The recent pathy patients. An ideal tendinopathy drug should blunt
initiation of a phase I clinical study of a synthetic miRNA the proinflammatory response and promote tendon
mimic to miR‑29b (MRG‑201) with possible extension pathology resolution by sparing inflammation-induced
to patients with cutaneous scleroderma may therefore healing moieties and encouraging robust and rapid ECM
lead to future miRNA therapies in tendinopathy. repair. Appropriately applied, modern ‘omic’ techniques
including transcriptome and proteome analysis should
Conclusions elucidate the key functional pathways implicated in
The precise functional contribution of inflammation in clinical disease. It is critical for the tendinopathy field to
human tendinopathy is not established among musculo- now embrace such experimental medicine approaches
skeletal clinicians and the scientific community. In several and move towards trialling therapies based on emerging
musculoskeletal diseases, rationalizing immunobiology molecular candidates.

1. Riley, G. Chronic tendon pathology: molecular basis 15. Xu, Y. & Murrell, G. A. The basic science of tendinopathy. 29. Cyranoski, D. Stem cells boom in vet clinics. Nature
and therapeutic implications. Expert Rev. Mol. Med. Clin. Orthop. Relat. Res. 466, 1528–1538 (2008). 496, 148–149 (2013).
7, 1–25 (2005). This article provides an overview of 30. Bahr, R., Fossan, B., Loken, S. & Engebretsen, L.
2. Riley, G. Tendinopathy — from basic science to pathophysiological mechanisms in tendon disease. Surgical treatment compared with eccentric training
treatment. Nat. Clin. Pract. Rheumatol. 4, 82–89 16. Abate, M. et al. Pathogenesis of tendinopathies: for patellar tendinopathy (Jumper’s Knee). A
(2008). inflammation or degeneration? Arthritis Res. Ther. 11, randomized, controlled trial. J. Bone Joint Surg. Am.
3. McGonagle, D., Marzo-Ortega, H., Benjamin, M. & 235 (2009). 88, 1689–1698 (2006).
Emery, P. Report on the Second International Informative review that debates the relative merits 31. Kirkendall, D. & Garrett, W. Function and
Enthesitis Workshop. Arthritis Rheum. 48, 896–905 of an inflammatory concept in the pathogeneis of biomechanics of tendons. Scand. J. Med. Sci. Sports
(2003). tendon disease. 7, 62–66 (1997).
4. Khan, K., Cook, J., Kannus, P., Maffulli, N. & Bonar, S. 17. Medzhitov, R. Origin and physiological roles of 32. Maffulli, N. & Benazzo, F. Basic Science of
Time to abandon the “tendinitis” myth: painful, inflammation. Nature 454, 428–435 (2008). Tendons. Sports Med. Arthrosc. Rev. 8, 1–5
overuse tendon conditions have a non-inflammatory 18. Dakin, S. G., Dudhia, J. & Smith, R. K. Resolving an (2000).
pathology. BMJ 324, 626 (2002). inflammatory concept: the importance of inflammation 33. O’Brien, M. Structure and metabolism of tendons.
5. Khan, K. M., Cook, J. L., Taunton, J. E. & Bonar, F. and resolution in tendinopathy. Vet. Immunol. Scand. J. Med. Sci. Sports 7, 55–61 (1997).
Overuse tendinosis, not tendinitis. Phys. Sportsmed. Immunopathol. 158, 121–127 (2014). 34. Thorpe, C. T. et al. Tendon overload results in
28, 38–48 (2000). 19. Maffulli, N., Khan, K. M. & Puddu, G. Overuse tendon alterations in cell shape and increased markers of
6. Felson, D. T. et al. Risk factors for incident conditions: time to change a confusing terminology. inflammation and matrix degradation. Scand. J. Med.
radiographic knee osteoarthritis in the elderly: the Arthroscopy 14, 840–843 (1998). Sci. Sports 25, e381–e391 (2015).
Framingham Study. Arthritis Rheum. 40, 728–733 This article describes the modern terminology and Important study that highlights the link between
(1997). classification of overuse tendinopathy. mechanical tendon overload and inflammatory
7. Pelletier, J. P., Martel-Pelletier, J. & Howell, D. S. in 20. Couppe, C., Svensson, R. B., Silbernagel, K. G., mechanisms in tendon disease.
Arthritis and Allied Conditions: a Textbook of Langberg, H. & Magnusson, S. P. Eccentric or 35. Thorpe, C. T., Riley, G. P., Birch, H. L., Clegg, P. D. &
Rheumatology 14th edn (eds Koopman, W. J.) concentric exercises for the treatment of Screen, H. R. Effect of fatigue loading on structure
2195–2245 (Lippincott Williams and Wilkins, tendinopathies? J. Orthop. Sports Phys. Ther. 45, and functional behaviour of fascicles from energy-
2000). 853–863 (2015). storing tendons. Acta Biomater. 10, 3217–3224
8. Berenbaum, F. Osteoarthritis as an inflammatory 21. Rio, E. et al. Isometric exercise induces analgesia and (2014).
disease (osteoarthritis is not osteoarthrosis!). reduces inhibition in patellar tendinopathy. Br. 36. Khan, K. M., Cook, J. L., Bonar, F., Harcourt, P. &
Osteoarthritis Cartilage 21, 16–21 (2013). J. Sports Med. 49, 1277–1283 (2015). Astrom, M. Histopathology of common
9. Kapoor, M., Martel-Pelletier, J., Lajeunesse, D., 22. Andres, B. M. & Murrell, G. A. Treatment of tendinopathies. Update and implications for
Pelletier, J. P. & Fahmi, H. Role of proinflammatory tendinopathy: what works, what does not, and what clinical management. Sports Med. 27, 393–408
cytokines in the pathophysiology of osteoarthritis. is on the horizon. Clin. Orthop. Relat. Res. 466, (1999).
Nat. Rev. Rheumatol. 7, 33–42 (2011). 1539–1554 (2008). This review describes the classical pathological
10. Sokolove, J. & Lepus, C. M. Role of inflammation in 23. Alfredson, H. & Cook, J. A treatment algorithm for features of tendinopathy.
the pathogenesis of osteoarthritis: latest findings and managing Achilles tendinopathy: new treatment 37. Kannus, P. & Jozsa, L. Histopathological changes
interpretations. Ther. Adv. Musculoskelet. Dis. 5, options. Br. J. Sports Med. 41, 211–216 (2007). preceding spontaneous rupture of a tendon. A
77–94 (2013). 24. Chan, K. M. & Fu, S. C. Anti-inflammatory controlled study of 891 patients. J. Bone Joint Surg.
11. Benjamin, M. & McGonagle, D. Basic concepts of management for tendon injuries — friends or foes? Am. 73, 1507–1525 (1991).
enthesis biology and immunology. J. Rheumatol. Sports Med. Arthrosc. Rehabil. Ther. Technol. 1, 23 38. Kannus, P., Jozsa, L. & Jarvinen, M. in Principles
Suppl. 83, 12–13 (2009). (2009). and Practice of Orthopaedic Sports Medicine
12. Benjamin, M. & McGonagle, D. The enthesis organ 25. Paoloni, J., De Vos, R. J., Hamilton, B., Murrell, G. A. (eds Garrett, W. E. Jr, Speer, K. P. &
concept and its relevance to the spondyloarthropathies. & Orchard, J. Platelet-rich plasma treatment for Kirkendall, D. T.) 21–37 (Lippincott Williams
Adv. Exp. Med. Biol. 649, 57–70 (2009). ligament and tendon injuries. Clin. J. Sport Med. 21, and Wilkins, 2000).
This review describes the concept of enthesitis as 37–45 (2011). 39. Maffulli, N., Barrass, V. & Ewen, S. W.
this condition has been considered analogous to 26. Krogh, T. P. et al. Treatment of lateral epicondylitis Light microscopic histology of achilles tendon
tendinopathy with platelet-rich plasma, glucocorticoid, or saline: a ruptures. A comparison with unruptured tendons.
13. Sherlock, J. P. et al. IL‑23 induces randomized, double-blind, placebo-controlled trial. Am. J. Sports Med. 28, 857–863 (2000).
spondyloarthropathy by acting on ROR-γ+CD3+CD4– Am. J. Sports Med. 41, 625–635 (2013). 40. Maffulli, N., Ewen, S. W., Waterston, S. W., Reaper, J.
CD8– entheseal resident T cells. Nat. Med. 18, 27. de Vos, R. J. et al. Platelet-rich plasma injection for & Barrass, V. Tenocytes from ruptured and
1069–1076 (2012). chronic Achilles tendinopathy: a randomized tendinopathic achilles tendons produce greater
14. Jacques, P. et al. Proof of concept: enthesitis and new controlled trial. JAMA 303, 144–149 (2010). quantities of type III collagen than tenocytes from
bone formation in spondyloarthritis are driven by 28. Smith, R. K. Mesenchymal stem cell therapy for equine normal achilles tendons. An in vitro model of human
mechanical strain and stromal cells. Ann. Rheum. Dis. tendinopathy. Disabil. Rehabil. 30, 1752–1758 tendon healing. Am. J. Sports Med. 28, 499–505
73, 437–445 (2014). (2008). (2000).

120 | FEBRUARY 2017 | VOLUME 13 www.nature.com/nrrheum


©
2
0
1
7
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
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

41. Maeda, E., Noguchi, H., Tohyama, H., Yasuda, K. & 63. Battery, L. & Maffulli, N. Inflammation in overuse tendon 86. Bauge, C., Leclercq, S., Conrozier, T. & Boumediene, K.
Hayashi, K. The tensile properties of collagen fascicles injuries. Sports Med. Arthrosc. 19, 213–217 (2011). TOL19‑001 reduces inflammation and MMP
harvested from regenerated and residual tissues in 64. Lories, R. J. & McInnes, I. B. Primed for inflammation: expression in monolayer cultures of tendon cells.
the patellar tendon after removal of the central third. enthesis-resident T cells. Nat. Med. 18, 1018–1019 BMC Complement. Altern. Med. 15, 217 (2015).
Biomed. Mater. Eng. 17, 77–85 (2007). (2012). 87. Dakin, S. G. et al. Proteomic analysis of tendon
42. Arnoczky, S. P., Lavagnino, M. & Egerbacher, M. 65. Buckley, C. D., Barone, F., Nayar, S., Benezech, C. & extracellular matrix reveals disease stage-specific
The mechanobiological aetiopathogenesis of Caamano, J. Stromal cells in chronic inflammation and fragmentation and differential cleavage of COMP
tendinopathy: is it the over-stimulation or the under- tertiary lymphoid organ formation. Annu. Rev. (cartilage oligomeric matrix protein). J. Biol. Chem.
stimulation of tendon cells? Int. J. Exp. Pathol. 88, Immunol. 33, 715–745 (2015). 289, 4919–4927 (2014).
217–226 (2007). 66. Iwamoto, T., Okamoto, H., Toyama, Y. & Momohara, S. 88. Schmitz, J. et al. IL‑33, an interleukin‑1‑like cytokine
43. Cook, J. L., Feller, J. A., Bonar, S. F. & Khan, K. M. Molecular aspects of rheumatoid arthritis: that signals via the IL‑1 receptor-related protein ST2
Abnormal tenocyte morphology is more prevalent chemokines in the joints of patients. FEBS J. 275, and induces T helper type 2‑associated cytokines.
than collagen disruption in asymptomatic athletes’ 4448–4455 (2008). Immunity 23, 479–490 (2005).
patellar tendons. J. Orthop. Res. 22, 334–338 67. Croft, A. P. et al. Rheumatoid synovial fibroblasts 89. Kakkar, R., Hei, H., Dobner, S. & Lee, R. T.
(2004). differentiate into distinct subsets in the presence of Interleukin 33 as a mechanically responsive
44. Jozsa, L. & Kannus, P. Histopathological findings in cytokines and cartilage. Arthritis Res. Ther. 18, 270 cytokine secreted by living cells. J. Biol. Chem. 287,
spontaneous tendon ruptures. Scand. J. Med. Sci. (2016). 6941–6948 (2012).
Sports 7, 113–118 (1997). 68. Dakin, S. G. et al. Persistence of tendon inflammation: a 90. Liew, F. Y., Pitman, N. I. & McInnes, I. B. Disease-
45. Pingel, J. et al. 3D ultrastructure and collagen leading role for the stroma? Proc. 4th Int. Scientif. associated functions of IL‑33: the new kid in the IL‑1
composition of healthy and overloaded human Tendinopathy Symposium. http://webcms.uct.ac.za/ family. Nat. Rev. Immunol. 10, 103–110 (2010).
tendon: evidence of tenocyte and matrix buckling. sites/default/files/image_tool/images/311/Documents/ 91. Millar, N. L. et al. MicroRNA29a regulates
J. Anat. 224, 548–555 (2014). ISTS%202016%20Programme%20booklet.pdf (2016) IL‑33‑mediated tissue remodelling in tendon disease.
46. Khan, K. M., Cook, J. L., Maffulli, N. & Kannus, P. 69. Kietrys, D. M. et al. Aging contributes to inflammation Nat. Commun. 6, 6774 (2015).
Where is the pain coming from in tendinopathy? It in upper extremity tendons and declines in forelimb This article describes cytokine and extracellular
may be biochemical, not only structural, in origin. agility in a rat model of upper extremity overuse. PLoS matrix (ECM) crosstalk and provides a mechanistic
Br. J. Sports Med. 34, 81–83 (2000). ONE 7, e46954 (2012). dissection of changes in collagen in the ECM in
47. De Jonge, S. et al. Relationship between 70. Pingel, J. et al. Increased mast cell numbers in a tendon disease.
neovascularization and clinical severity in Achilles calcaneal tendon overuse model. Scand. J. Med. Sci. 92. [No authors listed.] IL‑1 receptor–like 1 (IL1RL1; ST2);
tendinopathy in 556 paired measurements. Scand. Sports 23, e353–e360 (2013). IL‑33 (NF‑HEV). SciBX http://dx.doi.org/10.1038/
J. Med. Sci. Sports 24, 773–778 (2014). 71. Mantovani, A., Sica, A. & Locati, M. Macrophage scibx.2014.488 (2014).
48. Tol, J. L., Spiezia, F. & Maffulli, N. Neovascularization polarization comes of age. Immunity 23, 344–346 93. Scheller, J. & Rose-John, S. Interleukin‑6 and its
in Achilles tendinopathy: have we been chasing a red (2005). receptor: from bench to bedside. Med. Microbiol.
herring? Knee Surg. Sports Traumatol. Arthrosc. 20, 72. Mantovani, A., Sozzani, S., Locati, M., Allavena, P. & Immunol. 195, 173–183 (2006).
1891–1894 (2012). Sica, A. Macrophage polarization: tumor-associated 94. Legerlotz, K., Jones, E. R., Screen, H. R. & Riley, G. P.
49. Yuan, J., Murrell, G. A., Wei, A. Q. & Wang, M. X. macrophages as a paradigm for polarized M2 Increased expression of IL‑6 family members in
Apoptosis in rotator cuff tendonopathy. J. Orthop. mononuclear phagocytes. Trends Immunol. 23, tendon pathology. Rheumatology (Oxford) 51,
Res. 20, 1372–1379 (2002). 549–555 (2002). 1161–1165 (2012).
50. Arnoczky, S. P., Tian, T., Lavagnino, M. & Gardner, K. 73. Murray, P. J. et al. Macrophage activation and 95. Lin, T. W., Cardenas, L., Glaser, D. L. & Soslowsky, L. J.
Ex vivo static tensile loading inhibits MMP‑1 polarization: nomenclature and experimental Tendon healing in interleukin‑4 and interleukin‑6
expression in rat tail tendon cells through a guidelines. Immunity 41, 14–20 (2014). knockout mice. J. Biomech. 39, 61–69 (2006).
cytoskeletally based mechanotransduction 74. Dakin, S. G. et al. Macrophage sub-populations and 96. Legerlotz, K., Jones, G. C., Screen, H. R. & Riley, G. P.
mechanism. J. Orthop. Res. 22, 328–333 (2004). the lipoxin A4 receptor implicate active inflammation Cyclic loading of tendon fascicles using a novel fatigue
51. Mokone, G. G., Schwellnus, M. P., Noakes, T. D. & during equine tendon repair. PLoS ONE 7, e32333 loading system increases interleukin‑6 expression by
Collins, M. The COL5A1 gene and Achilles tendon (2012). tenocytes. Scand. J. Med. Sci. Sports 23, 31–37
pathology. Scand. J. Med. Sci. Sports 16, 19–26 75. Hackett, L., Millar, N. L., Lam, P. & Murrell, G. A. Are (2013).
(2006). the symptoms of calcific tendinitis due to 97. Jelinsky, S. A. et al. Regulation of gene expression in
52. Dean, B. J., Franklin, S. L. & Carr, A. J. The peripheral neoinnervation and/or neovascularization? J. Bone human tendinopathy. BMC Musculoskelet. Disord.
neuronal phenotype is important in the pathogenesis Joint Surg. Am. 98, 186–192 (2016). 12, 86 (2011).
of painful human tendinopathy: a systematic review. 76. Matthews, T. J., Hand, G. C., Rees, J. L., Important array data from human tendon samples
Clin. Orthop. Relat. Res. 471, 3036–3046 (2013). Athanasou, N. A. & Carr, A. J. Pathology of the torn that highlight many of the inflammatory pathways
53. Millar, N. L., Wei, A. Q., Molloy, T. J., Bonar, F. & rotator cuff tendon. Reduction in potential for repair described subsequently.
Murrell, G. A. Cytokines and apoptosis in as tear size increases. J. Bone Joint Surg. Br. 88, 98. Langberg, H., Olesen, J. L., Gemmer, C. & Kjaer, M.
supraspinatus tendinopathy. J. Bone Joint Surg. Br. 489–495 (2006). Substantial elevation of interleukin‑6 concentration in
91, 417–424 (2009). First paper to highlight resident and infiltrating peritendinous tissue, in contrast to muscle, following
This article introduces the concept of cytokine immune cells in spectrum of tendon disease. prolonged exercise in humans. J. Physiol. 542,
biology in human tendon disease 77. Behzad, H., Sharma, A., Mousavizadeh, R., Lu, A. & 985–990 (2002).
54. Del Buono, A., Battery, L., Denaro, V., Maccauro, G. & Scott, A. Mast cells exert pro-inflammatory effects of 99. Waugh, C. M. et al. In vivo biological response to
Maffulli, N. Tendinopathy and inflammation: some relevance to the pathophyisology of tendinopathy. extracorporeal shockwave therapy in human
truths. Int. J. Immunopathol. Pharmacol. 24, 45–50 Arthritis Res. Ther. 15, R184 (2013). tendinopathy. Eur. Cells Mater. 29, 268–280 (2015).
(2011). 78. Marsolais, D., Cote, C. H. & Frenette, J. 100. Pedersen, B. K., Steensberg, A. & Schjerling, P.
55. Sharma, P. & Maffulli, N. Tendon injury and Neutrophils and macrophages accumulate Muscle-derived interleukin‑6: possible biological
tendinopathy: healing and repair. J. Bone Joint Surg. sequentially following Achilles tendon injury. effects. J. Physiol. 536, 329–337 (2001).
Am. 87, 187–202 (2005). J. Orthop. Res. 19, 1203–1209 (2001). 101. Hosaka, Y., Kirisawa, R., Ueda, H., Yamaguchi, M. &
56. Cook, J. L. & Purdam, C. R. Is tendon pathology a 79. Dean, B. J., Gettings, P., Dakin, S. G. & Carr, A. J. Takehana, K. Differences in tumor necrosis factor
continuum? A pathology model to explain the clinical Are inflammatory cells increased in painful human (TNF)alpha and TNF receptor‑1‑mediated intracellular
presentation of load-induced tendinopathy. Br. tendinopathy? A systematic review. Br. J. Sports Med. signaling factors in normal, inflamed and scar-formed
J. Sports Med. 43, 409–416 (2009). 50, 216–220 (2016). horse tendons. J. Vet. Med. Sci. 67, 985–991 (2005).
57. Fu, S. C., Rolf, C., Cheuk, Y. C., Lui, P. P. & Chan, K. M. A systematic review that highlights clear evidence 102. John, T. et al. Effect of pro-inflammatory and
Deciphering the pathogenesis of tendinopathy: a of immune cells in tendinopathy. immunoregulatory cytokines on human tenocytes.
three-stages process. Sports Med. Arthrosc. Rehabil. 80. Millar, N. L. et al. Inflammation is present in early J. Orthop. Res. 28, 1071–1077 (2010).
Ther. Technol. 2, 30 (2010). human tendinopathy. Am. J. Sports Med. 38, 103. de Mos, M. et al. Tendon degeneration is not
58. Alfredson, H., Thorsen, K. & Lorentzon, R. In situ 2085–2091 (2010). mediated by regulation of Toll-like receptors 2 and 4
microdialysis in tendon tissue: high levels of 81. Kragsnaes, M. S. et al. Stereological quantification of in human tenocytes. J. Orthop. Res. 27, 1043–1047
glutamate, but not prostaglandin E2 in chronic immune-competent cells in baseline biopsy specimens (2009).
Achilles tendon pain. Knee Surg. Sports Traumatol. from achilles tendons: results from patients with 104. Gaida, J. E., Alfredson, H., Forsgren, S. & Cook, J. L. A
Arthrosc. 7, 378–381 (1999). chronic tendinopathy followed for more than 4 years. pilot study on biomarkers for tendinopathy: lower
59. Alfredson, H. & Lorentzon, R. Chronic tendon pain: no Am. J. Sports Med. 42, 2435–2445 (2014). levels of serum TNF-alpha and other cytokines in
signs of chemical inflammation but high 82. Schubert, T. E., Weidler, C., Lerch, K., Hofstadter, F. & females but not males with Achilles tendinopathy.
concentrations of the neurotransmitter glutamate. Straub, R. H. Achilles tendinosis is associated with BMC Sports Sci. Med. Rehabil. 8, 5 (2016).
Implications for treatment? Curr. Drug Targets 3, sprouting of substance P positive nerve fibres. Ann. 105. Hershey, G. K. IL‑13 receptors and signaling
43–54 (2002). Rheum. Dis. 64, 1083–1086 (2005). pathways: an evolving web. J. Allergy Clin. Immunol.
60. Khan, K. M., Cook, J. L., Kannus, P., Maffulli, N. & 83. Lui, P. P., Maffulli, N., Rolf, C. & Smith, R. K. What are 111, 677–690 (2003).
Bonar, S. F. Time to abandon the “tendinitis” myth. the validated animal models for tendinopathy? Scand. 106. Lin, T. W., Cardenas, L. & Soslowsky, L. J. Tendon
BMJ 324, 626–627 (2002). J. Med. Sci. Sports 21, 3–17 (2011). properties in interleukin‑4 and interleukin‑6 knockout
61. Millar, N. L., Dean, B. J. & Dakin, S. G. Inflammation 84. Archambault, J., Tsuzaki, M., Herzog, W. & mice. J. Biomech. 38, 99–105 (2005).
and the continuum model: time to acknowledge the Banes, A. J. Stretch and interleukin‑1beta induce 107. Courneya, J. P. et al. Interleukins 4 and 13 modulate
molecular era of tendinopathy. Br. J. Sports Med. 50, matrix metalloproteinases in rabbit tendon cells gene expression and promote proliferation of primary
1486 (2016). in vitro. J. Orthop. Res. 20, 36–39 (2002). human tenocytes. Fibrogen. Tissue Repair 3, 9 (2010).
62. Blazina, M. E., Kerlan, R. K., Jobe, F. W., Carter, V. S. 85. Tsuzaki, M. et al. IL‑1 beta induces COX2, MMP‑1, -3 108. Nurieva, R. et al. Essential autocrine regulation by
& Carlson, G. J. Jumper’s knee. Orthop. Clin. North and -13, ADAMTS‑4, IL‑1 beta and IL‑6 in human IL‑21 in the generation of inflammatory T cells. Nature
Am. 4, 665–678 (1973). tendon cells. J. Orthop. Res. 21, 256–264 (2003). 448, 480–483 (2007).

NATURE REVIEWS | RHEUMATOLOGY VOLUME 13 | FEBRUARY 2017 | 121


©
2
0
1
7
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
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

109. Spolski, R. & Leonard, W. J. Interleukin‑21: a double- 130. Murrell, G. A. Oxygen free radicals and tendon 149. Busch, F., Mobasheri, A., Shayan, P., Stahlmann, R. &
edged sword with therapeutic potential. Nat. Rev. healing. J. Shoulder Elbow Surg. 16 (5 Suppl.), Shakibaei, M. Sirt‑1 is required for the inhibition of
Drug Discov. 13, 379–395 (2014). S208–S214 (2007). apoptosis and inflammatory responses in human
110. Foster, P. S. & Mattes, J. IL‑21 comes of age. Immunol. 131. Murrell, G. A. Using nitric oxide to treat tendinopathy. tenocytes. J. Biol. Chem. 287, 25770–25781
Cell Biol. 87, 359–360 (2009). Br. J. Sports Med. 41, 227–231 (2007). (2012).
111. Campbell, A. L. et al. IL‑21 receptor expression in 132. Xia, W., Szomor, Z., Wang, Y. & Murrell, G. A. 150. Buhrmann, C. et al. Curcumin modulates nuclear
human tendinopathy. Mediators Inflamm. 2014, Nitric oxide enhances collagen synthesis in cultured factor κB (NF‑κB)-mediated inflammation in
481206 (2014). human tendon cells. J. Orthop. Res. 24, 159–172 human tenocytes in vitro: role of the
112. Lubberts, E. The IL‑23–IL‑17 axis in inflammatory (2006). phosphatidylinositol 3‑kinase/Akt pathway.
arthritis. Nat. Rev. Rheumatol. 11, 415–429 (2015). 133. Molloy, T. J., de Bock, C. E., Wang, Y. & Murrell, G. A. J. Biol. Chem. 286, 28556–28566 (2011).
113. Lubberts, E. et al. IL‑1‑independent role of IL‑17 in Gene expression changes in SNAP-stimulated and 151. McInnes, I. B. & O’Dell, J. R. State‑of‑the-art:
synovial inflammation and joint destruction during iNOS-transfected tenocytes — expression of rheumatoid arthritis. Ann. Rheum. Dis. 69,
collagen-induced arthritis. J. Immunol. 167, extracellular matrix genes and its implications for 1898–1906 (2010).
1004–1013 (2001). tendon-healing. J. Orthop. Res. 24, 1869–1882 152. Genovese, M. C. Inhibition of p38: has the fat lady
114. Millar, N. L. et al. IL‑17A mediates inflammatory and (2006). sung? Arthritis Rheum. 60, 317–320 (2009).
tissue remodelling events in early human 134. Serhan, C. N. & Savill, J. Resolution of inflammation: 153. Boyle, D. L. et al. The JAK inhibitor tofacitinib
tendinopathy. Sci. Rep. 6, 27149 (2016). the beginning programs the end. Nat. Immunol. 6, suppresses synovial JAK1–STAT signalling in
115. McInnes, I. B. et al. Secukinumab, a human anti- 1191–1197 (2005). rheumatoid arthritis. Ann. Rheum. Dis. 74, 1311–1316
interleukin‑17A monoclonal antibody, in patients with 135. Buckley, C. D., Gilroy, D. W. & Serhan, C. N. (2015).
psoriatic arthritis (FUTURE 2): a randomised, double- Proresolving lipid mediators and mechanisms in the 154. Berkoff, D. J., Kallianos, S. A., Eskildsen, S. M. &
blind, placebo-controlled, phase 3 trial. Lancet 386, resolution of acute inflammation. Immunity 40, Weinhold, P. S. Use of an IL1‑receptor antagonist to
1137–1146 (2015). 315–327 (2014). prevent the progression of tendinopathy in a rat
116. Mease, P. J. et al. Ixekizumab, an interleukin‑17A 136. Serhan, C. N. Pro-resolving lipid mediators are leads model. J. Orthop. Res. 34, 616–622 (2016).
specific monoclonal antibody, for the treatment of for resolution physiology. Nature 510, 92–101 155. Ma, Y., Yan, X., Zhao, H. & Wang, W. Effects of
biologic-naive patients with active psoriatic arthritis: (2014). interleukin‑1 receptor antagonist on collagen and
results from the 24‑week randomised, double-blind, 137. Dakin, S. G. et al. Inflamm-aging and arachadonic acid matrix metalloproteinases in stress-shielded achilles
placebo-controlled and active (adalimumab)-controlled metabolite differences with stage of tendon disease. tendons of rats. Orthopedics 35, e1238–e1244
period of the phase III trial SPIRIT‑P1. Ann. Rheum. PLoS ONE 7, e48978 (2012). (2012).
Dis. 76, 79–87 (2016). 138. Dakin, S. G. et al. Inflammation activation and 156. Fredberg, U. & Ostgaard, R. Effect of ultrasound-
117. Marchand, F., Perretti, M. & McMahon, S. B. Role of resolution in human tendon disease. Sci. Transl. Med. guided, peritendinous injections of adalimumab and
the immune system in chronic pain. Nat. Rev. 7, 311ra173 (2015). anakinra in chronic Achilles tendinopathy: a pilot
Neurosci. 6, 521–532 (2005). Study showing inflammatory signatures defining study. Scand. J. Med. Sci. Sports 19, 338–344
118. Basbaum, A. I., Bautista, D. M., Scherrer, G. & resolution of symptoms in supraspinatus tendons (2009).
Julius, D. Cellular and molecular mechanisms of pain. from patients experiencing pain before and after 157. Andersen, M. B., Pingel, J., Kjaer, M. & Langberg, H.
Cell 139, 267–284 (2009). surgical treatment. Interleukin‑6: a growth factor stimulating collagen
119. Ohishi, S. et al. Evidence for involvement of 139. Schett, G., Elewaut, D., McInnes, I. B., Dayer, J. M. & synthesis in human tendon. J. Appl. Physiol. (1985)
prostaglandin I2 as a major nociceptive mediator in Neurath, M. F. How cytokine networks fuel 110, 1549–1554 (2011).
acetic acid-induced writhing reaction: a study using inflammation: toward a cytokine-based disease 158. Gilroy, D. W., Lawrence, T., Perretti, M. & Rossi, A. G.
IP‑receptor disrupted mice. Adv. Exp. Med. Biol. 469, taxonomy. Nat. Med. 19, 822–824 (2013). Inflammatory resolution: new opportunities for drug
265–268 (1999). Good general overview for musculoskeletal discovery. Nat. Rev. Drug Discov. 3, 401–416
120. Ferry, S. T., Afshari, H. M., Lee, J. A., Dahners, L. E. & practitioners on understanding how cytokines are (2004).
Weinhold, P. S. Effect of prostaglandin E2 injection on involved in rheumatic disease. 159. Bushati, N. & Cohen, S. M. microRNA functions.
the structural properties of the rat patellar tendon. 140. Andia, I., Rubio-Azpeitia, E. & Maffulli, N. Platelet-rich Annu. Rev. Cell Dev. Biol. 23, 175–205 (2007).
Sports Med. Arthrosc. Rehabil. Ther. Technol. 4, 2 plasma modulates the secretion of inflammatory/ 160. Bartel, D. P. MicroRNAs: target recognition and
(2012). angiogenic proteins by inflamed tenocytes. Clin. regulatory functions. Cell 136, 215–233 (2009).
121. Jones, E. R., Jones, G. C., Legerlotz, K. & Riley, G. P. Orthop. Relat. Res. 473, 1624–1634 (2015). 161. Zeng, L. et al. MicroRNA‑210 overexpression
Cyclical strain modulates metalloprotease and matrix 141. Young, M. Stem cell applications in tendon disorders: induces angiogenesis and neurogenesis in the
gene expression in human tenocytes via activation of a clinical perspective. Stem Cells Int. 2012, 637836 normal adult mouse brain. Gene Ther. 21, 37–43
TGFbeta. Biochim. Biophys. Acta 1833, 2596–2607 (2012). (2014).
(2013). 142. de la Durantaye, M., Piette, A. B., van Rooijen, N. & 162. Usman, M. A. et al. The effect of administration of
122. Kidd, B. L. & Urban, L. A. Mechanisms of Frenette, J. Macrophage depletion reduces cell double stranded microRNA‑210 on acceleration of
inflammatory pain. Br. J. Anaesth. 87, 3–11 (2001). proliferation and extracellular matrix accumulation Achilles tendon healing in a rat model. J. Orthop. Sci.
123. Molloy, T. J., Kemp, M. W., Wang, Y. & Murrell, G. A. but increases the ultimate tensile strength of injured 20, 538–546 (2015).
Microarray analysis of the tendinopathic rat Achilles tendons. J. Orthop. Res. 32, 279–285 163. Chen, C. H. et al. Effectiveness of microRNA in
supraspinatus tendon: glutamate signaling and its (2014). down-regulation of TGF-β gene expression in digital
potential role in tendon degeneration. J. Appl. 143. Mounier, R. et al. AMPKα1 regulates macrophage flexor tendons of chickens: in vitro and in vivo
Physiol. 101, 1702–1709 (2006). skewing at the time of resolution of inflammation study. J. Hand Surg. Am. 34, 1777–1784.e1
124. Dean, B. J. et al. Differences in glutamate during skeletal muscle regeneration. Cell. Metab. 18, (2009).
receptors and inflammatory cell numbers are 251–264 (2013). 164. Chen, Q., Lu, H. & Yang, H. Chitosan inhibits
associated with the resolution of pain in human 144. Manning, C. N. et al. Adipose-derived mesenchymal fibroblasts growth in Achilles tendon via TGF‑β1/
rotator cuff tendinopathy. Arthritis Res. Ther. 17, stromal cells modulate tendon fibroblast responses to Smad3 pathway by miR‑29b. Int. J. Clin. Exp. Pathol.
176 (2015). macrophage-induced inflammation in vitro. Stem Cell 7, 8462–8470 (2014).
125. Palmer, R. M., Ashton, D. S. & Moncada, S. Vascular Res. Ther. 6, 74 (2015).
endothelial cells synthesize nitric oxide from 145. Scott, A. et al. Tenocyte responses to mechanical Acknowledgements
L‑arginine. Nature 333, 664–666 (1988). loading in vivo: a role for local insulin-like growth N.L.M is supported by a Wellcome Trust Postdoctoral
126. Murrell, G. A. et al. Modulation of tendon healing by factor 1 signaling in early tendinosis in rats. Arthritis Fellowship (WT100651MA) and grants from Arthritis
nitric oxide. Inflamm. Res. 46, 19–27 (1997). Rheum. 56, 871–881 (2007). Research UK (Ref 21346) and the Royal College of Surgeons
127. Szomor, Z. L. et al. Differential expression of cytokines 146. Schwartz, A. J. et al. p38 MAPK signaling in postnatal of Edinburgh.
and nitric oxide synthase isoforms in human rotator tendon growth and remodeling. PLoS ONE 10,
cuff bursae. Ann. Rheum. Dis. 60, 431–432 (2001). e0120044 (2015). Author contributions
128. Lin, J. H. et al. Temporal expression of nitric oxide 147. Poulsen, R. C., Carr, A. J. & Hulley, P. A. Protection N.L.M. researched data for the article. N.L.M. and I.B.M.
synthase isoforms in healing Achilles tendon. against glucocorticoid-induced damage in human contributed substantially to discussions of the content and
J. Orthop. Res. 19, 136–142 (2001). tenocytes by modulation of ERK, Akt, and forkhead wrote the article. All of the authors reviewed and edited the
129. Lin, J., Wang, M. X., Wei, A., Zhu, W. & Murrell, G. A. signaling. Endocrinology 152, 503–514 (2011). manuscript before submission.
The cell specific temporal expression of nitric oxide 148. Millar, N. L. et al. Hypoxia: a critical regulator of early
synthase isoforms during achilles tendon healing. human tendinopathy. Ann. Rheum. Dis. 71, 302–310 Competing interests statement
Inflamm. Res. 50, 515–522 (2001). (2012). The authors declare no competing interests.

122 | FEBRUARY 2017 | VOLUME 13 www.nature.com/nrrheum


©
2
0
1
7
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
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like