You are on page 1of 16

757443

research-article2018
JHCXXX10.1369/0022155418757443Tenascin-C and Innate ImmunityMarzeda and Midwood

Review
Journal of Histochemistry & Cytochemistry 2018, Vol. 66(4) 289–304
© The Author(s) 2018
Reprints and permissions:
sagepub.com/journalsPermissions.nav
DOI: 10.1369/0022155418757443
https://doi.org/10.1369/0022155418757443
journals.sagepub.com/home/jhc

Internal Affairs: Tenascin-C as a Clinically Relevant,


Endogenous Driver of Innate Immunity

Anna M. Marzeda and Kim S. Midwood


Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford,
UK (AMM, KSM)

Journal of Histochemistry & Cytochemistry


Summary
To protect against danger, the innate immune system must promptly and accurately sense alarm signals, and mount an
appropriate response to restore homeostasis. One endogenous trigger of immunity is tenascin-C, a large hexameric protein
of the extracellular matrix. Upregulated upon tissue injury and cellular stress, tenascin-C is expressed during inflammation
and tissue remodeling, where it influences cellular behavior by interacting with a multitude of molecular targets, including
other matrix components, cell surface proteins, and growth factors. Here, we discuss how these interactions confer upon
tenascin-C distinct immunomodulatory capabilities that make this matrix molecule necessary for efficient tissue repair.
We also highlight in vivo studies that provide insight into the consequences of misregulated tenascin-C expression on
inflammation and fibrosis during a wide range of inflammatory diseases. Finally, we examine how its unique expression
pattern and inflammatory actions make tenascin-C a viable target for clinical exploitation in both diagnostic and therapeutic
arenas. (J Histochem Cytochem 66:289–304, 2018)

Keywords
extracellular matrix, fibroblast, fibrosis, inflammation, inflammatory disease, innate immunity, integrin, macrophage, toll-like
receptor 4

Introduction However, infection is not the sole threat to homeo-


stasis. Inflammatory responses are also activated by
The innate immune system is constantly challenged entities of non-microbiological origin, for example, by a
by danger of diverse origin. Traditionally, its primary wide variety of endogenous stimuli generated as a
function was considered to be the recognition of non- result of cellular stress and tissue damage. These sig-
self entities. Indeed, upon infection, the presence of a nals include molecules released upon cell death and
pathogen is sensed by innate immune cells through components of damaged extracellular matrix, which
membrane-bound and intracellular pattern recognition are vital for triggering pathways designed to clear cel-
receptors (PRRs). The identification of highly con- lular debris and restore tissue integrity.2–4 Furthermore,
served pathogen-derived molecular motifs, collectively
referred to as pathogen-associated molecular patterns
Received for publication September 29, 2017; accepted November 29,
(PAMPs), by PRRs triggers cells to activate an inflam-
2017.
matory response. Following the release of chemo-
kines, phagocytes and antigen-presenting cells are Corresponding Author:
Kim S. Midwood, Kennedy Institute of Rheumatology, Nuffield
recruited to swiftly destroy the interloping pathogen,
Department of Orthopaedics, Rheumatology and Musculoskeletal
prevent widespread tissue colonization, and shape Sciences, University of Oxford, Roosevelt Drive, Headington, Oxford,
targeted adaptive responses to confer long-lasting OX3 7FY, UK.
immunity against recurrent invasion.1 E-mail: kim.midwood@kennedy.ox.ac.uk
290 Marzeda and Midwood Marzeda and Midwood

the immune system can be activated by inorganic irri- is an important regulator of the matrix itself, modulat-
tants, such as asbestos5; by deposits of endogenous ing its assembly and the biophysical properties of tis-
molecules, such as amyloid beta and cholesterol6,7; sue. Within the extracellular matrix, tenascin-C binding
and by tumor-associated antigens.8 Thus, it appears to partners include collagen, fibronectin, periostin, fibril-
be not a simple matter of discriminating between lin-2, and proteoglycans: perlecan, aggrecan, neuro-
harmful foreign and benign self, but rather detecting can, and phosphacan/receptor protein tyrosine
any potential threat, regardless of origin. phosphatase β/ζ. Tenascin-C also binds to proteins
Many questions around this model of immunity exposed on the cell surface, such as syndecan 4, con-
remain unanswered, not least how the identity of endog- tactin, glypican, annexin II, and CALEB/CSPG5. It can
enous inflammatory molecules is defined, and the further directly influence internal cell signaling by bind-
mechanisms by which their activity must be tightly regu- ing to cell surface receptors such as epidermal growth
lated to avoid catastrophic autoimmune destruction. factor receptor (EGFR), toll-like receptor 4 (TLR4), and
Moreover, while the induction of inflammatory responses integrins. Moreover, tenascin-C is able to bind numer-
by PAMPs is relatively well understood,9 the mode of ous growth factors, including members of the fibroblast
action of endogenous inflammatory triggers remains growth factor family and the transforming growth factor
elusive. Answering these questions will not only provide beta (TGF-β) superfamily. This multitude of interac-
insight into an intriguing immunological puzzle but may tions makes tenascin-C a diverse modulator of cellular
also open new therapeutic avenues to treat inflamma- behavior, such as proliferation, migration, adhesion,
tory diseases of a non-infectious origin, where pathol- and differentiation, capable of producing highly con-
ogy is driven by endogenous inflammatory stimuli. text- and cell-type-specific responses. Here, we dis-
Tenascin-C is a large hexameric protein of the cuss further a selection of these interactions by which
extracellular matrix that exhibits limited expression in tenascin-C mediates, and modulates, inflammatory
healthy tissues, but which is rapidly upregulated upon responses.
tissue damage. Typically, its expression is transient,
with mRNA downregulated and protein cleared by the
The Proinflammatory Capabilities of
time tissue repair is complete.10 However, persistent
expression of tenascin-C is associated with a wide Tenascin-C
variety of pathological conditions, where it accumu- To date, three main binding partners have been identi-
lates at sites of inflammation, for example, in autoim- fied to be responsible for the inflammatory effects of
mune, fibrotic, and metabolic diseases, and in many tenascin-C: TLR4, and integrins α9β1 and αVβ3. The
cancers.11 Despite this well-established pattern of impact of tenascin-C activation of these receptors is
expression, it was not until comparatively recently that summarized in Table 1.
the ability of tenascin-C to trigger inflammation, and TLR4 is a cell surface receptor present on leuko-
directly shape immune responses, was reported. In cytes and stromal cells. It was first identified as the
this review, we present the current understanding of PRR responsible for mediating innate immune
tenascin-C’s involvement in endogenous immunity, responses to the potent bacterial virulence factor, lipo-
and the data emerging that implicate this matrix mol- polysaccharide (LPS) or endotoxin, but it also has a
ecule as a promising biomarker of inflammatory dis- growing number of endogenous activators; this list
ease status and a tractable target for therapeutic includes tenascin-C, biglycan, fibrinogen, and S100
applications. proteins, among many others.26 The most widely
reported effect of TLR4 activation by tenascin-C is the
Tenascin-C Up-Close: A Diverse induction of soluble proinflammatory mediators, such
as interleukin (IL)-6, IL-8, and tumor necrosis factor
Interaction Partner (TNF). This has been observed in a number of cell
Tenascin-C is characterized by a distinct domain orga- types, including macrophages,13,18,21,22 dendritic cells
nization. An assembly domain at the N-terminus, (DCs),17 fibroblasts,13,20 and chondrocytes.14 More
responsible for the formation of the six-armed oligomer recently, tenascin-C signaling through TLR4 has been
known as the hexabrachion, is followed by a series of implicated in inflammasome priming. Inflammasomes
epidermal growth factor-like (EGF-L) repeats, constitu- are multiprotein, intracellular complexes that drive
tively expressed and alternatively spliced fibronectin specific inflammatory responses including the release
type III-like repeats (FNIII), and a C-terminal fibrino- of IL-1β and IL-18, and induction of pyroptosis, a type
gen-like globe (FBG). This complex structure enables of programmed cell death.27 Typically, inflammasomes
tenascin-C to establish a wide variety of molecular require two separate signals for activation: The first
interactions (reviewed in Midwood et al.12). Tenascin-C “priming” signal comprises PRR-driven procytokine
Table 1. Tenascin-C as a Modulator of Cellular Behavior in Inflammation.
Receptor Cell Type TN-C Source Response TN-C Domain Evidence Pathway Reference

TLR4 Primary human macrophages Recombinant human full-length TN-C ↑ IL-6, IL-8, and TNF FBG anti-TLR4 Ab and TLR4 KO mouse NF-kB Myd88- Midwood et al.13
purified from HEK cells; recombinant macrophages dependent
Human synovial fibroblasts human TN-C domainsa,b ↑ IL-6 Myd88 and TLR4 KO mouse
embryonic fibroblasts
Primary human Full-length TN-C purified from human ↑ IL-6, IL-8, prostaglandin E2, TLR4 inhibitor TAK242 Patel et al.14
chondrocytes glioblastoma cell linea,b nitrate, and ADAMTS4
Mouse bone marrow ↑ MMP-9 TLR4 KO neutrophils Kuriyama et al.15
Tenascin-C and Innate Immunity

neutrophils
Human THP-1-derived Recombinant human TN-C purified from ↑ macrophage differentiation anti-TLR4 Ab ↑ CD36, oxLDL Liu et al.16
macrophages mouse myeloma cell line into foam cells scavenger receptor
Mouse bone marrow– Full-length TN-C purified from human ↑ IL-6, IL-1α, and IL-1β; ↑ IL-17 TLR4 inhibitor TAK242 NF-kB Machino-Ohtsuka
derived DCs glioblastoma cell line in cocultures of naive T cells et al.17
and DCs
a,b
Human peripheral blood Recombinant human FBG domain ↑ IL-6, IL-8, TNF, IL-10, FBG anti-TLR4 Ab and TLR4 inhibitor p38, JNK, and NF-kB Piccinini et al.18
monocyte–derived collagen (expression and TAK242
macrophages phosphorylation), MMP-1, and
MMP-14
Human and mouse Full-length TN-C purified from human ↑ type I collagen, αSMA, and TLR4 inhibitor TAK242, Myd88 NF-kB Myd88- Bhattacharyya et al.19
fibroblasts glioblastoma cell lineb TLR4 blocking peptide, and TLR4 KO cells dependent
Human cardiac Full-length TN-C purified from human ↑ IL-6 and MMP-3 FBG anti-TLR4 Ab (for IL-6) Maqbool et al.20
myofibroblasts glioblastoma cell line, recombinant
human FBG domaina,b
Mouse macrophage cell line ↑ IL-6, TNF TLR4 inhibitor VIPER Benbow et al.21
Human hepatoma cell line ↑ epithelial-mesenchymal
transition
Primary human macrophages Recombinant human FBG domaina,b ↑ IL-6, IL-8, and TNF FBG anti-TLR4 Ab and TLR4 inhibitor Zuliani-Alvarez et al.22
TAK242
α9 Mouse synovial macrophages Recombinant FNIII3 domain (RGD to ↑ IL-6, IL-1α, IL-1β, TNF, CCL2, FNIII3 anti-α9 Ab and mutant FNIII3 Kanayama et al.23
RAA) CCL3, CCL4, CXCL2, and (removed α9 binding sequence) (IL-6
CXCL5 reduced)
Mouse synovial fibroblasts ↑ IL-6, IL-1α, CCL2, CCL4, anti-α9 Ab (IL-6 reduced)
CXCL5, CXCL12, and MMP-9
↓ MMP-2
Mouse dendritic cells Full-length TN-C purified from human ↑ IL-6, IL-23, Th17 FNIII3 anti-α9 Ab and mutant FNIII3 NFκB, MEK/ERK Kanayama et al.24
glioblastoma cell line, recombinant differentiation
mutant FNIII3 domain (RGD to RAA)
αVβ3 Mouse peritoneal ↑ IL-6, IL-1β, and TNF NFκB inhibitor (all cytokines), two NFκB Shimojo et al.25
macrophages different αVβ3 inhibitors and anti-β3
Ab (NFκB phosphorylation and IL-6
upregulation)

“TN-C source” column was left blank if no details about source and type of used tenascin-C were given in the article. Abbreviations: TN-C, tenascin-C; TLR4, toll-like receptor 4; HEK, human embryonic kidney; IL, interleukin;
TNF, tumor necrosis factor; FBG, fibrinogen-like globe; KO = gene knockout; NFκB, nuclear factor kappa-light-chain-enhancer of activated B cells; ADAMTS4, a disintegrin-like and metalloproteinase with thrombospondin
motifs-4; TAK242, small-molecule inhibitor of TLR4; MMP, matrix metalloproteinase; THP-1 = monocyte-like acute monocytic leukemia-derived cell line; oxLDL, oxidized low-density lipoprotein; DC, dendritic cell; JNK, c-Jun
N-terminal kinase; αSMA, alpha-smooth muscle actin; FNIII3, third fibronectin type III repeat; RAA, tripeptide Arg-Ala-Ala; CCL, C-C motif chemokine ligand; CXCL, chemokine receptor; MEK, mitogen-activated protein kinase
kinase; ERK, extracellular signal–regulated kinase.
a
Limulus amebocyte lysate (LAL) test has been used to measure endotoxin contamination in protein samples.
b
Preincubation with polymyxin B has been used as endotoxin contamination control.
291
292 Marzeda and Midwood Marzeda and Midwood

production and synthesis of inflammasome compo- phosphorylation, resulting in macrophage behavior


nents; this is followed by inflammasome assembly and more suited to tissue repair.18 Tenascin-C has also been
caspase-1-mediated maturation and release of bioac- shown to help drive macrophages to become foam
tive IL-1β and IL-18.28 Stimulation of rat epicardium– cells. Tenascin-C potentiated oxidized low-density lipo-
derived cells with tenascin-C induced TLR4-dependent protein (oxLDL)–induced foam cell formation, in a
pro-IL-1β transcription, implicating elevated tenascin- TLR4-dependent manner. Tenascin-C was also upregu-
C expression in rat cardiac tissue following ischemic lated upon macrophage stimulation by oxLDL, and was
injury as a potential priming step for inflammasome capable of inducing the expression of CD36, a scaven-
assembly that can be driven by adenosine or ATP- ger receptor involved in LDL uptake, suggesting the
mediated purinergic stimulation in this cell type.29 In existence of a positive feedback loop promoting the
addition, increased levels of secreted mature IL-1β generation of these cells key to the development of ath-
could be detected in cell culture medium of mouse erosclerosis.16 These data collectively show that tenas-
bone marrow–derived DCs stimulated with tenascin-C,17 cin-C profoundly influences macrophage polarization,
indicating that tenascin-C may be capable of driving that it induces a macrophage phenotype quite different
IL-1 synthesis in the absence of an exogenous second to that induced by pathogenic activation of TLR4, and
signal. However, a direct link between tenascin-C acti- that macrophage subsets created by tenascin-C activa-
vation of TLR4 and purinergic signaling, or the require- tion can differ depending on context.
ment for any other second signal, has not been An increasingly wide range of cell types are reported
investigated. to respond to tenascin-C via activation of TLR4. In
Tenascin-C upregulation therefore would be murine bone marrow isolated neutrophils, tenascin-C
expected to increase tissue levels of proinflammatory has been shown to upregulate the expression of matrix
cytokines and chemokines, orchestrating innate metalloproteinase (MMP)-9 in a TLR4-dependent
immune cell recruitment and activation. In addition, a manner.15 Activation of the TLR4/tenascin-C axis has
number of reports suggest that tenascin-C-mediated been also reported to induce epithelial-mesenchymal
cytokine release is also important for creating a micro- transition and promote migration of human hepatoma
environment permissive for selective T-cell polariza- cell line.21 Tenascin-C also induces TLR4-mediated
tion. DCs derived from tenascin-C-deficient mice cytokine synthesis in fibroblasts. For example, in syno-
exhibited diminished proinflammatory cytokine release vial fibroblasts it promotes high levels of IL-6 secretion,
in response to LPS stimulation, and a reduced capac- although it does not induce IL-8 in these cells, in con-
ity to drive naive T cells toward Th17, but not Th1, Th2, trast to macrophages.13 Moreover, other outputs have
or Treg, cell differentiation.30 Stimulation of DCs with been reported in tenascin-C-activated stromal cells.
exogenous tenascin-C promoted their ability to drive TLR4-dependent activation of primary human foreskin
the generation of Th17 cells, which effect could be fibroblasts by tenascin-C induces synthesis of type I
ablated with anti-IL-6 and anti-TLR4 antibodies, sug- collagen and alpha-smooth muscle actin.19 These data
gesting that IL-6 synthesis induced by tenascin-C acti- support a role for tenascin-C not just in triggering
vation of TLR4 is a key determinant of T-cell subset immune cell–mediated inflammation but also in stro-
polarization.17 Interestingly, tenascin-C also stimulated mally driven defense responses, suggesting that
chondrocyte secretion of prostaglandin E2,14 a com- this matrix molecule contributes to both inflammatory
plex mediator of inflammation known to be involved in processes immediately following tissue damage and
Th17 polarization.31,32 subsequent tissue remodeling by activating cell-type-
Wider profiling of the impact of tenascin-C activation specific responses via TLR4.
of TLR4 in primary human macrophages highlighted That tenascin-C drives very different TLR4-mediated
functional consequences beyond cytokine synthesis. inflammatory signaling than LPS supports the idea
Comparison of macrophages activated by tenascin-C, that the immune system is able to distinguish different
or by a pathogenic ligand, LPS, revealed overlapping, alarm signals using common sensors, to activate dis-
but distinct downstream impacts on cell behavior. While tinct, tailored responses to particular threats, that is,
both ligands induce classical NFκB and mitogen-acti- injury versus infection. However, it is not yet clear on
vated protein kinase (MAPK) signaling, LPS drove mac- the molecular level how different outputs arise from
rophages toward an aggressive phenotype designed to activation of the same PRR. We still do not fully under-
fight infection, characterized by high levels of proin- stand how tenascin-C binds to and activates TLR4.
flammatory cytokines and tissue degrading matrix What we do know is that the binding site comprises
metalloproteinases, while tenascin-C favored the upreg- three distinct epitopes within the FBG domain of
ulation of extracellular matrix protein expression and tenascin-C,22 and that the classical LPS coreceptors,
Tenascin-C and Innate Immunity 293

myeloid differentiation protein-2 (MD-2) and CD14, are modulation of the inflammatory response. Furthermore,
not essential for TLR4 activation by tenascin-C.13 evidence is emerging that supports a role for tenascin-
In addition to TLR4, tenascin-C activation of two C in PRR-mediated responses to pathogen invasion.
other cell surface receptors has been shown to drive In murine bone marrow–derived macrophages, rapid
inflammatory responses. Integrins α9β1 and αVβ3 are upregulation of tenascin-C upon LPS detection by
transmembrane receptors capable of binding to a TLR4 controls the maturation of the micro-RNA miR-
number of matrix proteins, including fibronectin and 155. Impaired maturation of miR-155 in the absence of
tenascin-C. The binding of tenascin-C by both integ- tenascin-C prevents effective translation of LPS-
rins induces proinflammatory cytokine synthesis, with induced proinflammatory cytokines including TNF.38
the interaction site responsible for the observed Finally, tenascin-C is capable of interfering with HIV
effects pinpointed to the third FNIII repeat for α9β1.23 transmission. Tenascin-C present naturally in human
Although αVβ3 is known to bind to two different sites breast milk was shown to bind HIV-1 Envelope protein,
within tenascin-C, the third FNIII repeat33 and the neutralizing the infectious properties of this virus in
FBG domain,34 it is still unknown which of them, if any, reporter cell lines and peripheral blood mononuclear
is responsible for conferring the proinflammatory cells.39 This cooperative impact of microbiological and
activity. In murine synovial macrophages and fibro- endogenous alarm signals demonstrates an amazing
blasts, tenascin-C-induced α9 integrin–mediated sig- complexity of signaling within the innate immune
naling upregulates a wide array of proinflammatory system.
molecules, including IL-6, IL-1α, CCL2, CCL4, and
CXCL5.23 α9 integrin activation by tenascin-C is also Tenascin-C and the Resolution of
involved in Th17 cell differentiation through induction
of IL-6 and IL-23 in DCs. In this study, stimulation of
Inflammation
IL-6 secretion by tenascin-C was shown to be a syner- The induction of tenascin-C expression is a transient
gistic effect of both α9 integrin- and TLR4-mediated process during the progression of inflammation; it is
signaling.24 The outcome of αVβ3 activation by tenas- not detectable following the completion of tissue repair.
cin-C was studied in peritoneal macrophages, where However, persistent tenascin-C expression is associ-
it resulted in the upregulation of IL-6, IL-1β, and TNF. ated with chronic inflammation, where the proinflam-
This effect of tenascin-C on cytokine secretion could matory properties of tenascin-C mediated by
be ablated with an NFκB inhibitor.25 These data show interactions with both TLR4 and the integrins might
that tenascin-C can induce cytokine synthesis by acti- offer an interesting insight into the mechanism of its
vating more than one receptor family; how this inter- involvement in inflammatory diseases. Upregulated as
play is mediated on the cellular level remains to be a result of tissue damage, cellular stress, and inflam-
elucidated, but such receptor redundancy may illus- matory mediators, and subsequently exacerbating the
trate the importance of tenascin-C as a danger signal, inflammatory response, tenascin-C is likely to activate
ensuring that it does not go unnoticed at times of a positive feedback loop driving disease. At this point,
cellular stress. the vital importance of strict control over tenascin-C
In addition to its influence through binding to mem- expression becomes evident, and the factors regulat-
brane receptors, tenascin-C may also indirectly modu- ing tissue levels of tenascin-C have recently been
late intracellular inflammatory signaling. In a human reviewed.11,12,40
glioblastoma cell line, tenascin-C downregulates the The question arises whether tenascin-C plays an
Wnt inhibitor DKK1, resulting in increased classical active role during the resolution of inflammation, or
Wnt signaling by stabilization of β-catenin.35 Tenascin-C whether its disappearance from tissues simply creates
also interacts with Wnt3a, and human adenocarci- a less proinflammatory environment. Tenascin-C has
noma cell line cells plated on tenascin-C in the pres- been shown to bind to TGF-β,41 a factor that plays a
ence of Wnt3a exhibit enhanced classical Wnt key role in the resolution of inflammation,42 and inter-
signaling. Interestingly, when cells were incubated with actions with tenascin-C might increase local concen-
soluble tenascin-C, the signaling was markedly trations or activity of this immunosuppressive factor.
reduced.36 This might suggest that while soluble Moreover, tenascin-C has been shown itself to exert
tenascin-C would sequester Wnt ligands, within the immunosuppressive effects on lymphocytes. A number
ECM it could be responsible for accumulating ligands of studies found tenascin-C to be capable of arresting
close to the cell surface enabling signaling activation. T-cell activation induced by various stimuli.43–47 The
Due to the fact that a complex crosstalk exists between blocking of anti-CD3 MAb/fibronectin-induced activa-
Wnt and NFκB pathways,37 regulation of Wnt signaling tion of human peripheral blood T cells by tenascin-C
by tenascin-C could exert both positive and negative was shown to be mediated through the alternatively
294 Marzeda and Midwood Marzeda and Midwood

spliced repeats A1 and A2 within FNIII domain and surgical subarachnoid hemorrhage model.63,64 Remarkable
was caused by a blockade of TCR/CD3 complex inter- anti-inflammatory effects of tenascin-C deficiency
nalization.46 In addition, in murine CD4+ and CD8+ were also reported in models of joint inflammation.
splenocytes activated with a cocktail of IL-2 and anti- During acute inflammation induced by zymosan, the
CD3 and anti-CD28 beads, tenascin-C-mediated reor- resolution of synovial inflammation was significantly
ganization of the actin cytoskeleton was implicated in quicker in TNC KO animals compared with wild type.
regulating T-cell behavior.47 On the whole, these data Furthermore, tenascin-C-deficient mice were pro-
add to the evidence supporting tenascin-C as a tected from prolonged synovitis, and cartilage and
sophisticated regulator of the inflammation, capable of bone destruction, in a model of erosive arthritis
conveying both pro- and anti-inflammatory responses. induced by methylated BSA.13 During this erosive
arthritis, tenascin-C deficiency resulted in decreased
The Wider Picture: Conclusions From cytokine synthesis in the joint, including attenuated
IL-6 and IL-23 expression, and this altered cytokine
Animal Models
profile created a microenvironment that could not sup-
The intricate inflammatory functions of tenascin-C port Th17 polarization, manifesting in significantly
described in vitro prompt questions about the signifi- reduced IL-17 expression.30 TNC KO mice also exhib-
cance of its contribution to inflammation in vivo. The ited a decrease in the production of IL-17 by Th17 cells
macroscopic effects of tenascin-C gene deletion have during experimental autoimmune encephalomyelitis, a
been examined using two independently generated mouse model of multiple sclerosis, accompanied by a
knockout mice.48,49 Surprisingly, few gross phenotypic reduction in the number of interferon gamma (IFNγ)–
differences could be detected in the tenascin-C knock- producing Th1 cells.60 Moreover, during ovalbumin
out (TNC KO) animals, compared with wild type. They (OVA)-induced bronchial asthma, TNC KO animals
were born alive and fertile, normal in size, and exhib- exhibited significantly attenuated allergic inflammation,
ited standard development and lifespan. Abnormalities with reduced eosinophil infiltration and reduced levels
in TNC KO mice were observed during tissue repair of IgE levels. This dampening of the inflammatory
following corneal50,51 and myocardial injury,52 including response was shown to be a result of impaired differ-
reduced macrophage infiltration, delayed myofibro- entiation of Th2 cells, leading to a decrease in the
blast recruitment, and reduced TGF-β, fibronectin, and expression of the classical type 2 cytokines, IL-5 and
collagen synthesis, resulting in delayed healing. These IL-13.54 These studies support in vitro data suggesting
phenotypes stimulated further interest in the protein’s that tenascin-C can modulate T cell polarization, but
relevance during pathological conditions. Indeed, highlight that tenascin-C exerts complex and highly
investigation of inflammatory responses in TNC KO context-specific effects, affecting T-cell subsets shown
mice challenged with experimental disease has pro- to orchestrate particular pathologies: Th2 in asthma,68
vided considerable insight into the role of tenascin-C in Th17 in rheumatoid arthritis (RA),69 and Th17 and Th1
immunity in vivo (summarized in Table 2). in autoimmune encephalomyelitis.70
In line with the proinflammatory capabilities of In addition to exacerbating inflammatory responses,
tenascin-C reported in vitro, tenascin-C persistently tenascin-C has been shown to play a particularly dele-
emerges as a key player in chronic inflammation in terious role promoting tissue fibrosis. For example, in
vivo. The reduction of proinflammatory cytokine secre- the cardiovascular system, tenascin-C deficiency
tion and subsequent diminished inflammatory cell relieves postsurgical neointimal hyperplasia62,71 and
recruitment as a result of tenascin-C deficiency have improves cardiac function after myocardial infarction.61
been shown to exert a protective effect in a number of Moreover, in a concanavalin A–induced hepatitis
inflammatory disease models. Tenascin-C ablation in model, a reduction in collagen expression and deposi-
transgenic mice overexpressing mutated amyloid pre- tion was observed in the livers of TNC KO mice. This
cursor protein resulted in suppression of Alzheimer’s was associated with reduced secretion of inflammatory
disease–specific inflammation. Lower levels of proin- mediators IFNγ, TNF, and IL-4, and lower lymphocyte
flammatory mediators, such as TNF, IL-1β, and IL-6, and neutrophil infiltration into the liver, as well as a sig-
and higher levels of anti-inflammatory mediators, such nificant downregulation of TGF-β expression compared
as IL-10, MRC1, and CCL2, in the central nervous sys- with wild-type mice.58 Impaired TGF-β signaling, with
tem were associated with reduced amyloid deposition diminished nuclear translocation of Smad-2 and Smad-
and ameliorated Alzheimer’s disease pathology.53 3, was also shown to protect tenascin-C-deficient ani-
Knockout of tenascin-C was reported to reduce mals from lung fibrosis in a bleomycin-induced acute
immune cell infiltration and prevent brain edema and lung injury model.59 Moreover, a decrease in TGF-β
blood–brain barrier disruption following insult in a expression and attenuation of collagen deposition
Tenascin-C and Innate Immunity 295

Table 2. Tenascin-C in Mouse Models of Inflammatory Diseases.


Pathology Model Tenascin-C Deficiency Effect Ref.

Tenascin-C deficiency alleviating inflammatory response


Alzheimer’s disease Mutated APP-overexpression ↓ proinflammatory cytokine synthesis and amyloid deposits Xie et al.53
AD model in the brain
Asthma Ovalbumin-induced bronchial ↓ inflammatory cell infiltration, goblet cell production, Nakahara et al.54
asthma model IL-5, IL-13, MCP-1, and MMP-9 expression, levels of IgE,
airway hyperresponsiveness
Cancer Transgenic metastatic breast Altered tumor organization Talts et al.55
cancer model ↑ macrophage infiltration
Xenografting melanoma model ↓ tumor size and vascularization, VEGF expression Tanaka et al.56
Transgenic pancreatic cancer ↓ tumor vascularization, vessel leakage, level of tumor Saupe et al.57
model cell–specific marker insulin mRNA
↑ levels of Wnt pathway inhibitor DKK1
Hepatic ischemia/ Warm hepatic IRI model ↑ liver regeneration, ↓ necrosis, levels of apoptotic Kuriyama et al.15
reperfusion injury markers, IL-1β, IL-6, CXCL2, and MMP-9 expression,
impaired leukocyte recruitment, altered expression of
adhesion molecules
Liver fibrosis Concanavalin A–induced ↓ collagen expression and deposition, inflammatory cell El-Karef et al.58
hepatitis model infiltration, IFNγ, TNF, IL-4, and TGF-β production,
numbers of αSMA positive cells
Lung fibrosis Bleomycin-induced acute lung ↓ collagen deposition, numbers of αSMA and Smad2/3- Carey et al.59
injury model positive fibroblasts, fibroblast–myofibroblast
transformation
Multiple sclerosis MOG-induced ↓ number of IFNγ producing Th1 cells and IL-17 Momcilovic et al.60
experimental autoimmune production by Th17 cells
encephalomyelitis model
Myocardial infarction Surgical MI model ↓ collagen deposition, myocardial stiffness Nishioka et al.61
↑ cardiac function
Neointimal hyperplasia Surgical aortomy model ↓ aorta scaring, cell migration and proliferation, deposition Yamamoto et al.62
of proteoglycans
Rheumatoid arthritis Zymosan-induced acute ↑ resolution of paw swelling, no synovitis, no cell Midwood et al.13
arthritis model infiltration
mBSA-induced erosive ↑ synovial inflammation resolution, no fibrin or cell
arthritis model aggregation, no pannus formation, no proteoglycan loss,
no chondrocyte death, no cartilage or bone erosion
Subarachnoid Surgical SAH model ↓ brain edema, blood-brain barrier disruption Fujimoto et al.63
hemorrhage ↓ cerebral vasospasm and inflammatory cell infiltration Fujimoto et al.64
Systemic sclerosis Bleomycin-induced pulmonary ↓ fibrosis scores, pleural thickening, numbers of fibrotic Bhattacharyya
fibrosis model foci, inflammatory cell infiltration, collagen deposition, et al.19
numbers of αSMA- and Smad2-positive fibroblasts
Bleomycin-induced dermal ↑ resolution of fibrotic dermis thickening
fibrosis model ↓ inflammatory cell infiltration, collagen, fibronectin, IL-6,
MCP-1, and TGF-β expression, Smad-2/3 activation
Tsk dermal fibrosis model ↓ hypodermis thickening, collagen and αSMA deposition
Tenascin-C deficiency disturbing tissue remodeling
Dermatitis chemically induced ear ↑ ear thickening, disorganized ECM, prolonged granulocyte Koyama et al.65
dermatitis model infiltration, formation of elastosis-like structures
Glomerulonephritis Habu-snake venom Abnormal regeneration, ↑ proteinuria and scar tissue Nakao et al.66
induced proliferative formation, ↓ mesangial cells proliferation, PDGF-BB
glomerulonephritis model expression, abnormal TGF-β, collagen and fibronectin
expression
Osteoarthritis spontaneous age-related knee ↑ articular cartilage degeneration Okamura et al.67
OA model
surgical degenerative knee OA Delayed cartilage regeneration
model

Abbreviations: APP, β-amyloid precursor protein; AD, Alzheimer’s disease; IL, interleukin; MCP-1, monocyte chemotactic protein 1; MMP, matrix metalloproteinase; VEGF,
vascular endothelial growth factor; DKK1, Dickkopf-1; IRI, ischemia/reperfusion injury; CXCL2, C-X-C motif chemokine ligand 2; IFNγ, interferon gamma; TNF, tumor
necrosis factor; TGF-β, transforming growth factor beta; αSMA, alpha-smooth muscle actin; MOG, myelin oligodendrocyte glycoprotein; MI, myocardial infarction; mBSA,
methylated BSA; SAH, subarachnoid hemorrhage; ECM, extracellular matrix; PDGF-BB, platelet-derived growth factor chain B dimer; OA, osteoarthritis.
296 Marzeda and Midwood Marzeda and Midwood

caused reduced dermal and pleural thickening, as well However, these data also highlight that tenascin-C dis-
as accelerated resolution of inflammation, in the plays a remarkable functional dichotomy; in some
absence of tenascin-C in bleomycin-induced mouse models, it positively influences tissue repair, protecting
models of systemic sclerosis.19 Consistent with in vitro from excessive inflammation and remodeling, and pre-
studies, these data indicate that tenascin-C impacts venting fibrotic tissue formation. In contrast, in other
both the immune and the stromal compartment during models, it is capable of driving chronic inflammation
inflammatory disease models, and suggest that and promoting profibrotic responses often to the detri-
enhanced fibrosis may be mediated at least in part by ment of the organism. Temporal control of tissue levels
raising tissue levels of TGF-β, enabling this factor to of tenascin-C might be of importance here; where
exert fibrogenic effects by promoting fibroblast activa- expression is downregulated tenascin-C mediated
tion, and stimulating excessive production of extracel- inflammation and repair can proceed in a controlled
lular matrix.72 Finally, liver tissue regeneration was manner, while persistent expression of tenascin-C
enhanced as a result of tenascin-C ablation in a model would fuel proinflammatory and profibrotic responses.
of hepatic ischemia/reperfusion injury. In the absence Another key influencing factor might be the tissue
of tenascin-C, the secretion of IL-6, IL-1β, and CXCL2 microenvironment where expression of distinct tenas-
was decreased and the recruitment of leukocytes cin-C splicing variants, differential proteolytic process-
impaired. This inflammatory phenotype was accompa- ing, and the presence of different tenascin-C receptors
nied by attenuated catabolic processes, with depleted exposed on the cell surface would also be expected to
necrosis, lower levels of apoptotic markers, and shape the context of tenascin-C-mediated responses.
decreased expression and activation of MMP-9, an In considering the role of tenascin-C in inflamma-
extracellular matrix–degrading enzyme,15 suggesting tion in vivo, more intriguing questions continue to
tenascin-C can impact tissue fibrosis by both promot- emerge. Tenascin-C has been shown to contribute to
ing matrix deposition and inhibiting matrix removal. the immune response to LPS-induced sepsis. TNC KO
These data indicate that tenascin-C expression in animals were protected from systemic inflammation
experimental models contributes to prolonged inflam- following the cytokine storm caused by LPS adminis-
mation and fibrosis during disease pathogenesis. tration, supporting a role for tenascin-C in host defense
However, a number of studies provide evidence sup- against infection in vivo. Interestingly, this phenotype in
porting a positive role for tenascin-C during inflamma- tenascin-C-deficient mice could be partially rescued
tion in vivo. Following renal injury in a snake by bone marrow transplant from wild-type littermates,38
venom–induced model of proliferative glomerulone- highlighting that both myeloid and stromally derived
phritis, an inflammatory kidney disease, TNC KO ani- tenascin-C participate in these immune responses.
mals showed drastically impaired renal regeneration, Within the bone marrow compartment, a decrease in
with increased proteinuria and scar tissue formation, cell colony–forming capacity has been reported in
decreased mesangial cells proliferation, and aberrant TNC KO animals, yet the steady-state hematopoiesis
patterns of TGF-β, collagen, and fibronectin expres- was not affected.73 However, tenascin-C-deficient
sion. The severity of observed abnormalities resulted mice did exhibit impaired hematopoietic recovery as a
in death of all knockout animals within 4 months, result of a bone marrow ablation by irradiation.74
despite the injury being reversible in wild-type controls.66 Tenascin-C was also shown to regulate bone marrow–
Tenascin-C has been also reported to attenuate inflam- mediated angiogenesis.75 In addition, a recent study
mation in chemically induced dermatitis. Tenascin-C- found that tenascin-C expressed in the bone marrow,
null mice exhibited increased swelling and prolonged but not the myocardium, was responsible for the atten-
polymorphonuclear cell infiltration. During postinsult uation of fibrosis in a mouse model of cardiac hyper-
regeneration, a disorganization of extracellular matrix trophy. TNC KO animals showed an exacerbation of
and formation of elastosis-like structures were inflammation and impaired cardiac function following
observed.65 A significant delay in tissue regeneration the transverse aortic constriction surgery. This pheno-
was also observed in tenascin-C-deficient mice in a type could be rescued by a bone marrow transplant
surgical degenerative knee osteoarthritis model. In from a wild-type donor, and simultaneously, a trans-
contrast to controls, the injured cartilage could not be plant from a tenascin-C-null donor to a wild-type recip-
regenerated in TNC KO animals, and the defects were ient resulted in a declined cardiac function.76 Together,
filled with fibrous tissue. Moreover, age-related sponta- these data pose questions about the site of action of
neous articular cartilage degeneration was acceler- tenascin-C during inflammation, suggesting a role not
ated in knockout animals.67 only locally, at the site of inflammation, but also distally,
Collectively, these studies confirm tenascin-C is a impacting inflammatory cell behavior at sites some
potent regulator of inflammation and fibrosis in vivo. distance from pathology. These data also raise a
Tenascin-C and Innate Immunity 297

cautionary note that it will be important to take into “flare”) than conventionally used biomarkers, poten-
account the background strain of mice used in studies, tially allowing for quicker identification of the need to
as different strains of mice are known to exhibit differ- escalate immunosuppressive therapy. The diagnostic
ent immune compartment biases that could define the value of tenascin-C was also investigated in arthritis,
ultimate impact of tenascin-C deletion in vivo. revealing upregulation in the synovial fluid of osteoar-
thritis patients, with levels correlating to disease severity.86
Tenascin-C in the Clinic Similarly, tenascin-C was elevated in RA, although
here tenascin-C did not correlate with the levels of
In humans, a missense mutation in the TNC gene is
CRP in synovial fluid.87 Upon further investigation,
responsible for autosomal dominant hear loss, caused
markedly raised circulating levels of tenascin-C in RA
by irreversible abnormalities in developing cochlea.77
patients correlate with joint erosion.88 Moreover, in RA,
Polymorphisms in the gene have been also associated
a subset of autoantibodies recognizing a citrullinated
with increased risk of asthma, rhinoconjunctivitis, and
epitope in the FBG domain of tenascin-C could be
Achilles tendinopathy.78–80 However, it is the wild-type
detected years before any manifestation of the dis-
protein that attracts most interest as a potential target
ease and thus could help in early clinical diagnosis,89
for diagnostics and therapy.
as well as predicting among people presenting with
Successful treatment of a disease must be pre-
early synovitis, those whose inflammation will not
ceded by a correct diagnosis. This is often challenging,
resolve but who will go on to develop RA.90 Tenascin-C
as signs and symptoms can be nonspecific, or their
has been also considered an attractive candidate bio-
severity highly subjective. Hence the widespread use
marker in cardiac pathologies (reviewed in Franz et al.91).
of biomarkers, easily measurable objective factors
For example, tenascin-C has been proven a useful
indicating the medical state of the patient. Biomarkers
biomarker in a retrospective study on Kawasaki dis-
can be employed to predict disease incidence and
ease. Serum levels of tenascin-C were shown to pre-
severity, treatment outcome, or surgical intervention
dict the risk of acquiring resistance to the high-dose
effects. Although tenascin-C is upregulated in a num-
intravenous immunoglobulin therapy, implemented in
ber of human pathologies, it has been most thoroughly
Kawasaki disease to reduce pathological chronic
studied in the context of cancer, and was shown largely
inflammation, and also subsequent development of
to correlate with metastasis and indicate poor progno-
coronary artery lesions, one of the most dangerous
sis. So far its prognostic significance has been con-
complications of the acute form of the disease.92
firmed in breast, lung, and bladder cancer, glioma,
Moreover, in line with in vitro data highlighting the pro-
squamous cell carcinoma, melanoma, and colorectal
fibrotic activity of tenascin-C, this matrix molecule
and hepatocellular carcinoma (reviewed in Lowy and
could also be useful in the stratification of patients with
Oskarsson.81). Recently, tenascin-C levels have been
conditions caused by pathological cardiac tissue
investigated as a diagnostic factor in preeclampsia, a
remodeling. High tenascin-C levels in patients suffer-
hypertensive disorder of pregnancy and one of the
ing from hypertrophic cardiomyopathy correlated with
most common cause of maternal deaths worldwide.82
high incidence of heart failure, a potential cause of
Consistent with its involvement in driving inflamma-
premature death that is hard to predict with echocar-
tion, tenascin-C has also been widely considered as a
diographic examination.93 In addition, in systemic scle-
biomarker of patient inflammatory status in a number
rosis, a disease driven by persistent fibrosis,
of chronic inflammatory diseases.
significantly elevated tenascin-C expression was found
A screen of tenascin-C in sera from patients suffer-
in skin biopsies from patients with inflammatory subset
ing diverse conditions, including cancer, malaria, and
of the disease. What is more, tenascin-C levels were
sepsis (from a routine chemistry laboratory of a large
also markedly higher in circulation of patients with
hospital), found a positive correlation between the lev-
both early- and late-stage disease.19 Together, these
els of tenascin-C and C-reactive protein (CRP), high-
data highlight tenascin-C as a prospective diagnostic
lighting that elevated serum tenascin-C is associated
target in a number of inflammatory and fibrotic condi-
with pathological inflammation, but is not disease spe-
tions, useful in predicting disease progression and
cific.83 Subsequent studies have shown that circulat-
determining patient response to treatment.
ing tenascin-C levels correlate with disease activity in
inflammatory conditions including ulcerative colitis, Tenascin-C is an immunomodulatory protein that
Crohn’s disease, and systemic lupus erythematosus plays a key role in physiological tissue repair, but
(SLE).84,85 The clinical relevance of tenascin-C was which can also drive pathological inflammation and
particularly evident in SLE, as it was found to be a better fibrosis (Fig. 1). While an increasing body of data sup-
predictive factor of disease aggravation (so-called port a role for tenascin-C in influencing inflammation,
298 Marzeda and Midwood Marzeda and Midwood

Figure 1. Tenascin-C modulates physiological and pathological inflammation. (A) Tenascin-C, upregulated upon cellular stress or tissue
injury, is capable of binding to TLR4 and integrins α9β1 and αVβ3 on the cell surface and inducing NFκB-mediated gene expression. In
macrophages and DCs, tenascin-C stimulates the secretion of a wide array of proinflammatory cytokines and chemokines, provoking
an inflammatory response. Among these soluble mediators, IL-1β, IL-6, and IL-23 promote the differentiation of Th17 cells. Tenascin-C
also polarizes myeloid cell phenotypes in a context-specific manner, for example, generating reparative macrophages or facilitating lipid
uptake, promoting macrophages to form foam cells. In fibroblasts, tenascin-C stimulates expression of a different subset of mediators;
this includes the cytokine IL-6 but also molecules such as αSMA, TGF-β and high levels of collagen that promote tissue remodeling.
Tenascin-C is also capable of inhibiting naive T-cell activation. Typically, tenascin-C expression is downregulated, allowing for the resolu-
tion of inflammation and completion of tissue repair. However, a number of questions regarding the mode of action of tenascin-C still
need answering: (1) What are the exact mechanisms leading to tenascin-C upregulation? (2) Does the form of tenascin-C (hexabrachion/
monomer/domains cleaved by proteolytic enzymes) influence receptor binding and activation? (3) How exactly, and why, is tenascin-C
expression downregulated and protein cleared from the tissue? Answering each of these questions will be crucial to understanding how
and why persistent expression of tenascin-C leads to a chronic inflammation that can trigger the development of a number of inflamma-
tory diseases, summarized in (B). Abbreviations: TLR4, toll-like receptor 4; NFκB, nuclear factor kappa-light-chain-enhancer of activated
B cells; DC, dendritic cell; IL = interleukin; αSMA, alpha-smooth muscle actin; TGF-β, transforming growth factor beta; TNF, tumor
necrosis factor; MMP, matrix metalloproteinase; CCL, C-C motif chemokine ligand; CXCL, chemokine receptor; oxLDL, oxidized low-
density lipoprotein.

this field is still in its infancy. Although tenascin-C immune populations to stromal cell lineages, and the
impacts immune signaling in a wide range of cell molecular basis of the cell-type-specific responses
types, the details of these signaling pathways remain reported so far to be dissected. Furthermore, there
to be fully defined in each target cell, from distinct may be as yet uncovered roles for tenascin-C in
Tenascin-C and Innate Immunity 299

impacting other effector cells that may provide a fuller profound effect in reported cases of acute tissue repair,
picture of how this matrix molecule orchestrates suggesting an association with immune persistence
inflammation. For example, myeloid-derived suppres- that may provide an avenue to treat chronic diseases
sor cells (MDSCs), a heterogeneous population of without significantly affecting underlying biology.
immature myeloid cells capable of inhibiting T cell– Alternatively, elucidating mechanisms that control the
mediated inflammation, are emerging as key players induction of tenascin-C expression at the transcrip-
in immune responses underlying tissue repair94 as tional level, or tissue removal at the protein level, would
well as cancer95 and autoimmune diseases such as provide information that might enable manipulation in
RA, inflammatory bowel disease, or SLE.96 Due to a tissue- or disease-specific manner, adding a further
their unique immune suppressive potential, MDSCs level of refinement to this approach.
are emerging as potential cellular immunotherapeutic The second approach would involve targeting
agents.97 The expansion of MDSCs can be stimulated tenascin-C signaling by specifically preventing its acti-
by activation of the TLR4 pathway,98 for example, in vation of receptors that mediate undesirable inflamma-
the lungs where MDSCs induced by activation of tory signaling. However, this strategy also comes with
TLR4 signaling by LPS inhibit Th2 effector functions.99 its own challenges. Little is known about how different
In a mammary carcinoma mouse model, S100A8/A9 domains in tenascin-C interact with each other at the
proteins, endogenous TLR4 activators, have been functional level; for example the extent of synergy
shown to induce MDSCs accumulation in tissue, between domain signaling is unclear, nor do we know
albeit this was triggered by binding to glycoprotein how individual interactions are influenced by the struc-
receptors on the cell surface.100 These data raise the tural context of the molecule as a whole. We also have
intriguing possibility that MDSCs may respond to limited knowledge of the different ligand binding modes
tenascin-C and that this interaction could serve as a of this matrix molecule; this includes understanding
natural means of turning off tenascin-C-driven inflam- whether interactions are mediated as a result of ligand
mation. Whether this is the case, and whether this binding exclusively to a unique epitope within tenas-
mechanism goes awry in chronic or pathological cin-C, or to more general motifs with multiple potential
inflammation, remains to be seen. ligands, or whether a combination of both possibilities
Due to its unique properties and characteristic exists. For example, RGD sequences bind to more
expression pattern, tenascin-C has long been regarded than one combination of integrin dimers, and integrins
as an attractive therapeutic candidate for targeting in can also bind to non-RGD containing motifs in tenas-
clinics. A substantial part of ongoing work focuses on cin-C. In addition, one site among the tripartite TLR4-
utilizing tenascin-C as a “postcode” for delivery of ther- binding epitope within tenascin-C comprises an
apeutics directly to the tumor microenvironment with exposed cationic ridge,22 which might conceivably
the means of aptamer or antibody technologies interact with other charged molecules. Understanding
(reviewed in Spenle et al.101). However, we are yet to more about how this multimodular molecule binds to
explore applications targeting the proinflammatory its wide range of ligands therefore is key to under-
potential of tenascin-C. To regain control over detri- standing not only its biological complexity but also
mental inflammation fueled by tenascin-C, two main inhibitor development.
strategies could be implemented. The first approach
might involve regulation of tenascin-C production at Competing Interests
the level of mRNA transcription and protein expres- KSM has filed patents around the use of tenascin-C in dis-
sion. This would in principle resolve the key issue ease diagnosis and treatment, and is the founder of, and
underlying tenascin-C pathology—its misregulated consultant to, Nascient Ltd.
expression. Global inhibition of tenascin-C expression,
however, would ablate all the effects exerted by this Author Contributions
large multimodular molecule in all tissues, which would AMM and KSM contributed to the conception and prepara-
include undesirable, prolonged engagement of proin- tion of the manuscript, and wrote the manuscript. Both
flammatory signaling, but which also might prevent authors reviewed and approved the final version of the
desirable functions, for example, those supporting manuscript.
physiological tissue remodeling. The restricted pattern
of prolonged expression of tenascin-C in the adult to Funding
pathological sites may make this a viable option. The author(s) disclosed receipt of the following financial
Moreover, the phenotype of the tenascin-C knockout support for the research, authorship, and/or publication of
mouse implies a level of redundancy for this molecule this article: This work was supported by Arthritis Research
during development, and its absence does not have a UK and the Kennedy Trust for Rheumatology Research.
300 Marzeda and Midwood Marzeda and Midwood

Literature Cited 14. Patel L, Sun WY, Glasson SS, Morris EA, Flannery
CR, Chockalingam PS. Tenascin-C induces inflamma-
1. Janeway CA, Medzhitov R. Innate immune rec-
tory mediators and matrix degradation in osteoarthritic
ognition. Annu Rev Immunol. 2002;20:197–216.
cartilage. BMC Musculoskelet Disord. 2011;12:14.
doi:10.1146/annurev.immunol.20.083001.084359.
doi:10.1186/1471-2474-12-164.
2. Bianchi ME. DAMPs, PAMPs and alarmins: all we need
15. Kuriyama N, Duarte S, Hamada T, Busuttil RW, Coito
to know about danger. J Leukoc Biol. 2007;81(1):1–5.
AJ. Tenascin-C: a novel mediator of hepatic ischemia
doi:10.1189/jlb.0306164.
and reperfusion injury. Hepatology. 2011;54(6):2125–
3. Schaefer L. Complexity of danger: the diverse nature
36. doi:10.1002/hep.24639.
of damage-associated molecular patterns. J Biol
16. Liu R, He Y, Li B, Liu J, Ren YG, Han W, Wang X, Zhang
Chem. 2014;289(51):35237–45. doi:10.1074/jbc.
L. Tenascin-C produced by oxidized LDL-stimulated
R114.619304.
macrophages increases foam cell formation through
4. Bryant CE, Gay NJ, Heymans S, Sacre S, Schaefer
Toll-like receptor-4. Mol Cells. 2012;34(1):35–41.
L, Midwood KS. Advances in Toll-like receptor biol-
doi:10.1007/s10059-012-0054-x.
ogy: modes of activation by diverse stimuli. Crit Rev 17. Machino-Ohtsuka T, Tajiri K, Kimura T, Sakai S,
Biochem Mol Biol. 2015;50(5):359–79. doi:10.3109/1 Sato A, Yoshida T, Hiroe M, Yasutomi Y, Aonuma
0409238.2015.1033511. K, Imanaka-Yoshida K. Tenascin-C aggravates
5. Dostert C, Petrilli V, Van Bruggen R, Steele C, autoimmune myocarditis via dendritic cell activa-
Mossman BT, Tschopp J. Innate immune activa- tion and Th17 cell differentiation. J Am Heart Assoc.
tion through Nalp3 inflammasome sensing of asbes- 2014;3(6):e001052. doi:10.1161/jaha.114.001052.
tos and silica. Science. 2008;320(5876):674–7. 18. Piccinini AM, Zuliani-Alvarez L, Lim JMP, Midwood KS.
doi:10.1126/science.1156995. Distinct microenvironmental cues stimulate divergent
6. Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer TLR4-mediated signaling pathways in macrophages.
G, Bauernfeind FG, Abela GS, Franchi L, Nuñez Sci Signal. 2016;9(443):ra86. doi:10.1126/scisignal.
G, Schnurr M, Espevik T, Lien E, Fitzgerald KA, aaf3596.
Rock KL, Moore KJ, Wright SD, Hornung V, Latz 19. Bhattacharyya S, Wang WX, Morales-Nebreda L, Feng
E. NLRP3 inflammasomes are required for ath- G, Wu MH, Zhou XD, Lafyatis R, Lee J, Hinchcliff M,
erogenesis and activated by cholesterol crystals. Feghali-Bostwick C, Lakota K, Budinger GR, Raparia
Nature. 2010;464(7293):1357–61. doi:10.1038/ K, Tamaki Z, Varga J. Tenascin-C drives persis-
nature08938. tence of organ fibrosis. Nat Commun. 2016;7:11703.
7. Halle A, Hornung V, Petzold GC, Stewart CR, Monks doi:10.1038/ncomms11703.
BG, Reinheckel T, Fitzgerald KA, Latz E, Moore KJ, 20. Maqbool A, Spary EJ, Manfield IW, Ruhmann M,
Golenbock DT. The NALP3 inflammasome is involved Zuliani-Alvarez L, Gamboa-Esteves FO, Porter KE,
in the innate immune response to amyloid-beta. Nat Drinkhill MJ, Midwood KS, Turner NA. Tenascin C
Immunol. 2008;9(8):857–65. doi:10.1038/ni.1636. upregulates interleukin-6 expression in human car-
8. Woo SR, Corrales L, Gajewski TF. Innate immune diac myofibroblasts via toll-like receptor 4. World
recognition of cancer. In: Littman DR, Yokoyama WM, J Cardiol. 2016;8(5):340–50. doi:10.4330/wjc.
editors. Annual review of immunology, vol. 33. Palo v8.i5.340.
Alto: Annual Reviews; 2015. p. 445–74. 21. Benbow JH, Thompson KJ, Cope HL, Brandon-
9. Mogensen TH. Pathogen recognition and inflam- Warner E, Culberson CR, Bossi KL, Li T, Russo MW,
matory signaling in innate immune defenses. Clin Gersin KS, McKillop IH, deLemos AS, Schrum LW.
Microbiol Rev. 2009;22(2):240–73. doi:10.1128/ Diet-induced obesity enhances progression of hepa-
cmr.00046-08. tocellular carcinoma through tenascin-C/Toll-like
10. Chiquet-Ehrismann R, Chiquet M. Tenascins: regu- receptor 4 signaling. Am J Pathol. 2016;186(1):145–
lation and putative functions during pathological 58. doi:10.1016/j.ajpath.2015.09.015.
stress. J Pathol. 2003;200(4):488–99. doi:10.1002/ 22. Zuliani-Alvarez L, Marzeda AM, Deligne C, Schwenzer
path.1415. A, McCann FE, Marsden BD, Piccinini AM, Midwood
11. Udalova IA, Ruhmann M, Thomson SJP, Midwood KS. Mapping tenascin-C interaction with toll-like
KS. Expression and immune function of tenascin-C. receptor 4 reveals a new subset of endogenous
Crit Rev Immunol. 2011;31(2):115–45. inflammatory triggers. Nat Commun. 2017;8(1):1595.
12. Midwood KS, Chiquet M, Tucker RP, Orend G. doi:10.1038/s41467-017-01718-7.
Tenascin-C at a glance. J Cell Sci. 2016;129(23):4321– 23. Kanayama M, Kurotaki D, Morimoto J, Asano T,
7. doi:10.1242/jcs.190546. Matsui Y, Nakayama Y, Saito Y, Ito K, Kimura C,
13. Midwood K, Sacre S, Piccinini AM, Inglis J, Trebaul Iwasaki N, Suzuki K, Harada T, Li HM, Uehara J,
A, Chan E, Drexler S, Sofat N, Kashiwagi M, Orend Miyazaki T, Minami A, Kon S, Uede T. Alpha(9) inte-
G, Brennan F, Foxwell B. Tenascin-C is an endoge- grin and its ligands constitute critical joint microenvi-
nous activator of Toll-like receptor 4 that is essential ronments for development of autoimmune arthritis. J
for maintaining inflammation in arthritic joint disease. Immunol. 2009;182(12):8015–25. doi:10.4049/jimmu-
Nat Med. 2009;15(7):774–80. doi:10.1038/nm.1987. nol.0900725.
Tenascin-C and Innate Immunity 301

24. Kanayama M, Morimoto J, Matsui Y, Ikesue M, 35. Ruiz C, Huang WT, Hegi ME, Lange K, Hamou MF,
Danzaki K, Kurotaki D, Ito K, Yoshida T, Uede T. Fluri E, Oakeley EJ, Chiquet-Ehrismann R, Orend G.
α9β1 integrin-mediated signaling serves as an intrin- Dilfferential gene expression analysis reveals activation
sic regulator of pathogenic Th17 cell generation. J of growth promoting signaling pathways by tenascin-C.
Immunol. 2011;187(11):5851–64. doi:10.4049/jimmu- Cancer Res. 2004;64(20):7377–85. doi:10.1158/0008-
nol.1101524. 5472.can-04-1234.
25. Shimojo N, Hashizume R, Kanayama K, Hara M, 36. Hendaoui S, Tucker RP, Zingg D, Bichet S, Schittny
Suzuki Y, Nishioka T, Hiroe M, Yoshida T, Imanaka- J, Chiquet-Ehrismann R. Tenascin-C is required for
Yoshida K. Tenascin-C may accelerate cardiac fibro- normal Wnt/beta-catenin signaling in the whisker
sis by activating macrophages via the integrin alpha follicle stem cell niche. Matrix Biol. 2014;40:46–53.
V beta 3/nuclear factor-kappa B/interleukin-6 axis. doi:10.1016/j.matbio.2014.08.017.
Hypertension. 2015;66(4):757–66. doi:10.1161/ 37. Ma B, Hottiger MO. Crosstalk between wnt/beta-
hypertensionaha.115.06004. catenin and NF-kappa B signaling pathway during
26. Yu L, Wang LT, Chen SW. Endogenous toll-like recep- inflammation. Front Immunol. 2016;7:14. doi:10.3389/
tor ligands and their biological significance. J Cell Mol fimmu.2016.00378.
Med. 2010;14(11):2592–603. doi:10.1111/j.1582- 38. Piccinini AM, Midwood KS. Endogenous control of
4934.2010.01127.x. immunity against infection: tenascin-C regulates
27. Broz P, Dixit VM. Inflammasomes: mechanism of TLR4-mediated inflammation via MicroRNA-155.
assembly, regulation and signalling. Nat Rev Immunol. Cell Rep. 2012;2(4):914–26. doi:10.1016/j.cel-
2016;16(7):407–20. doi:10.1038/nri.2016.58. rep.2012.09.005.
28. Jo EK, Kim JK, Shin DM, Sasakawa C. Molecular 39. Mansour RG, Stamper L, Jaeger F, McGuire E, Fouda
mechanisms regulating NLRP3 inflammasome G, Amos J, Barbas K, Ohashi T, Alam SM, Erickson H,
activation. Cell Mol Immunol. 2016;13(2):148–59. Permar SR. The presence and anti-HIV-1 function of
doi:10.1038/cmi.2015.95. tenascin C in breast milk and genital fluids. PLoS ONE.
29. Hesse J, Leberling S, Boden E, Friebe D, Schmidt T, 2016;11(5):e0155261. doi:10.1371/journal.pone.
Ding ZP, Dieterich P, Deussen A, Roderigo C, Rose 0155261.
CR, Floss DM, Scheller J, Schrader J. CD73-derived 40. Chiovaro F, Chiquet-Ehrismann R, Chiquet M.
adenosine and tenascin-C control cytokine produc- Transcriptional regulation of tenascin genes. Cell Adh
tion by epicardium-derived cells formed after myo- Migr. 2015;9(1–2):34–47. doi:10.1080/19336918.201
cardial infarction. FASEB J. 2017;31(7):3040–53. 5.1008333.
doi:10.1096/fj.201601307R. 41. De Laporte L, Rice JJ, Tortelli F, Hubbell JA.
30. Ruhmann M, Piccinini AM, Kong PL, Midwood KS. Tenascin C promiscuously binds growth factors via
Endogenous activation of adaptive immunity: tenas- its fifth fibronectin type III-like domain. PLoS ONE.
cin-C drives interleukin-17 synthesis in murine arthritic 2013;8(4):e62076. doi:10.1371/journal.pone.0062076.
joint disease. Arthritis Rheum. 2012;64(7):2179–90. 42. Kelly A, Houston SA, Sherwood E, Casulli J, Travis
doi:10.1002/art.34401. MA. Regulation of innate and adaptive immunity by
31. Boniface K, Bak-Jensen KS, Li Y, Blumenschein TGF beta. In: Alt FW, editor. Advances in immunol-
WM, McGeachy MJ, McClanahan TK, McKenzie ogy, vol. 134. San Diego: Elsevier Academic Press;
BS, Kastelein RA, Cua DJ, de Waal Malefyt R. 2017. p. 137–233.
Prostaglandin E2 regulates Th17 cell differentia- 43. Ruegg CR, Chiouetehrismann R, Alkan SS. Tenascin,
tion and function through cyclic AMP and EP2/EP4 an extracellular-matrix protein, exerts immuno-
receptor signaling. J Exp Med. 2009;206(3):535–48. modulatory activities. Proc Natl Acad Sci U S A.
doi:10.1084/jem.20082293. 1989;86(19):7437–41. doi:10.1073/pnas.86.19.7437.
32. Napolitani G, Acosta-Rodriguez EV, Lanzavecchia 44. Hemesath TJ, Marton LS, Stefansson K. Inhibition
A, Sallusto F. Prostaglandin E2 enhances Th17 of T cell activation by the extracellular-matrix protein
responses via modulation of IL-17 and IFN-gamma tenascin. J Immunol. 1994;152(11):5199–207.
production by memory CD4(+) T cells. Eur J Immunol. 45. Hibino S, Kato K, Kudoh S, Yagita H, Okumura K.
2009;39(5):1301–12. doi:10.1002/eji.200838969. Tenascin suppresses CD3-mediated T cell activation.
33. Prieto AL, Edelman GM, Crossin KL. Multiple integ- Biochem Biophys Res Commun. 1998;250(1):119–
rins mediate cell attachment to cytotactin/tenascin. 24. doi:10.1006/bbrc.1998.9258.
Proc Natl Acad Sci U S A. 1993;90(21):10154–8. 46. Navazo MDP, Valmori D, Ruegg C. The alternatively
doi:10.1073/pnas.90.21.10154. spliced domain TnFnIII A1A2 of the extracellular matrix
34. Yokoyama K, Erickson HP, Ikeda Y, Takada Y. protein tenascin-C suppresses activation-induced T
Identification of amino acid sequences in fibrinogen lymphocyte proliferation and cytokine production. J
gamma-chain and tenascin CC-terminal domains Immunol. 2001;167(11):6431–40.
critical for binding to integrin alpha(v)beta(3). J Biol 47. Jachetti E, Caputo S, Mazzoleni S, Brambillasca CS,
Chem. 2000;275(22):16891–8. doi:10.1074/jbc. Parigi SM, Grioni M, Piras IS, Restuccia U, Calcinotto
M000610200. A, Freschi M, Bachi A, Galli R, Bellone M. Tenascin-C
302 Marzeda and Midwood Marzeda and Midwood

protects cancer stem-like cells from immune surveil- 58. El-Karef A, Yoshida T, Gabazza EC, Nishioka T,
lance by arresting T-cell activation. Cancer Res. Inada H, Sakakura T, Imanaka-Yoshida K. Deficiency
2015;75(10):2095–108. doi:10.1158/0008-5472.can- of tenascin-C attenuates liver fibrosis in immune-
14-2346. mediated chronic hepatitis in mice. J Pathol.
48. Saga Y, Yagi T, Ikawa Y, Sakakura T, Aizawa S. 2007;211(1):86–94. doi:10.1002/path.2099.
Mice develop normally without tenascin. Genes Dev. 59. Carey WA, Taylor GD, Dean WB, Bristow JD.
1992;6(10):1821–31. doi:10.1101/gad.6.10.1821. Tenascin-C deficiency attenuates TGF-beta-mediated
49. Forsberg E, Hirsch E, Frohlich L, Meyer M, Ekblom P, fibrosis following murine lung injury. Am J Physiol Lung
Aszodi A, Werner S, Fässler R. Skin wounds and sev- Cell Mol Physiol. 2010;299(6):L785–93. doi:10.1152/
ered nerves heal normally in mice lacking tenascin- ajplung.00385.2009.
C. Proc Natl Acad Sci U S A. 1996;93(13):6594–9. 60. Momcilovic M, Stamenkovic V, Jovanovic M, Andjus
doi:10.1073/pnas.93.13.6594. PR, Jakovcevski I, Schachner M, Miljković Đ.
50. Matsuda A, Yoshiki A, Tagawa Y, Matsuda H, Tenascin-C deficiency protects mice from experimen-
Kusakabe M. Corneal wound healing in tenas- tal autoimmune encephalomyelitis. J Neuroimmunol.
cin knockout mouse. Invest Ophthalmol Vis Sci. 2017;302:1–6. doi:10.1016/j.jneuroim.2016.12.001.
1999;40(6):1071–80. 61. Nishioka T, Onishi K, Shimojo N, Nagano Y, Matsusaka H,
51. Sumioka T, Kitano A, Flanders KC, Okada Y, Ikeuchi M, Ide T, Tsutsui H, Hiroe M, Yoshida T, Imanaka-
Yamanaka O, Fujita N, Iwanishi H, Kao WW, Saika Yoshida K. Tenascin-C may aggravate left ventricular
S. Impaired cornea wound healing in a tenascin remodeling and function after myocardial infarction in mice.
C-deficient mouse model. Lab Invest. 2013;93(2):207– Am J Physiol Heart Circ Physiol. 2010;298(3):H1072–8.
17. doi:10.1038/labinvest.2012.157. doi:10.1152/ajpheart.00255.2009.
52. Tamaoki M, Imanaka-Yoshida K, Yokoyama K, 62. Yamamoto K, Onoda K, Sawada Y, Fujinaga K,
Nishioka T, Inada H, Hiroe M, Sakakura T, Yoshida Imanaka-Yoshida K, Shimpo H, Yoshida T, Yada
T. Tenascin-C regulates recruitment of myofibro- I. Tenascin-C is an essential factor for neointimal
blasts during tissue repair after myocardial injury. hyperplasia after aortotomy in mice. Cardiovasc
Am J Pathol. 2005;167(1):71–80. doi:10.1016/s0002- Res. 2005;65(3):737–42. doi:10.1016/j.cardio-
9440(10)62954-9. res.2004.10.034.
53. Xie K, Liu Y, Hao WL, Walter S, Penke B, Hartmann T, 63. Fujimoto M, Shiba M, Kawakita F, Liu L, Shimojo N,
Schachner M, Fassbender K. Tenascin-C deficiency Imanaka-Yoshida K, Yoshida T, Suzuki H. Deficiency
ameliorates Alzheimer’s disease-related pathology of tenascin-C and attenuation of blood-brain barrier
in mice. Neurobiol Aging. 2013;34(10):2389–98. disruption following experimental subarachnoid hem-
doi:10.1016/j.neurobiolaging.2013.04.013. orrhage in mice. J Neurosurg. 2016;124(6):1693–702.
54. Nakahara H, Gabazza EC, Fujimoto H, Nishii Y, doi:10.3171/2015.4.jns15484.
D’Alessandro-Gabazza CN, Bruno NE, Takagi T, 64. Fujimoto M, Shiba M, Kawakita F, Liu L, Shimojo N,
Hayashi T, Maruyama J, Maruyama K, Imanaka- Imanaka-Yoshida K, Yoshida T, Suzuki H. Effects of
Yoshida K, Suzuki K, Yoshida T, Adachi Y, Taguchi O. tenascin-C knockout on cerebral vasospasm after experi-
Deficiency of tenascin C attenuates allergen-induced mental subarachnoid hemorrhage in mice. Mol Neurobiol.
bronchial asthma in the mouse. Eur J Immunol. 2017. doi:10.1007/s12035-017-0466-x.
2006;36(12):3334–45. doi:10.1002/eji.200636271. 65. Koyama Y, Kusubata M, Yoshiki A, Hiraiwa N, Ohashi
55. Talts JF, Wirl G, Dictor M, Muller WJ, Fassler R. T, Irie S, Kusakabe M. Effect of tenascin-C deficiency
Tenascin-C modulates tumor stroma and monocyte on chemically induced dermatitis in the mouse. J
macrophage recruitment but not tumor growth or Invest Dermatol. 1998;111(6):930–5. doi:10.1046/
metastasis in a mouse strain with spontaneous mam- j.1523-1747.1998.00401.x.
mary cancer. J Cell Sci. 1999;112(12):1855–64. 66. Nakao N, Hiraiwa N, Yoshiki A, Ike F, Kusakabe
56. Tanaka K, Hiraiwa N, Hashimoto H, Yamazaki M. Tenascin-C promotes healing of habu-snake
Y, Kusakabe M. Tenascin-C regulates angiogen- venom-induced glomerulonephritis: studies in knock-
esis in tumor through the regulation of vascular out congenic mice and in culture. Am J Pathol.
endothelial growth factor expression. Int J Cancer. 1998;152(5):1237–45.
2004;108(1):31–40. doi:10.1002/ijc.11509. 67. Okamura N, Hasegawa M, Nakoshi Y, Iino T, Sudo A,
57. Saupe F, Schwenzer A, Jia YD, Gasser I, Spenle Imanaka-Yoshida K, Yoshida T, Uchida A. Deficiency
C, Langlois B, Kammerer M, Lefebvre O, Hlushchuk of tenascin-C delays articular cartilage repair in
R, Rupp T, Marko M, van der Heyden M, Cremel mice. Osteoarthritis Cartilage. 2010;18(6):839–48.
G, Arnold C, Klein A, Simon-Assmann P, Djonov V, doi:10.1016/j.joca.2009.08.013.
Neuville-Méchine A, Esposito I, Slotta-Huspenina J, 68. Busse WW, Lemanske RF. Advances in immunol-
Janssen KP, de Wever O, Christofori G, Hussenet ogy—asthma. N Engl J Med. 2001;344(5):350–62.
T, Orend G. Tenascin-C downregulates wnt inhibitor 69. van den Berg WB, Miossec P. IL-17 as a future
dickkopf-1, promoting tumorigenesis in a neuroen- therapeutic target for rheumatoid arthritis. Nat
docrine tumor model. Cell Rep. 2013;5(2):482–92. Rev Rheumatol. 2009;5(10):549–53. doi:10.1038/
doi:10.1016/j.celrep.2013.09.014. nrrheum.2009.179.
Tenascin-C and Innate Immunity 303

70. Petermann F, Korn T. Cytokines and effector T cell sub- allergic diseases. Hum Mol Genet. 2008;17(11):1673–
sets causing autoimmune CNS disease. FEBS Lett. 82. doi:10.1093/hmg/ddn058.
2011;585(23):3747–57. doi:10.1016/j.febslet.2011.03.064. 80. Saunders CJ, van der Merwe L, Posthumus M, Cook
71. Sawada Y, Onoda K, Imanaka-Yoshida K, Maruyama J, Handley CJ, Collins M, September AV. Investigation
J, Yamamoto K, Yoshida T, Shimpo H. Tenascin-C of variants within the COL27A1 and TNC genes and
synthesized in both donor grafts and recipients Achilles tendinopathy in two populations. J Orthop
accelerates artery graft stenosis. Cardiovasc Res. 2013;31(4):632–7. doi:10.1002/jor.22278.
Res. 2007;74(3):366–76. doi:10.1016/j.cardio- 81. Lowy CM, Oskarsson T. Tenascin C in metastasis: a
res.2007.02.028. view from the invasive front. Cell Adh Migr. 2015;9(1–
72. Pohlers D, Brenmoehl J, Loffler I, Muller CK, Leipner 2):112–24. doi:10.1080/19336918.2015.1008331.
C, Schultze-Mosgau S, Stallmach A, Kinne RW, 82. Orak U, Celik E, Kavak SB, Demirel I, Atilgan R, Aydin
Wolf G. TGF-beta and fibrosis in different organs S, Sapmaz E. Tenascin C levels in patients with mild
- molecular pathway imprints. Biochim Biophys and severe preeclampsia. J Matern Fetal Neonatal
Acta. 2009;1792(8):746–56. doi:10.1016/j.bba- Med. 2016;29(2):270–3. doi:10.3109/14767058.2014.
dis.2009.06.004. 998191.
73. Ohta M, Sakai T, Saga Y, Aizawa S, Saito M. 83. Schenk S, Muser J, Vollmer G, Chiquetehrismann
Suppression of hematopoietic activity in tenascin-C- R. Tenascin-C in serum: a questionable tumor-
deficient mice. Blood. 1998;91(11):4074–83. marker. Int J Cancer. 1995;61(4):443–9. doi:10.1002/
74. Nakamura-Ishizu A, Okuno Y, Omatsu Y, Okabe K, ijc.2910610402.
Morimoto J, Uede T, Nagasawa T, Suda T, Kubota Y. 84. Riedl S, Tandara A, Reinshagen M, Hinz U, Faissner A,
Extracellular matrix protein tenascin-C is required in Bodenmuller H, Buhr HJ, Herfarth C, Möller P. Serum
the bone marrow microenvironment primed for hema- tenascin-C is an indicator of inflammatory bowel dis-
topoietic regeneration. Blood. 2012;119(23):5429–37. ease activity. Int J Colorectal Dis. 2001;16(5):285–91.
doi:10.1182/blood-2011-11-393645. doi:10.1007/s003840100312.
75. Ballard VLT, Sharma A, Duignan I, Holm JM, Chin A, 85. Zavada J, Uher M, Svobodova R, Olejarova M, Husakova
Choi R, Hajjar KA, Wong SC, Edelberg JM. Vascular M, Ciferska H, Hulejová H, Tomčík M, Šenolt L,
tenascin-C regulates cardiac endothelial phenotype Vencovský J. Serum tenascin-C discriminates patients
and neovascularization. FASEB J. 2006;20(2):717–9. with active SLE from inactive patients and healthy con-
doi:10.1096/fj.05-5131fje. trols and predicts the need to escalate immunosup-
76. Song L, Wang L, Li FQ, Yukht A, Qin MH, Ruther pressive therapy: a cohort study. Arthritis Res Ther.
H, Yang M, Chaux A, Shah PK, Sharifi BG. Bone 2015;17:341. doi:10.1186/s13075-015-0862-4.
marrow-derived tenascin-C attenuates cardiac 86. Hasegawa M, Hirata H, Sudo A, Kato K, Kawase D,
hypertrophy by controlling inflammation. J Am Kinoshita N, Yoshida T, Uchida A. Tenascin-C con-
Coll Cardiol. 2017;70(13):1601–15. doi:10.1016/j. centration in synovial fluid correlates with radiographic
jacc.2017.07.789. progression of knee osteoarthritis. J Rheumatol.
77. Zhao YL, Zhao FF, Zong L, Zhang P, Guan LP, Zhang 2004;31(10):2021–6.
JG, Wang D, Wang J, Chai W, Lan L, Li Q, Han B, 87. Hasegawa M, Nakoshi Y, Muraki M, Sudo A, Kinoshita
Yang L, Jin X, Yang W, Hu X, Wang X, Li N, Li Y, Petit N, Yoshida T, Uchida A. Expression of large tenas-
C, Wang J, Wang HY, Wang Q. Exome sequencing cin-C splice variants in synovial fluid of patients with
and linkage analysis identified tenascin-C (TNC) as a rheumatoid arthritis. J Orthop Res. 2007;25(5):563–8.
novel causative gene in nonsyndromic hearing loss. doi:10.1002/jor.20366.
PLoS ONE. 2013;8(7):e69549. doi:10.1371/journal. 88. Page TH, Charles PJ, Piccinini AM, Nicolaidou V,
pone.0069549. Taylor PC, Midwood KS. Raised circulating tenas-
78. Matsuda A, Hirota T, Akahoshi M, Shimizu M, cin-C in rheumatoid arthritis. Arthritis Res Ther.
Tamari M, Miyatake A, Takahashi A, Nakashima K, 2012;14(6):R260. doi:10.1186/ar4105.
Takahashi N, Obara K, Yuyama N, Doi S, Kamogawa 89. Schwenzer A, Jiang X, Mikuls TR, Payne JB, Sayles
Y, Enomoto T, Ohshima K, Tsunoda T, Miyatake HR, Quirke AM, Kessler BM, Fischer R, Venables PJ,
S, Fujita K, Kusakabe M, Izuhara K, Nakamura Y, Lundberg K, Midwood KS. Identification of an immuno-
Hopkin J, Shirakawa T.Coding SNP in tenascin-C dominant peptide from citrullinated tenascin-C as a
Fn-III-D domain associates with adult asthma. Hum major target for autoantibodies in rheumatoid arthritis.
Mol Genet. 2005;14(19):2779–86. doi:10.1093/hmg/ Ann Rheum Dis. 2016;75(10):1876–83. doi:10.1136/
ddi311. annrheumdis-2015-208495.
79. Orsmark-Pietras C, Melen E, Vendelin J, Bruce S, 90. Raza K, Schwenzer A, Juarez M, Venables P, Filer A,
Laitinen A, Laitinen LA,Lauener R, Riedler J, von Mutius Buckley CD, Midwood KS. Detection of antibodies to
E, Doekes G, Wickman M, Van Hage M, Pershagen G, citrullinated tenascin-C in patients with early synovi-
Scheynius A, Nyberg F, Kere J, PARSIFAL Genetics tis is associated with the development of rheumatoid
Study Group. Biological and genetic interaction arthritis. RMD Open. 2016;2(2):e000318. doi:10.1136/
between Tenascin C and Neuropeptide S receptor 1 in rmdopen-2016-000318.
304 Marzeda and Midwood Marzeda and Midwood

91. Franz M, Jung C, Lauten A, Figulla HR, Berndt A. 96. Boros P, Ochando J, Zeher M. Myeloid derived sup-
Tenascin-C in cardiovascular remodeling: potential pressor cells and autoimmunity. Hum Immunol.
impact for diagnosis, prognosis estimation and tar- 2016;77(8):631–6. doi:10.1016/j.humimm.2016.05.024.
geted therapy. Cell Adh Migr. 2015;9(1–2):90–5. doi 97. Baniyash M. Myeloid-derived suppressor cells as intrud-
:10.1080/19336918.2014.1000075. ers and targets: clinical implications in cancer therapy.
92. Okuma Y, Suda K, Nakaoka H, Katsube Y, Mitani Cancer Immunol Immunother. 2016;65(7):857–67.
Y, Yoshikane Y, Ichida F, Matsushita T, Shichino doi:10.1007/s00262-016-1849-y.
H, Shiraishi I, Abe J, Hiroe M, Yoshida T, Imanaka- 98. Ray A, Chakraborty K, Ray P. Immunosuppressive
Yoshida K. Serum tenascin-C as a novel predictor for risk MDSCs induced by TLR signaling during infec-
of coronary artery lesion and resistance to intravenous tion and role in resolution of inflammation. Front
immunoglobulin in Kawasaki disease—a multicenter Cell Infect Microbiol. 2013;3:52. doi:10.3389/
retrospective study. Circ J. 2016;80(11):2376–81. fcimb.2013.00052.
doi:10.1253/circj.CJ-16-0563. 99. Arora M, Poe SL, Oriss TB, Krishnamoorthy N,
93. Kitaoka H, Kubo T, Baba Y, Yamasaki N, Matsumura Yarlagadda M, Wenzel SE, Billiar TR, Ray A, Ray P.
Y, Furuno T, Doi YL. Serum tenascin-C levels as TLR4/MyD88-induced CD11b+Gr-1 int F4/80+ non-
a prognostic biomarker of heart failure events in migratory myeloid cells suppress Th2 effector func-
patients with hypertrophic cardiomyopathy. J Cardiol. tion in the lung. Mucosal Immunol. 2010;3(6):578–93.
2012;59(2):209–14. doi:10.1016/j.jjcc.2011.11.008. doi:10.1038/mi.2010.41.
94. Saiwai H, Kumamaru H, Ohkawa Y, Kubota K, Kobayakawa 100. Sinha P, Okoro C, Foell D, Freeze HH, Ostrand-
K, Yamada H, Yokomizo T, Iwamoto Y, Okada S. Ly6C+ Rosenberg S, Srikrishna G. Proinflammatory
Ly6G- Myeloid-derived suppressor cells play a critical role S100 proteins regulate the accumulation of
in the resolution of acute inflammation and the subsequent myeloid-derived suppressor cells. J Immunol.
tissue repair process after spinal cord injury. J Neurochem. 2008;181(7):4666–75.
2013;125(1):74–88. doi: 10.1111/jnc.12135. 101. Spenle C, Saupe F, Midwood K, Burckel H, Noel
95. Kumar V, Patel S, Tcyganov E, Gabrilovich DI. The G, Orend G. Tenascin-C: exploitation and collat-
nature of myeloid-derived suppressor cells in the tumor eral damage in cancer management. Cell Adh Migr.
microenvironment. Trends Immunol. 2016;37(3):208– 2015;9(1–2):141–53. doi:10.1080/19336918.2014.10
20. doi:10.1016/j.it.2016.01.004. 00074.

You might also like