You are on page 1of 21

Cytokine and Growth Factor Reviews 39 (2018) 71–91

Contents lists available at ScienceDirect

Cytokine and Growth Factor Reviews


journal homepage: www.elsevier.com/locate/cytogfr

Review article

Cytokines in the pathogenesis of hemophilic arthropathy T


a,b,c,⁎
Piotr Wojdasiewicz , Łukasz A. Poniatowski , Paweł Nauman , Tomasz Mandat , d e e,f

Agnieszka Paradowska-Goryckag, Katarzyna Romanowska-Próchnickaa,h, Dariusz Szukiewicza,


Andrzej Kotelai,j, Łukasz Kubaszewskik,l, Ireneusz Kotelaj, Iwona Kurkowska-Jastrzębskam,
Robert Gasikc
a
Department of General and Experimental Pathology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Pawińskiego 3C, 02-106
Warsaw, Poland
b
Department of Rheumaorthopaedics, Eleonora Reicher National Institute of Geriatrics, Rheumatology and Rehabilitation, Spartańska 1, 02-637 Warsaw, Poland
c
Department of Neuroorthopaedics and Neurology, Eleonora Reicher National Institute of Geriatrics, Rheumatology and Rehabilitation, Spartańska 1, 02-637 Warsaw, Poland
d
Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Banacha 1B, 02-097
Warsaw, Poland
e
Department of Neurosurgery, Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957 Warsaw, Poland
f
Department of Neurosurgery, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, W. K. Roentgena 5, 02-781 Warsaw, Poland
g
Department of Biochemistry and Molecular Biology, Eleonora Reicher National Institute of Geriatrics, Rheumatology and Rehabilitation, Spartańska 1, 02-637 Warsaw,
Poland
h
Department of Systemic Connective Tissue Diseases, Eleonora Reicher National Institute of Geriatrics, Rheumatology and Rehabilitation, Spartańska 1, 02-637 Warsaw, Poland
i
Department of Orthopaedics and Traumatology, 1st Faculty of Medicine, Medical University of Warsaw, Lindleya 4, 02-005 Warsaw, Poland
j
Department of Orthopaedics and Traumatology, Central Clinical Hospital of the Ministry of the Interior and Administration, Wołoska 137, 02-507 Warsaw, Poland
k
Department of Spondyloorthopaedics and Biomechanics of the Spine, Wiktor Dega Orthopaedic and Rehabilitation Clinical Hospital, Poznań University of Medical
Sciences, 28 Czerwca 1956 135/147, 61-545 Poznań, Poland
l
Department of Orthopaedics and Traumatology, Wiktor Dega Orthopaedic and Rehabilitation Clinical Hospital, Poznań University of Medical Sciences, 28 Czerwca 1956
135/147, 61-545 Poznań, Poland
m
2nd Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957 Warsaw, Poland

A R T I C L E I N F O A B S T R A C T

Keywords: Hemophilic arthropathy (HA) is one of the most common and typical manifestation in the course of recurrent
Cytokines bleeding episodes in patients with hemophilia. Clinical and subclinical joint bleeding episodes gradually lead to
Hemophilic arthropathy irreversible changes manifesting themselves as pain, progressing ankylosis, marked limitation of the range of
Hemophilia motion, muscle atrophy and osteoporosis commonly concomitant with joint deformity resulting from chronic
Arthritis
proliferative synovitis and both cartilage and bone degeneration leading to the final functional impairment of the
Inflammation
Synovitis
joint. In spite of numerous studies, the pathophysiology of HA has not been fully elucidated, especially as regards
immunopathological mechanisms which are associated with the subclinical and early stage of the disease and to
be more precise, with chronic joint inflammation. It needs to be emphasized that the pathophysiological pro-
cesses occurring in a joint with HA are most probably highly mediated by interactions within the cytokine
network and other inflammatory mediators present in the tissues of affected joint. Among numerous compounds
participating in the induction of an inflammatory process in the pathogenesis of HA, cytokines seem to play a
leading role. The most important group controlling the disease seems to be well known inflammatory cytokines,
including IL-1β, TNFα and IL-6. The second group with antagonistic effect is formed by anti-inflammatory
cytokines such as IL-4 and IL-10. The role of inflammatory and anti-inflammatory cytokines in the pathogenesis
of HA with respect to cellular and intracellular signaling pathways is still under investigation. This review,
summarizes and discusses the current knowledge about cytokine network in the pathogenesis of HA, indicating
possible molecular and cellular mechanisms that may provide potential new therapeutic directions.


Corresponding author at: Department of General and Experimental Pathology, Centre for Preclinical Research and Technology (CePT), Medical University of Warsaw, Pawińskiego
3C, 02-106 Warsaw, Poland.
E-mail addresses: piotr.wojdasiewicz@wum.edu.pl (P. Wojdasiewicz), lukasz.poniatowski@gmail.com (Ł.A. Poniatowski), pnauman@ipin.edu.pl (P. Nauman),
tomaszmandat@yahoo.com (T. Mandat), paradowska_aga@interia.pl (A. Paradowska-Gorycka), katarzyna.prochnicka@gmail.com (K. Romanowska-Próchnicka),
dszukiewicz@hotmail.com (D. Szukiewicz), andrzejkotela@op.pl (A. Kotela), pismiennictwo1@gmail.com (Ł. Kubaszewski), ikotela@op.pl (I. Kotela),
ikurkowska@ipin.edu.pl (I. Kurkowska-Jastrzębska), robertgasik2@gmail.com (R. Gasik).

https://doi.org/10.1016/j.cytogfr.2017.11.003
Received 4 November 2017; Accepted 9 November 2017
Available online 13 November 2017
1359-6101/ © 2017 Elsevier Ltd. All rights reserved.
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

Fig. 1. Schematic representation of the coagulation cascade and simplified life cycle of FVIII and FIX including their domain structure and processing under physiological conditions.
Hemostasis mechanisms defined as a system of proteolytic reactions, are activated by two distinct pathways, through intrinsic pathway associated with the formation of ITC (FVIIIa/FIXa/
FX/PM/Ca2+) and by extrinsic pathway associated with the formation of ETC (FVIIa/FX/TF/PM/Ca2+). Both of these converge on the activation of common pathway associated with the
formation of PTC (FII/FVa/FXa/PM/Ca2+) and catalytic generation of FIIa which is essential for conversion of FI into FIa. Ultimately, FIa monomers polymerization along with platelet
aggregation leads to formation of a stable cross-linked fibrin clot. The tightly controlled process such a hemostasis can be disrupted by genetic deficiency or reduction in the activity of
FVIII or FIX. In this conditions assembly and functioning of ITC is impaired leading to prolonged bleeding (APTT ≥ 30–40 s). This types of congenital disturbances in hemostatic system
are known as hemophilia A and B. Abbreviations: APTT activated partial thromboplastin time, HMWK high molecular weight kininogen (Fitzgerald factor), PK prekallikrein (Fletcher
factor), K kallikrein, FXII coagulation factor XII (Hageman factor), FXIIa active coagulation factor XII, FXI coagulation factor XI (plasma thromboplastin antecedent), FXIa active
coagulation factor XI, FIX coagulation factor IX (Christmas factor), GLA gamma-carboxyglutamic acid-rich domain (1–40 aa), EGF1 epidermal growth factor 1-like domain (47–83 aa),
EGF2 epidermal growth factor 2-like domain (88–127 aa), AP activation peptide (146–180 aa), CAT catalytic domain (181–415 aa), FIXa active coagulation factor IX, FVIII coagulation
factor VIII (anti-hemophilic factor), A1 A1 domain (1–336 aa), a1 a1 acidic region (337–372 aa), A2 A2 domain (373–710 aa), a2 a2 acidic region (711–740 aa), B B domain (741–1648
aa), a3 a3 acidic region (1649–1689 aa), A3 A3 domain (1690–2019 aa), C1 C1 domain (2020–2172 aa), C2 C2 domain (2173–2332 aa), vWF von Willebrand factor, FVIIIHC coagulation
factor VIII heavy chain (A1-a1-A2-a2-B), FVIIILC coagulation factor VIII light chain (a3-A3-C1-C2), FVIIIa active coagulation factor VIII, PM phospholipid membrane, ITC intrinsic tenase
complex, PT prothrombin time, INR international normalized ratio, TF (FIII) tissue factor (coagulation factor III), FVII coagulation factor VII (proconvertin), FVIIa active coagulation
factor VII, ETC extrinsic tenase complex, TT thrombin time, FX coagulation factor X (Stuart factor), FXa active coagulation factor X, FV coagulation factor V (proaccelerin), FVa active
coagulation factor V, PTC prothrombinase complex, FII prothrombin, FIIa thrombin, FI fibrinogen, FIa fibrin, FXIII coagulation factor XIII (fibrin-stabilizing factor), FXIIIa active
coagulation factor XIII.

72
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

1. Introduction Active FIX (FIXa), and acting as a cofactor active FVIII (FVIIIa), parti-
cipate in ITC forming in the presence of phospholipid membrane (PM)
Hemophilia is a congenital coagulation disorder caused by the re- and calcium ions (Ca2+) [9,10]. The kinetics of ITC complex in the
duced activity or the lack of plasma glycoproteins such as coagulation catalytic generation of FXa is approximately 50-fold more efficient
factor VIII (FVIII), which leads to the development of hemophilia A, and (kcat/Km) than extrinsic tenase complex (ETC) and therefore, its in-
analogous reduction in the activity or the absence of coagulation factor sufficiency results in inhibition of assembly of prothrombinase complex
IX (FIX), which leads to the development of hemophilia B (Christmas (PTC) and the impairment of thrombin (FIIa) production
disease) [1,2]. Both hemophilia A and B are sex-linked disorders de- (VITC→FXa ≥ 50 x VETC→FXa) [11,12]. Depending on the level of FVIII
termined by the presence of alleles on chromosome X (Xh), inherited in and FIX in the serum, we distinguish the following forms of the disease:
a monogenic recessive pattern, where females (46,XX) are carriers severe < 0.01 IU/mL (< 1% of normal level), moderate 0.01–0.05 IU/
(46,XhX), and the disease is present in males (46,XY) having a complete mL (1–5% of normal level) and mild > 0.05 to < 0.40 IU/mL (> 5%
expression of the recessive gene (46,XhY) [2]. Around 1/3 of the cases to < 40% of normal level) [13].
are caused by spontaneous mutations (de novo), without no prior fa- The main phenotypic manifestations of the mutations on chromo-
mily history of hemophilia [1]. The reduced activity of FVIII and FIX some X are hemorrhages which are a part of the comprehensive clinical
oscillating around the reference values may be commonly observed in picture in hemophilia patients. The type, location and severity of he-
hemophilia gene carriers and in some rare cases the carriers may also morrhages are directly connected with factors including the type and
suffer from hemophilia (46,XhXh) [1,2]. Moreover, the clinically ob- level of coagulation factor deficiency, patient’s age and a potentially
servable picture of the disease may be also affected by other genetic introduced treatment, however, the majority of cases are heterogenous
factors leading to a multifactorial disease [3]. The analysis of currently bleeding phenotypes [14]. According to the WFH, the most common
available data from long-term epidemiological research shows that and typical manifestations in the course of severe hemophilia are non-
hemophilia is a disease carrying a heavy economic burden with its di- traumatic (spontaneous) hemorrhages within the musculoskeletal
rect and indirect costs [4]. It is also associated with limiting the health- system, with spontaneous intraarticular bleeding (hemarthrosis) ac-
related quality of life (HRQoL) [4]. counting for 70–80% of all the hemorrhages, and intramuscular he-
Taking into consideration global epidemiological data on hemo- morrhages (the second largest group) accounting for 10–20% of all the
philia, according to the monitoring by the Centers for Disease Control hemorrhages occurring in patients [15]. Bleeding episodes are most
and Prevention (CDC) and World Federation of Hemophilia (WFH) the frequently reported in large synovial joints, like the elbow, knee and
prevalence of hemophilia A is around 1 in 5000 male births, and 1 in ankle [15]. Less frequently in multiaxial joints such as the shoulder and
30000 male births for hemophilia B and occurs in approximately 1 out the hip joint [15]. Recurrent clinical and subclinical joint bleeding
of 7500 live male births, affecting all racial and socioeconomic groups episodes gradually lead to irreversible changes manifesting themselves
equally [5–8]. as pain, progressing ankylosis, marked limitation of the range of motion
The disturbances of hemostasis that are considered to be associated (ROM), muscle atrophy and osteoporosis commonly concomitant with
with the reduction of activity or absence of FVIII and FIX are related to joint deformity resulting from chronic proliferative synovitis (syno-
the impairment of intrinsic tenase complex (ITC) functioning in the vium-mediated component) and cartilage degeneration (cartilage-
activation of coagulation factor X (FX) to active FX (FXa) [9] (Fig. 1). mediated component) leading to the final functional impairment of the

Fig. 2. Radiographs showing the advanced hemo-


philic arthropathy of the knee in anteroposterior (A)
and lateral (B) view. In this case, we can observe
symmetrical full-thickness articular cartilage loss at
both femoral and tibial condyle associated with
subchondral bone destruction resulting in cyst for-
mation.

73
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

Fig. 3. Schematic overview of the potential cellular and molecular mechanisms by which IL-1β and its associated signaling pathways are involved in expression and functional regulation
of various genes involved in inflammatory and catabolic processes in the pathogenesis of hemophilic arthropathy. Abbreviations: IL-1β interleukin 1 beta, IL-1RA interleukin 1 receptor
antagonist, anti-IL-1β antibodies against interleukin 1 beta, IL-1R1 (CD121a) interleukin 1 receptor type 1, IL-1R2 (CD121b) interleukin 1 receptor type 2, IL-1RAcP interleukin 1 receptor
accessory protein, D1 immunoglobulin-like domain 1, D2 immunoglobulin-like domain 2, D3 immunoglobulin-like domain 3, TIR Toll/IL-1R homology domain, MyD88 myeloid
differentiation primary response 88 protein, IRAK4 interleukin 1 receptor associated kinase 4, IRAK2 interleukin 1 receptor associated kinase 2, IRAK3 interleukin 1 receptor associated
kinase 3, IRAK1 interleukin 1 receptor associated kinase 1, TRAF6 TNF receptor associated factor 6, IKKα inhibitor of nuclear factor kappa B kinase subunit alpha, NEMO nuclear factor
kappa B essential modulator, IKKβ inhibitor of nuclear factor kappa B kinase subunit beta, IκB inhibitor kappa B kinase complex, p50 nuclear factor kappa B subunit 1, p65 nuclear factor
kappa B p65 subunit, TAB2 TGF-β activated kinase 1/MAP3K7 binding protein 2, TAK1 mitogen-activated protein kinase kinase kinase 7, TAB3 TGF-β activated kinase 1/MAP3K7
binding protein 3, ERK1/2 extracellular signal-regulated kinase 1/2, JNK c-Jun N-terminal kinase, p38 p38 mitogen-activated protein kinase, AP-1 activator protein 1, MMP-1 interstitial
collagenase, MMP-3 stromelysin 1, MMP-13 collagenase 3, ADAM-TS4 a disintegrin and metalloproteinase with thrombospondin motifs 4, TNFα tumor necrosis factor alpha, IL-6
interleukin 6, IL-8 interleukin 8, CCL5 C-C motif chemokine ligand 5, PLA2 phospholipase A2, COX-2 cyclooxygenase 2, PGE2 prostaglandin E2, iNOS inducible nitric oxide synthase,
ACAN aggrecan, COL2 collagen type II; ↑ increase of expression, ↓ decrease of expression.

74
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

joint [16,17]. Hemophilic arthropathy (HA) is a syndrome connected and negative feedback exerted on cells and the production of other
with blood-induced joint disease (BIJD) in the course of hemophilia cytokines forms a complex multilevel system of correlations known as
[17] (Fig. 2). cytokine network [30]. It needs to be emphasized that the potential role
The correlation between hemorrhages occurring in the course of of cytokines and inflammatory mediators in the pathomechanism and
hemophilia and arthritis was first reported by a German surgeon Franz pathophysiology of numerous arthritis-associated diseases such as os-
König in 1892, who was also the first to perform joint surgery in he- teoarthritis (OA) and rheumatoid arthritis (RA) have been described
mophilia patients [18]. The natural history of HA on an animal model [31,32]. In this review, we describe the participation of the cytokine
was first described by Margaret Swanton in 1959 [19]. While there is no network in the course of HA, and will analyse the role of inflammatory
uniform consensus, it is considered that the annual joint bleeding rate and anti-inflammatory cytokine-driven processes occurring in HA-af-
(AJBR) at the level of 2–3 predisposes to the development of irrever- fected joints, late sequelae and possible clinical implications.
sible pathobiological and structural changes within the tissues of the
joint [20]. Regarding in vitro testing results, research conducted on
animals and histological analysis, it is assumed that the process may be 2. Inflammatory cytokines
due to a single clinical episode of hemarthrosis and potentially occur-
ring subclinical bleedings [20]. According to Fisher et al. the median The role of inflammatory cytokines in the pathogenesis of HA
age at the time of the first joint bleeding is 2.2 years (range: 0.2–5.8 mainly involves promoting catabolic processes in joint-forming tissues
years) and 90% of patients had experienced at least one joint bleeding and intensifying an inflammatory response [17,25]. In the described
at the age of 4.4 years [21]. Although an effective prophylactic factor context catabolic processes are mainly seen in the cells and extra-
replacement therapy (FRT) was developed which considerably reduces cellular matrix of the articular cartilage which, as a result of the dis-
the occurrence of joint bleeding episodes, there are still patients with a ruption of its metabolic homeostasis, undergoes gradual atrophy and
clinically advanced form of HA (30–50%) in many regions of the world, destruction [33]. However, the intensification of immune response
also in highly developed countries [15,22]. HA is the main cause of should be associated with immune system cells which may be activated
morbidity in patients with hemophilia [15,23]. In the most severe types and stimulated by cytokines as a result of autocrine, paracrine and
of hemostasis disturbances associated with hemophilia even appro- endocrine properties acting via pleiotropy, redundancy, synergy and
priate and regular use of prophylactic FRT may not prevent the de- antagonism [30]. Periarticular tissues are also sensitive to in-
velopment of advanced forms of HA, which may be due to the devel- flammatory cytokine activity, especially the synovial membrane or its
opment of alloimmune antibodies directed against epitopes of infused vascular endothelial cells, which become a potentially dynamic re-
FVIII (anti-FVIII) or FIX (anti-FIX) known as inhibitors, occurring in servoir of cells and mediators participating in an inflammation after the
approximately 10–30% hemophilia A patients and 2–5% of hemophilia activation of intracellular pathways propagating an inflammatory re-
B patients [24]. sponse [34]. Currently, interleukin 1 beta (IL-1β), tumor necrosis factor
In spite of numerous studies, the pathophysiology of HA has not alpha (TNFα) and interleukin 6 (IL-6) are the best described in-
been fully elucidated, especially as regards immunopathological me- flammatory cytokines which take part in the pathogenesis of HA.
chanisms which are associated with the subclinical and early stage of
the disease and to be more precise, with intraarticular inflammation
[15,25]. The presence of extravasated blood in the joint is the trigger of 2.1. Interleukin 1 beta (IL-1β)
an inflammation which initiates BIJD [17]. The morphotic elements of
blood include numerous immune system cells, such as granulocytes, 2.1.1. Overview of the IL-1β properties and its associated signaling
monocytes and lymphocytes, and it needs to be emphasized that after pathways
extravasation these elements remain active in the process of secretion of The activity of IL-1β as one of the main regulators of inflammatory
inflammatory mediators [26]. Then, blood and its metabolites are response consists of inducing catabolic processes in the articular carti-
gradually absorbed by macrophages (MΦ) and synovial cells [17]. In lage and other elements making up the joint [17,25] (Fig. 3). IL-1β
case of recurrent episodes of hemarthrosis, the potential of blood me- belongs to interleukin 1 family (IL-1F), which consists of 11 ligand
tabolite removal by immune system cells present in the joint and the members [35]. IL-1β precursor is synthesized as a cytosolic protein
synovial membrane gradually depletes [17]. It results in an extended (pro-IL-1β), which is composed of 269 amino acid residues [36]. After
presence of toxic blood metabolites, such as erythrocyte-derived iron the proteolysis by the cysteine-aspartic acid protease, caspase 1
(Fe2+) and deposition of hemosiderin in the synovium which is asso- (CASP1, ICE), a member of the multiprotein oligomer NLR family pyrin
ciated with the production of reactive oxygen species (ROS) via the domain containing 3 (NLRP3) inflammasome, the precursor is con-
Haber-Weiss/Fenton reaction (Fe2+ + H2O2 → Fe3+ + OH% + OH−) verted into an active form composed of 153 amino acid residues, and
and, additionally, induction of oxidative stress [27,28]. These processes then secreted to the extracellular space [25,37]. It has an ability to
lead to pathological changes including cartilage and subchondral bone adhere to two types of receptors. One of them is interleukin 1 receptor
destruction, increased fibroblast proliferation and angiogenesis within type 1 (IL-1R1, CD121a), belonging to Toll-like receptors (TLR) and
the synovial membrane resulting in its hypertrophy which is also as- interleukin 1 receptor (IL-1R) superfamily, which has the affinity for
sociated with overexpression of mouse double minute 2 homolog other cytokines from IL-1F family, such as interleukin 1 alpha (IL-1α)
(MDM2) and v-myc avian myelocytomatosis viral oncogene homolog and interleukin 1 receptor antagonist (IL-1RA) [38]. Another receptor is
(c-MYC) [17]. interleukin 1 receptor type 2 (IL-1R2, CD121b), which includes the
It has already been mentioned that in case of HA and other arthritis- same set of possible ligands as IL-1R1, with the reservation that IL-1R2
associated diseases the direct process of intensified degeneration of forms an inactive ligand-receptor complex which is unable to propagate
joint tissues is due to a chronic inflammation [25]. Among numerous further signals of intracellular communication [39]. Intracellular
compounds participating in the induction of an inflammatory process in phosphorylation of subsequent proteins due to IL-1R1, leads to the
HA pathogenesis cytokines seem to play a leading, but not fully eluci- activation of nuclear factor kappa-light-chain-enhancer of activated B
dated role [25]. Cytokines form a heterogeneous group of mediators cells (NF-κB) transcriptive factor and other transcriptive factors such as
responsible for such processes as activation and proliferation of im- c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein ki-
mune system cells and the change of the metabolism of various types of nases (p38MAPK) [40]. As a result, there is an increased expression of
cells including, what is important in this context, cells which make up various genes responsible for the synthesis of enzymes, adhesion mo-
the joint and potentially infiltrate its structures via blood vessels [29]. lecules or inflammatory mediators including cytokines and chemokines
Cytokines and their individual, highly specific influence via positive [41].

75
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

HA hemophilic arthropathy, MNC mononuclear cells, RBC red blood cells, LPS lipopolysaccharide, F8-KO F8 gene knock-out, CD14+ cluster of differentiation 14 cells, ELISA enzyme-linked immunosorbent assay, IHC immunohistochemistry.
2.1.2. Biological functioning of the IL-1β in joint tissues

Reference
IL-1β may be synthesized within joint tissues both by synovial

[115]
[118]
[131]
[54]
[55]
[56]
[57]
[58]
[60]
[62]
[63]
membrane cells, chondrocytes, osteoblasts and mononuclear cells in the
course of an inflammation [42]. IL-1β demonstrates a multidirectional
effect on the articular cartilage via simultaneous intensification of its

of incubation
of incubation

of incubation
of incubation
of incubation
disintegration and inhibition of regenerative processes [43]. Under the
influence of IL-1β, chondrocytes increase the synthesis of enzymes from
matrix metalloproteinase (MMP) group, including interstitial col-
lagenase (MMP-1), stromelysin 1 (MMP-3), collagenase 3 (MMP-13),

cultures after 4 days


cultures after 4 days

cultures after 4 days


cultures after 4 days
cultures after 4 days
which influence the excessive degradation of extracellular matrix mo-

subchondral bone
lecules [44]. IL-1β also results in an increased proteolysis of aggrecan

synovial fluid
supernatants
supernatants
supernatants

supernatants
(ACAN) molecules by promoting the secretory activity of cartilage cells
in order to produce proteinases from a disintegrin and metalloprotei-
nase with thrombospondin motifs (ADAM-TS) family, and more pre-
cisely ADAM-TS4 [44]. In addition to catabolic processes affecting the

concentration in the
concentration in the
concentration in the

concentration in the
concentration in the

concentration in the
secretion by the cell
secretion by the cell

secretion by the cell


secretion by the cell
secretion by the cell
extracellular matrix, IL-1β also causes a significant reduction in the
synthesis of the substances like collagen type II (COL2) and ACAN,

Reported results/timing
which leads to a gradual loss of cartilage density and its mechanical
resistance [45]. The combination of IL-1β and TNFα led to a tendency
towards accelerated ageing process and more frequent induction of

of
of
of
of
of
of
of
of
of
of
of
apoptosis in chondrocytes [46]. This may be directly related to the

Increase
Increase
Increase
Increase
Increase
Increase
Increase
Increase
Increase
Increase
Increase
property of IL-1β to increase the production of ROS and induce oxi-
dative stress [47]. IL-1β generates the development of substances such
as hydroxyl radicals (OH%) and peroxides (H2O2) which have been
shown to destroy the structure of articular cartilage [47]. Thus, the

Methodology for quantification


catabolic properties of IL-1β are not only due to a direct influence on
chondrocyte metabolism, but also to the stimulation of the production
of inflammatory mediators in all types of cells which make up the joint
[48]. IL-1β triggers the synthesis of numerous inflammatory cytokines
such as TNFα, IL-6, interleukin 8 (IL-8), C-C motif chemokine ligand 5

Luminex xMAP
(CCL5, RANTES) and also induces self-secretion via an autocrine me-
chanism [49]. Moreover, it increases the production and causes the
ELISA
ELISA
ELISA
ELISA
ELISA

ELISA
ELISA
ELISA
ELISA
overexpression of inflammatory enzymes and molecules including
phospholipase A2 (PLA2), cyclooxygenase 2 (COX-2), prostaglandin E2 IHC
(PGE2) and inducible nitric oxide synthase (iNOS) [50–52].
Subchondral bone samples

2.1.3. Role of IL-1β in the pathogenesis of hemophilic arthropathy


Synovial tissue samples
Synovial tissue samples
Synovial tissue samples

Synovial tissue samples


Synovial fluid samples

Numerous studies implicate IL-1β production in the pathophy-


Method of sampling

Cartilage cultures
Cartilage cultures

Cartilage cultures
Cartilage cultures
Cartilage cultures

siology of HA [17,25,37,53] (Table 1). The level of IL-1β is markedly


elevated in the synovial membranes harvested via synovectomy or joint
replacement from patients with HA compared to the control group
[54,55]. Roosendaal et al. analysed cell cultures from synovial tissue of
Observational studies showing the expression pattern of IL-1β in hemophilic arthropathy.

patients with HA and reported that the supernatants had higher levels
of IL-1β compared to the control group [56]. A significantly elevated
level of IL-1β secretion was also observed in the supernatants of human
Cartilage co-cultured with whole blood, MNC, MNC + RBC or MNC + LPS

articular cartilage cell cultures which underwent a short-term contact


Cartilage co-cultured with whole blood, MNC + RBC or RBC + CD14+

with the blood [57,58]. It is also currently suggested that IL-1β in-
creased transferrin (TFN)-bound iron uptake into synovial fibroblasts
which was significant in the context of chronic synovitis associated with
F8-KO mouse with needle-induced knee joint hemarthrosis

deposition of hemosiderin and IL-1β autocrine secretion [59]. A de-


Cartilage co-cultured with whole blood or MNC + RBC

tailed study by Øvlisen et al. conducted on a mouse model of hemo-


philia A demonstrated changes in the concentration of numerous cy-
tokines, including IL-1β, in the synovial fluid [60]. The study group
included FVIII gene (F8) knock-out (F8-KO) mice and the control group
included C57Bl/6NTac mice, divided into four subgroups. The first
Cartilage co-cultured with whole blood
Cartilage co-cultured with whole blood

group did not receive any clotting factor, only a carrier (an excipient
administered with clotting factors) was administered into the tail vein.
The remaining groups received various clotting factors: the second
group – recombinant FVIII (rFVIII), the third group − recombinant
activated factor VII (rFVIIa) and the fourth group − rFVII analogue
Clinical trial/model

(NN1731). All the animals from the control and study group, then had a
Patients with HA
Patients with HA
Patients with HA

Patients with HA

Patients with HA

knee injury made with a needle. After 3 days, a post-mortem analysis


was performed, which included parameters like the level of cytokines in
the synovial fluid of injured joints. The level of IL-1β in the synovial
Table 1

fluid in F8-KO mice which received placebo was considerably higher


(18.0 ± 26.2 pg/mL) than in the mice from the healthy control group

76
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

Fig. 4. Schematic overview of the potential cellular and molecular mechanisms by which TNFα and TNFR1 signaling complex are involved in expression and functional regulation of
various genes involved in inflammatory and catabolic processes in the pathogenesis of hemophilic arthropathy. Abbreviations: sTNFα soluble tumor necrosis factor alpha, mTNFα
membrane tumor necrosis factor alpha, anti-TNFα antibodies against tumor necrosis factor alpha, ADAM17 (TACE) a disintegrin and metalloproteinase domain 17 (tumor necrosis factor
alpha-converting enzyme), EGF epidermal growth factor-like domain, CR cysteine-rich domain, DS disintegrin domain, MP catalytic metalloproteinase domain, iRhom2 intramembrane
serine proteases rhomboid 5 homolog 2, TNFR1 tumor necrosis factor receptor 1, DD death domain, TRADD TNFRSF1A protein associated via death domain, FADD Fas associated via
death domain, CASP8 caspase 8, RIPK1 receptor interacting serine-threonine kinase 1, Ub ubiquitin, TRAF2 TNF receptor associated factor 2, cIAP1 baculoviral IAP repeat containing 2,
cIAP2 baculoviral IAP repeat containing 3, TAB2 TGF-β activated kinase 1/MAP3K7 binding protein 2, TAK1 mitogen-activated protein kinase kinase kinase 7, TAB3 TGF-β activated
kinase 1/MAP3K7 binding protein 3, ERK1/2 extracellular signal-regulated kinase 1/2, JNK c-Jun N-terminal kinase, p38 p38 mitogen-activated protein kinase, AP-1 activator protein 1,
IL-1β interleukin 1 beta, TNFα tumor necrosis factor alpha, IL-6 interleukin 6, C5a complement component 5a, VEGF vascular endothelial growth factor, FIIa thrombin, Hb hemoglobin,
Fe2+ erythrocyte-derived iron, MMP-1 interstitial collagenase, MMP-3 stromelysin 1, MMP-13 collagenase 3, ADAM-TS4 a disintegrin and metalloproteinase with thrombospondin motifs
4, IL-6 interleukin 6, IL-8 interleukin 8, CCL5 C-C motif chemokine ligand 5, SDF-1 stromal cell-derived factor 1, VEGF vascular endothelial growth factor, HIF-1α hypoxia inducible
factor 1 alpha, HIF-2α hypoxia inducible factor 2 alpha, ACAN aggrecan, COL2 collagen type II; ↑ increase of expression, ↓ decrease of expression.

77
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

(1.0 ± 6.5 pg/mL). Administration of clotting factors markedly re- correlated with changes observed in the joint with visual bleeding score
duced the level of IL-1β in the synovial fluid, with rFVIII (VBS) [61]. Control group mice and those after rFVIII administration
(3.7 ± 18.7 pg/mL) being the most efficient in this context. The results manifested significantly fewer cases of joint capsule oedema and he-
of the substitution of rFVIIa and NN1731 were comparable marthrosis than the mice that underwent the substitution of rFVIIa,
(10.0 ± 19.0 pg/mL and 11.0 ± 30.9 pg/mL, respectively). Im- NN1731 and after receiving placebo. The last group had a distended,
portantly, the levels of IL-1β and other inflammatory cytokines oedematous joint capsule and hemarthrosis in almost every case.

Fig. 5. Schematic overview of the potential cellular and molecular mechanisms by which TNFα and TNFR2 signaling complex are involved in expression and functional regulation of
various genes involved in inflammatory and catabolic processes in the pathogenesis of hemophilic arthropathy. Abbreviations: mTNFα membrane tumor necrosis factor alpha, anti-TNFα
antibodies against tumor necrosis factor alpha, TNFR2 tumor necrosis factor receptor 2, TRAF1 TNF receptor associated factor 1, TRAF2 TNF receptor associated factor 2, ETK BMX non-
receptor tyrosine kinase, PI3K phosphatidylinositol-4,5-bisphosphate 3-kinase, AKT protein kinase B, mTOR mechanistic target of rapamycin, RIPK1 receptor interacting serine-threonine
kinase 1, Ub ubiquitin, TRAF3 TNF receptor associated factor 3, cIAP1 baculoviral IAP repeat containing 2, cIAP2 baculoviral IAP repeat containing 3, IKKα inhibitor of nuclear factor
kappa B kinase subunit alpha, NEMO nuclear factor kappa B essential modulator, IKKβ inhibitor of nuclear factor kappa B kinase subunit beta, IκB inhibitor kappa B kinase complex, p50
nuclear factor kappa B subunit 1, p65 nuclear factor kappa B p65 subunit, MAP3K mitogen-activated protein kinase kinase kinase, ERK1/2 extracellular signal-regulated kinase 1/2, JNK
c-Jun N-terminal kinase, p38 p38 mitogen-activated protein kinase, AP-1 activator protein 1, NIK nuclear factor kappa B-inducing kinase, p100 nuclear factor kappa B p100 subunit, RelB
transcription factor RelB, p52 nuclear factor kappa B p52 subunit, MMP-1 interstitial collagenase, MMP-3 stromelysin 1, MMP-13 collagenase 3, ADAM-TS4 a disintegrin and me-
talloproteinase with thrombospondin motifs 4, IL-1β interleukin 1 beta, IL-6 interleukin 6, IL-8 interleukin 8, CCL5 C-C motif chemokine ligand 5, SDF-1 stromal cell-derived factor 1,
VEGF vascular endothelial growth factor, HIF-1α hypoxia inducible factor 1 alpha, HIF-2α hypoxia inducible factor 2 alpha, ACAN aggrecan, COL2 collagen type II; ↑ increase of
expression, ↓ decrease of expression.

78
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

Zhang et al. analysed the levels of IL-1β in subchondral bone de- within 8 h after the exposure of a colony to blood had a reduced effect
rived from femoral heads of patients with HA and RA [62]. The level of on extracellular matrix proteoglycan synthesis. In contrast, TNFαmAb
IL-1β was markedly elevated in the subchondral bone form patients had no significant effect on the synthesis of proteoglycans via inhibiting
with HA compared to the RA patients group. A study by Hooiveld et al. TNFα activity. Moreover, IL-1βmAb reduced the apoptotic activity of
corroborated the negative multidirectional effect of IL-1β on the ar- chondrocytes, whereas TNFαmAb did not. After four days of exposure
ticular cartilage on a study model which imitated intraarticular and adding IL-1RA, the expression of IL-1β and IL-6 was reduced, TNFα
bleeding, and reported a direct influence of IL-1β on the reduction of secretion was unaffected and no influence of TNFαmAb on the ex-
the synthesis of extracellular matrix proteoglycans (PG) of the articular pression of IL-1β was observed. Despite the limitations of an in vitro
cartilage by chondrocytes which was tested with liquid scintillation model, the authors concluded that IL-1β had a key role in the process of
analysis by measuring their rate of sulphate (35SO24−) incorporation cartilage destruction due to BIJD, a role that was not observed with
[63]. Moreover, as regards the pathomechanisms of HA, it was con- TNFα [65].
firmed that the cell membrane of erythrocytes (RBC) was damaged by
lysis, and stimulated monocytes/macrophages (CD14+) to secrete in-
2.2. Tumor necrosis factor alpha (TNFα)
flammatory cytokines, mainly IL-1β independent of inflammation [64].
It was also noted that the impaired secretion of extracellular matrix
2.2.1. Overview of the TNFα properties and its associated signaling
proteoglycans by chondrocytes had a longer lasting effect in case of a
pathways
synergistic activity of IL-1β with lysed RBC than if the factors acted
TNFα is another key inflammatory cytokine, a member of tumor
separately. After four days of the exposure of a colony of human
necrosis factor TNF superfamily with catabolic properties in articular
chondrocytes to isolated IL-1β, isolated lysed RBC and IL-1β combined
cartilage and joint tissues [66,67]. The activity of a protease from a
with lysed RBC, the respective levels of secreted extracellular matrix
disintegrin and metalloproteinase (ADAM) family, namely ADAM me-
proteoglycans were 50%, 37% and 31% of the level in the control
tallopeptidase domain 17 (ADAM17) or TNFα-converting enzyme
group, which was not subjected to the activity of any external factors.
(TACE) triggers the conversion of a homotrimeric transmembrane
Next, the researchers assessed the durability of the impairment of ex-
protein type II (mTNFα) into a soluble TNFα (sTNFα) form [68]. It is
tracellular matrix proteoglycans production by chondrocytes. All three
currently suggested that the signaling pathway associated with
assessed colonies were washed to eliminate pathogenetic factors. After
ADAM17 (TACE) and its regulator known as intramembrane serine
12 days the colonies were again assessed in terms of extracellular ma-
proteases rhomboid 5 homolog 2 (iRhom2) may have a role in HA pa-
trix proteoglycan production and compared to the control group. The
thogenesis [69]. TNFα binds two isotypes of membrane receptors,
result of the colony subjected to IL-1β activity was 90%. In the colony
tumor necrosis factor receptor 1 (TNFR1, TNFRSF1A, CD120a) and
incubated with lysed RBC the level of secretion returned to the level
tumor necrosis factor receptor 2 (TNFR2, TNFRSF1b, CD120b) [70]
observed in the control group. Third colony incubated with IL-1β
(Figs. 4 and 5). Both types of receptors are responsible for the stimu-
combined with lysed RBC indicated the secretion at the level of 56%.
lation of inflammatory and degenerative processes within the articular
Addition of dimethylsulphoxide (DMSO) free radical scavenger, in-
cartilage and other joint tissues; however the activity of TNFR1 re-
creased the production of proteoglycans and the disintegration of the
ceptor, which is sensitive to both forms of TNFα, seems to be sig-
extracellular matrix substances proving that oxidative radicals are in-
nificantly dominant [71]. The activation of TNFR2 receptor also affects
volved in pathological mechanism. A related study was proposed by van
the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/AKT/mam-
Vulpen et al. that analysed the effects of anti-IL-1β monoclonal anti-
malian target of rapamycin (mTOR) angiogenic pathway, independent
body (IL-1βmAb), IL-1RA and anti-TNFα monoclonal antibody
of TNF receptor associated factor 2 (TRAF2) activation, induced with
(TNFαmAb) on the synthesis of extracellular matrix proteoglycans,
angiogenesis, which may be of pathophysiological significance in the
chondrocyte apoptosis and the expression of IL-1β, TNFα and IL-6 in
context of HA and HA-related chronic hypertrophy of the synovial
human colonies of chondrocytes incubated with blood [65]. After four
membrane [72,73].
days of the exposure of the colony to blood, the respective levels of
extracellular matrix proteoglycan secretion dropped by 75% and 76%
compared to the control colony with no blood added. Gradually added 2.2.2. Biological functioning of the TNFα in joint tissues
IL-1βmAb and IL-1RA substantially increased the synthesis to the level The properties of TNFα are related to the inhibition of proteoglycan
of 91% at a dose of 100 ng/mL of IL-1βmAb and 74% at a dose of and COL2 synthesis by chondrocytes [45]. However, the expression of
1000 ng/mL of IL-1RA. A rapid administration of IL-1βmAb and IL-1RA catabolic compounds, such as MMP-1, MMP-3, MMP-13 and ADAM-
TS4, is increased [44]. As regards the promotion of inflammatory

Table 2
Observational studies showing the expression pattern of TNFα in hemophilic arthropathy.

Clinical trial/model Method of sampling Methodology for Reported results/timing Reference


quantification

Patients with HA Synovial tissue samples ELISA Increase of concentration in the supernatants [55]
Patients with HA Synovial tissue samples ELISA Increase of concentration in the supernatants [56]
Cartilage co-cultured with whole blood or Cartilage cultures ELISA Increase of secretion by the cell cultures after [57]
MNC + RBC 4 days of incubation
Cartilage co-cultured with whole blood, MNC, Cartilage cultures ELISA Increase of secretion by the cell cultures after [58]
MNC + RBC or MNC + LPS 4 days of incubation
F8-KO mouse with needle-induced knee joint Synovial fluid samples Luminex xMAP No difference in concentration in the synovial [60]
hemarthrosis fluid
Patients with HA Subchondral bone IHC Increase of concentration in the subchondral [62]
samples bone
Cartilage co-cultured with whole blood Cartilage cultures ELISA Increase of secretion by the cell cultures after [115]
4 days of incubation
Patients with HA Synovial tissue samples ELISA Increase of concentration in the supernatants [131]

HA hemophilic arthropathy, MNC mononuclear cells, RBC red blood cells, LPS lipopolysaccharide, F8-KO F8 gene knock-out, ELISA enzyme-linked immunosorbent assay, IHC im-
munohistochemistry.

79
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

phenomena, TNFα stimulates cells to increase the secretion of IL-6, IL- messenger RNA (mRNA) expression by synoviocytes [80]. Moreover,
8, CCL5, stromal cell-derived factor 1 (SDF-1) and mediators re- TM levels were higher in the synovial fluid of patients with HA
sponsible for the neoangiogenesis of substances like vascular en- (56 ± 25 ng/mL) compared to healthy controls (39 ± 21 ng/mL). In
dothelial growth factor (VEGF), hypoxia inducible factor 1 alpha (HIF- natural conditions TM binds thrombin in 1:1 stoichiometric ratio, then
1α) and hypoxia inducible factor 2 alpha (HIF-2α) [17,46,49,74,75]. activates protein C (PC) [81]. PC is a zymogen which belongs to a group
The regenerative abilities of the articular cartilage are inhibited due to of proteins which inhibit blood clotting via active coagulation factor V
the induction of chondrocyte apoptosis and via the disruption of (FVa) and FVIIIa degradation [81]. HA-related tendency of endothelial
chondrogenic progenitor cells (CPC) migration [46]. In the course of cells, synovial membrane and immune system cells towards an ex-
arthritis, a distinct synergism between the activity of TNFα and IL-1β cessive secretion of inflammatory cytokines, including TNFα, may lead
may be observed [76]. The cytokines activate identical groups of in- to additional disruption of coagulation processes, via an increased se-
tracellular signaling pathways and are secreted by the same types of cretion of TM, and the resultant excessive and inadequate activation of
cells, which is manifested by their increased concentration in the car- PC [80,82]. As regards congenital coagulation disturbances the above
tilage and synovial fluid, subchondral cortical bone layer and synovial cascade of reactions appears to be a potentially aggravating factor of
membrane [31]. the prognosis in patients with HA [82].

2.3. Interleukin 6 (IL-6)


2.2.3. Role of TNFα in the pathogenesis of hemophilic arthropathy
The significant role of TNFα and IL-1β in the pathogenesis of HA is
2.3.1. Overview of the IL-6 properties and its associated signaling pathways
corroborated by several studies (Table 2). Studies by Roosendaal et al.
IL-6 is a multi-functional cytokine composed of 184 amino acid
confirmed that the supernatants of cell cultures of synovial tissue of HA
residues of molecular weight of 21–28 kDa depending on the level of
patients have higher levels of TNFα compared to OA and RA study
glycosylation, with a post-translational four-α-helix bundle structure
groups and control groups, with the reservation that samples including
[83,84] (Fig. 7). The biological activity of IL-6 is mediated through
hemosiderin deposits had the highest level of TNFα, that may be as-
binding to the IL-6 receptor (IL-6R), a type I transmembrane protein
sociated with TNFα-induced TFN-bound iron uptake into synovial
and binding second membrane glycoprotein 130 (mgp130) that serves
macrophages [55,56,59]. Studies by Øvlisen et al. showed no sig-
as a signal transducer of IL-6 [85]. There are two forms of IL-6R such as
nificant differences of TNFα levels in the synovial fluid collected from
membrane IL-6R (mIL-6R) and soluble IL-6R (sIL-6R) [86]. Unlike other
study group and control group mice [60]. The effect of TNFα on ar-
cytokine receptors, both types of IL-6 receptor have activating proper-
ticular cartilage degeneration was shown by Sun et al., who compared
ties [86]. The secretion of IL-6 is mainly induced by IL-1β, TNFα, in-
the influence of isolated FIX and FIX with TNFα receptor antagonist
terferon gamma (IFNγ) and lipopolysaccharides (LPS) [49,87].
(etanercept) in FIX gene (F9) knock-out mice (F9-KO) with knee he-
marthrosis triggered by needle puncture [77,78]. After 3–4 weeks the
degree of destruction of the knees in the FIX and etanercept group was 2.3.2. Biological functioning of the IL-6 in joint tissues
lower than in the group which received FIX substitution and the control Numerous cell types produce IL-6, including: chondrocytes, osteo-
group. The results of the animal study are similar to the study by blasts, fibroblasts, macrophages and mononuclear cells [88]. Arthritis-
Melchiorre et al. and demonstrate a positive clinical impact of admin- associated diseases caused by inflammation, such as OA and RA involve
istration of anti-TNFα antibody (adalimumab) [79]. TNFα also influ- an increased concentration of IL-6 both in the synovial fluid and in the
enced the process of hemostasis occurring directly in joint (Fig. 6). serum, with a positive correlation between clinical signs and the
Dargaud et al. demonstrated that TNFα alone, and in combination with radiographic evidence of the severity of the destructive lesions [88,89].
interleukin 17 (IL-17), in human synovial cells culture from HA pa- IL-6 inhibits the synthesis of COL2 by chondrocytes and increases the
tients, induced a substantial reduction of thrombomodulin (TM) synthesis of MMPs, IL-1β and TNFα by osteoblasts [90,91].

Fig. 6. Schematic overview of the model showing role of TNFα and IL-17 in the context of hemostasis mechanisms occurring directly in joint of patient with hemophilic arthropathy.
Upregulated levels of thrombomodulin and inflammatory cytokines may lead to additional disruption of coagulation processes in the tissues of the affected joint. In case of hemostasis
mechanisms occurring in joint of patient with hemophilic arthropathy, TNFα and IL-17 seem to have a procoagulant activity apart from their direct inflammatory effects. Abbreviations:
TNFα tumor necrosis factor alpha, IL-17 interleukin 17, TM thrombomodulin, PS protein S, C4b-BP complement component 4b binding protein, FIIa thrombin, PC protein C, APC
activated protein C, FVa active coagulation factor V, FVai inactive coagulation factor V, FVIIIa active coagulation factor VIII, A1 A1 domain (1–336 aa), a1 a1 acidic region (337–372 aa),
A2 A2 domain (373–710 aa), a2 a2 acidic region (711–740 aa), A3 A3 domain (1690–2019 aa), C1 C1 domain (2020–2172 aa), C2 C2 domain (2173–2332 aa), FVIIIai inactive
coagulation factor VIII.

80
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

Fig. 7. Schematic overview of the potential cellular and molecular mechanisms by which IL-6 and its associated signaling pathways are involved in expression and functional regulation
of various genes involved in inflammatory and catabolic processes in the pathogenesis of hemophilic arthropathy. Abbreviations: IL-6 interleukin 6, anti-IL-6R antibodies against
interleukin 6 receptor, mIL-6R membrane interleukin 6 receptor, sIL-6R soluble interleukin 6 receptor, mgp130 membrane glycoprotein 130, JAK1 janus kinase 1, JAK2 janus kinase 2,
TYK2 tyrosine kinase 2, MAP3K mitogen-activated protein kinase kinase kinase, ERK1/2 extracellular signal-regulated kinase 1/2, JNK c-Jun N-terminal kinase, p38 p38 mitogen-
activated protein kinase, AP-1 activator protein 1, PI3K phosphatidylinositol-4,5-bisphosphate 3-kinase, AKT protein kinase B, mTOR mechanistic target of rapamycin, STAT3 signal
transducer and activator of transcription 3, P phosphate, MMP-1 interstitial collagenase, MMP-3 stromelysin 1, MMP-13 collagenase 3, ADAM-TS4 a disintegrin and metalloproteinase
with thrombospondin motifs 4, IL-1β interleukin 1 beta, TNFα tumor necrosis factor alpha, IL-6 interleukin 6, IL-8 interleukin 8, CCL5 C-C motif chemokine ligand 5, PLA2 phospholipase
A2, COX-2 cyclooxygenase 2, PGE2 prostaglandin E2, ACAN aggrecan, COL2 collagen type II; ↑ increase of expression, ↓ decrease of expression.

Additionally, IL-6 promotes the development of osteoclast formations administration of IL-6 had chondroprotective properties and reduced
which stimulates the process of osseous tissue resorption, especially in the loss of proteoglycans during the acute stage of inflammation. Fur-
the subchondral layer [92]. However, some research questions the in- thermore, chronic administration of IL-6 produced an increased ten-
flammatory role of IL-6 as regards the articular cartilage and periarti- dency towards osteophyte formation [95]. Seemingly, there is an opi-
cular tissues [93]. Mice which lacked the gene coding IL-6 (IL-6−/−) nion that IL-6 has a tendency towards regulating immunological
demonstrated a tendency towards developing much more advanced processes or may show anti-inflammatory properties at the physiolo-
degenerative changes in joints compared to wild-type (WT) mice [94]. gical level or during acute inflammation [96]. However, in case of a
Another study on IL-6−/− mice showed that intraarticular chronic inflammatory process, the biological activity of IL-6 turns into

81
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

an inflammatory one [97]. Altogether these results indicate that IL-6 development of HA, at least at the initial stage of the inflammation, in
mediates anti-inflammatory properties during acute inflammation, but which no significant activity of lymphocytes T CD4+ is observed [60].
inflammatory activity during chronic inflammation [96,97].

2.3.3. Role of IL-6 in the pathogenesis of hemophilic arthropathy 3. Anti-inflammatory cytokines


The inflammatory role of IL-6 seems to be confirmed in the patho-
genesis of HA (Table 3). Just like in case of IL-1β and TNFα, the su- Anti-inflammatory cytokines are associated with the regulation and
pernatants of cell cultures of synovial tissue in HA patients have higher modulation of the immune response in two ways: they inhibit the
levels of IL-6 compared to OA and RA study groups and control groups synthesis of inflammatory cytokines, which indirectly prevents cata-
[55,56,59]. Øvlisen et al. reported a significantly higher level of IL-6 in bolic processes and they possess a direct anabolic activity on articular
the synovial fluid in F8-KO mice (174 ± 319 pg/mL) than in healthy structures via the stimulation of the secretion of growth factors and
mice and those which received the substitution of rFVIII [60]. Ad- selective regulatory proteins [102,103]. In the large group of anti-in-
ditionally, the levels of IL-6 in the synovial fluid correlated with post flammatory cytokines interleukin 4 (IL-4) and interleukin 10 (IL-10)
mortem examination showing a more marked distension of the joint have been the best analysed in the context of HA pathogenesis.
capsule and hemarthrosis. In the study of Narkbunnam et al. analysed
the effect of MR16-1, a rat monoclonal immunoglobulin G1 (IgG1) 3.1. Interleukin 4 (IL-4)
antibody directed against mouse IL-6R (anti-IL-6R) on the knees of F8-
KO mouse with hemarthrosis [98]. The administration of isolated 3.1.1. Overview of the IL-4 properties and its associated signaling pathways
MR16-1 insignificantly reduced the proliferation of the synovial mem- IL-4 is a glycoprotein composed of 129 amino acid residues of
brane, the presence of neovascularization, hemosiderin deposits, mac- molecular weight of 15–20 kDa depending on the level of glycosylation,
rophage infiltration and articular cartilage erosion compared to the with a post-translational four-α-helix bundle structure, additionally
control group. Interestingly, the best anti-inflammatory activity to- stabilized by the presence of three disulfide bonds [104]. There are two
wards joints was observed in case of a substitution with MR16-1 com- types of membrane receptors binding IL-4 (IL-4R) forming heterodimers
bined with FVIII. A 100% survival rate was observed until the last day which are structurally a combination of two of the three receptor chains
of the study in F8-KO mice which received the combined therapy. It is [105]. Type I signaling complex contributes to the transfer of in-
noteworthy that supplementation with FVIII alone did not have such an tracellular signal via gradual phosphorylation of IL-4Rα/JAK1/STAT3/
anti-inflammatory effectiveness as the combination of MR16-1 and STAT6 cascade [106,107]. The activation of type II signaling complex is
FVIII. It may indicate that in early stage of HA, the role of IL-6 is not so associated with triggering IL-4Rα/JAK2/STAT3 cascade and also IL-
important as regards the development of destructive lesions in the joint. 13Rα1/TYK2/STAT1/STAT6 cascade in case of binding the second type
It confirms that the administration of isolated MR16-1 only slightly of receptor with IL-13 [106,107]. There is also a soluble form of a re-
inhibited degenerative processes. IL-6 seemed rather to specifically ceptor for IL-4 (sIL-4R), which binds and inhibits the biological activity
strengthen immune processes which is confirmed by the observed high of IL-4 [108].
chondroprotective effectiveness of the combined substitution of MR16-
1 and FVIII.
3.1.2. Biological functioning of the IL-4 in joint tissues
2.4. Other inflammatory cytokines IL-4 is mainly produced by Th2 lymphocytes which infiltrate the
synovial membrane of the joint via blood vessels [109]. Mast cells,
Other inflammatory cytokines which participate in the pathogenesis basophils and eosinophils also have the ability to express and secrete IL-
of HA include chemokine C-C motif ligand 2 (CCL2, MCP-1) and che- 4 [110]. IL-4 is a cytokine with strong chondroprotective properties,
mokine C-X-C motif ligand 1 (CXCL1, KC) [60]. Their level in the sy- that blocks the degradation of proteoglycans of articular cartilage via
novial fluid in mouse knees was significantly elevated in F8-KO mice inhibiting the secretion of MMPs in patients at an advanced stage of OA
compared to healthy controls. CCL2 mediates monocyte and macro- [42]. IL-4 also inhibits the synthesis of inflammatory mediators such as
phage migration, and their infiltration to the site of inflammation [99]. PGE2, COX-2, PLA2 and iNOS via chondrocytes and fibroblasts
CXCL1 is a strong chemoattractant for neutrophils, which potentially [42,111,112]. Moreover, fibroblast and chondrocyte cultures exposed
contributes to the promotion of the immune response and secretion of to IL-4 showed a reduction in the expression of inflammatory cytokines
numerous inflammatory mediators including IL-1β, TNFα and IL-6 by such as IL-1β, TNFα and IL-6 [113]. Fu et al. studied mice with col-
neutrophils and other cell types [100]. CXCL1 is an important cytokine lagen-induced arthritis which received recombinant murine IL-4 (rmIL-
that influences the severity of an inflammatory process in injuries with 4) with recombinant murine GM-CSF (rmGM-CSF) [114] and reported a
bleeding. Blocking its activity before the procedure of an artificially significant reduction in inflammation of the synovial membrane and
stimulated bleeding in a mouse with anti-CXCL1 resulted in reduction articular cartilage. Macrophages sampled from these mice showed a
of neutrophil infiltration to liver and lung tissues and diminished the diminished ability to express IL-1β and TNFα compared to the control
oedema of these organs compared to the control group [101]. However, group. The effect of rmIL-4 with rmGM-CSF was also manifested by the
there is a scarcity of studies concerning other inflammatory cytokines reduced proliferation of Th17 lymphocytes with the reduced in-
which might have a role in the pathogenesis of HA. Individual studies flammatory synthesis of IL-17 and increased secretion of IL-10 and
have not confirmed any significant effect of IL-17 or CCL5 on the transforming growth factor beta (TGF-β) by regulatory T (Treg) cells.

Table 3
Observational studies showing the expression pattern of IL-6 in hemophilic arthropathy.

Clinical trial/model Method of sampling Methodology for quantification Reported results/timing Reference

Patients with HA Synovial tissue samples ELISA Increase of concentration in the supernatants [55]
Patients with HA Synovial tissue samples ELISA Increase of concentration in the supernatants [56]
F8-KO mouse with needle-induced knee joint Synovial fluid samples Luminex xMAP Increase of concentration in the synovial fluid [60]
hemarthrosis

HA hemophilic arthropathy, F8-KO F8 gene knock-out, ELISA enzyme-linked immunosorbent assay.

82
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

3.1.3. Role of IL-4 in the pathogenesis of hemophilic arthropathy

Reference
Studies on HA pathogenesis confirm the anti-inflammatory and

[115]

[118]
[131]
[132]

[133]
chondroprotective role of IL-4, as well as a decrease in disease devel-
opment (Table 4). In the study of van Meegeren et al. analysed the

Prevent of blood-induced cartilage damage

Prevent of blood-induced cartilage damage


Prevent of blood-induced cartilage damage
Prevent of blood-induced cartilage damage

Prevent of blood-induced cartilage damage


effect of single IL-4, IL-10, and IL-4 combined with IL-10 on human
chondrocyte cultures, exposed to blood for four days, to mimic the
natural evacuation time of blood from the joint [115,116]. IL-4 treat-
ment increased proteoglycan production by chondrocytes to complete
normalization. The combination of IL-4 and IL-10 (10 ng/mL) in a dose-
dependent manner further increased the normalization of proteoglycan
secretion. Moreover, IL-4 reduced the expression of IL-1β and TNFα,
and also markedly inhibited blood-induced apoptosis of chondrocytes.

Reported results
Additionally, some samples of cartilage which had been exposed to
blood showed the increased expression of mRNA for IL-4 receptor
subunit alpha (IL-4Rα) and interleukin 2 (IL-2) receptor common

IL-4 interleukin 4, IL-10 interleukin 10, IL-1β interleukin 1 beta, TNFα tumor necrosis factor alpha, IL-6 interleukin 6; ↑ increase in mentioned variable, ↓ decrease in mentioned variable.
gamma (IL-2Rγc) chains [42] (Fig. 8). The another study in F8-KO mice
administering IL-4 and IL-10 demonstrated a slower progression of ar-
ticular cartilage destruction compared to the control group. Similar

IL-6









results were obtained when the analysis included canine and human

Effect on production
cartilage, with a marked reduction in the production of IL-1β by human

TNFα
chondrocytes after adding IL-4 combined with IL-10 [118]. Optimal
chondroprotective activity and potential BIJD prevention occurred with




early administration of anti-inflammatory cytokines, preferably im-
mediately after hemarthrosis occurred and no later than 4–8 h after an

IL-1β
artificially induced injury [115,117,118]. Some authors also suggested














the role of IL-4 in decreasing VEGF expression in the course of HA

Effect on chondrocyte apoptosis


[119]. It may be due to an intermediate activity of inhibiting the se-
cretion of IL-1β and TNFα which are indirectly responsible for in-
creased expression of VEGF [98,115,117–119]. Thanks to the limitation
of the pathological growth and permeability of blood vessels IL-4 and
other anti-inflammatory cytokines may contribute to the protection and
reduction of inflammatory and degenerative processes in the course of
HA [119]. Ravanbod et al. demonstrated that IL-4 deficiency enhanced
the tendency towards the development of destructive lesions in the


joints of HA patients [120]. A therapeutic effect of mesenchymal stem


cells (MSC) on rabbit joints with hemarthrosis were analyzed. The re-
duced degenerative manifestations were accompanied with the in-
Observational studies implicating the role of IL-4 and IL-10 in the pathogenesis of hemophilic arthropathy.

Effect on proteoglycan turnover

creased expression of IL-4 and simultaneous reduction in the level of the


Release

expression of IL-1β and TNFα in the synovial membrane of study group


animals. It may indicate that maintaining healthy joints requires the













constant presence of IL-4, and lowering its expression may be associated
with the progression of inflammatory changes related to BIJD. In this
case the use of MSC appears to be a promising therapeutic option in HA
Synthesis

patients [121]. Interestingly, presence of anti-FVIII in blood also in-


fluences the anti-inflammatory activity dependent on IL-4. Silveira












et al. reported changes in the activity of IL-4-positive cells in the per-


ipheral blood of patients with anti-FVIII [122]. These patients had a
and synovial tissue co-cultured with whole blood and IL-10

decreased level of IL-4 in CD4+ and CD8+ T cells compared to analo-


gous cells isolated from the peripheral blood of HA patients who lacked
co-cultured with whole blood and IL4-10 synerkine

co-cultured with whole blood and IL4-10 synerkine

anti-FVIII. Similar results are observed as regards B cells. Moreover, it


co-cultured with whole blood and IL-4 + IL-10
co-cultured with whole blood and IL-4 + IL-10

co-cultured with whole blood and IL-4 + IL-10

co-cultured with whole blood and IL-4 + IL-10

was demonstrated that the presence of anti-FVIII is associated with


decreased frequencies of IL-4-positive neutrophils. Some researchers
co-cultured with whole blood and IL-10

co-cultured with whole blood and IL-10

co-cultured with whole blood and IL-10


co-cultured with whole blood and IL-4

co-cultured with whole blood and IL-4

co-cultured with whole blood and IL-4

suggest that there is a relationship between level of anti-FVIII secretion


and an anti-inflammatory activity and regulatory pattern of IL-4
[123,124]. The study showed a correlation between the presence of
anti-FVIII and the reduced expression of IL-4, which may provide a
rationale for deteriorated prognosis in HA patients [122].

3.2. Interleukin 10 (IL-10)


Clinical trial/model

3.2.1. Overview of the IL-10 properties and its associated signaling


pathways
IL-10 is a homodimer of the total molecular weight of 37 kDa, built
Cartilage
Cartilage
Cartilage
Cartilage
Cartilage
Cartilage
Cartilage
Cartilage
Cartilage
Cartilage
Cartilage
Cartilage
Cartilage

of monomers composed of 160 amino acid residues of the molecular


Table 4

weight of 18.5 kDa, after post-translational modifications the homo-


dimer becomes a six-α-helix bundle structure [125,126]. The receptor

83
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

Fig. 8. Schematic overview of the potential cellular and molecular mechanisms by which IL-4 and IL-13 signaling complexes are involved in expression and functional regulation of
various genes involved in anti-inflammatory and anabolic processes in the pathogenesis of hemophilic arthropathy. Abbreviations: IL-4 interleukin 4, IL-13 interleukin 13, IL-4Rα
interleukin 4 receptor subunit alpha, IL-2Rγc interleukin 2 receptor common gamma, IL-13Rα1 interleukin 13 receptor subunit alpha 1, IL-13Rα2 interleukin 13 receptor subunit alpha 2,
JAK1 janus kinase 1, JAK3 janus kinase 3, STAT6 signal transducer and activator of transcription 6, P phosphate, TYK2 tyrosine kinase 2, STAT3 signal transducer and activator of
transcription 3, MMPs metalloproteinases, IL-1β interleukin 1 beta, TNFα tumor necrosis factor alpha, IL-6 interleukin 6, PGE2 prostaglandin E2, COX-2 cyclooxygenase 2, PLA2
phospholipase A2, iNOS inducible nitric oxide synthase, VEGF vascular endothelial growth factor, ACAN aggrecan, COL2 collagen type II; ↑ increase of expression, ↓ decrease of
expression.

for IL-10 (IL-10R) is a heterodimer composed of two chains such as IL- 3.2.2. Biological functioning of the IL-10 in joint tissues
10 receptor subunit 1 (IL-10R1) and IL-10 receptor subunit 2 (IL-10R2) IL-10 has multifunctional protective and anti-inflammatory prop-
[126]. The immune system cells which may secrete IL-10 include CD4+ erties as regards articular cartilage cells and periarticular tissues [42]
T lymphocytes (Th1, Th2, Th17, Treg), CD8+ T lymphocytes, B lym- (Fig. 9). IL-10 increases the synthesis of COL2 and ACAN by chon-
phocytes, monocytes, macrophages, myeloid dendritic cells (mDC), drocytes and also leads to an intensified general proteoglycan synthesis
eosinophils and mast cells [127]. Moreover, numerous other cell types and their increased percentage proportion in the extracellular matrix in
are capable of secreting IL-10, which makes them able to take part in in vitro conditions [42]. Just like IL-4, IL-10 exerts an inhibiting effect
the regulation of inflammatory response, such as keratinocytes, epi- on chondrocyte apoptosis and MMPs production [42]. These processes
thelial cells or fibroblasts [128]. are also influenced by triggering IL-1RA expression, a possible me-
chanism stimulated by IL-10, and tissue inhibitor of metallopeptidase 1
(TIMP-1) [129]. IL-10 is also antagonistic to TNFα activity towards

84
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

Fig. 9. Schematic overview of the potential cellular and molecular mechanisms by which IL-10 and its associated signaling pathways are involved in expression and functional regulation
of various genes involved in anti-inflammatory and anabolic processes in the pathogenesis of hemophilic arthropathy. Abbreviations: IL-10 interleukin 10, IL-10R1 interleukin 10 receptor
subunit 1, IL-10R2 interleukin 10 receptor subunit 2, JAK1 janus kinase 1, TYK2 tyrosine kinase 2, STAT3 signal transducer and activator of transcription 3, P phosphate, MMPs
metalloproteinases, TIMP-1 tissue inhibitor of metallopeptidase 1, IL-1RA interleukin 1 receptor antagonist, TNFα tumor necrosis factor alpha, IL-4Rα interleukin 4 receptor subunit
alpha, PLA2 phospholipase A2, COX-2 cyclooxygenase 2, PGE2 prostaglandin E2, BMP-2 bone morphogenetic protein 2, BMP-6 bone morphogenetic protein 6, ACAN aggrecan, COL2
collagen type II; ↑ increase of expression, ↓ decrease of expression.

fibroblasts of the synovial membrane via inhibiting the secretion of production of proteoglycans and decreased their degradation after IL-
PGE2, COX-2 and PLA2 [111]. Moreover, IL-10 impedes binding TNFα 10 administration [131]. IL-10 also reduced the expression of IL-1β and
to specific receptors via inhibiting their expression on the surface of TNFα in culture supernatants. Van Meegeren et al. confirmed these
fibroblasts [111]. It was also demonstrated that IL-10 induces the ex- observations, but showed that IL-4 had stronger anti-inflammatory
pression of numerous growth factors, including bone morphogenetic properties compared to IL-10 [115]. As mentioned before, IL-10 showed
protein 2 (BMP-2) and bone morphogenetic protein 6 (BMP-6) which the strongest protective activity when combined with IL-4 [117,118].
belong to TGF-β family and are responsible for chondrogenesis reg- From these results, van Meegeren and van Vulpen et al. analysed the
ulation involving an appropriate transformation of mesenchymal cells effect of fusion protein consisting IL-4 and IL-10 (IL4-10 synerkine) on
into chondrocytes [130]. IL-10 is viewed as a well-known cytokine with human chondrocyte cultures [132,133]. The IL4-10 synerkine pre-
protective and anti-inflammatory properties towards joints in the vented blood-induced cartilage damage similar to the combination of
course of OA and RA [42]. the individual components in a dose-dependent manner
(0.0001–100 ng/mL), and the use of IL4-10 synerkine also appeared to
3.2.3. Role of IL-10 in the pathogenesis of hemophilic arthropathy be a potent therapeutic option in HA patients. Interestingly, Silveira
Numerous studies have investigated the role of IL-10 in the patho- et al. demonstrated that the patients with hemophilia who developed
genesis of HA and its possible therapeutic activity (Table 4). Jansen anti-FVIII had increased frequencies of IL-10-positive neutrophils, B and
et al. demonstrated that chondrocyte and synovial membrane cell cul- T cells [122]. It may be associated with polymorphism in the IL-10
tures collected from patients with advanced HA lesions increased the gene. Apparently, a polymorphism in the IL-10 gene (134 allele in

85
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

cytosine-adenine repeat microsatellites of the IL-10 promoter) directly not be viewed in some cases as fully effective and satisfactory [143].
carries a significantly higher risk of anti-FVIII development [124]. IL-10 The use of prophylactic FRT is associated with the risk of inhibitor
is known to stimulate plasma cells to secrete numerous antibodies and development which considerably impedes the prophylaxis of articular
cytokines. Presence of allele 134 markedly increases this tendency. lesions [144]. At the same time, orthopaedic procedures, due to their
Most probably, the presence of this polymorphism in IL-10 gene con- specificity, carry a high perioperative risk and if conducted at a young
tributes to an intensified and permanent secretion of IL-10, and also age, they do not provide a long-term solution to the problem, but only a
anti-FVIII in hemophilia patients. Interestingly, polymorphisms in IL-1β short-term relief, depending on the severity of the disease [143].
and IL-4 genes demonstrated no correlation with a higher risk of anti- Therefore, it is important to continue the search for new sustainable
FVIII development [124]. Therefore, it needs to be emphasized that therapies which would help prevent the development of the disease and
although IL-10 has a protective role in the pathogenesis of HA as re- avoid the surgical treatment of patients with HA [145]. The regulation
gards the joint and periarticular tissues, its overexpression propably and modulation of relationships occurring within the cytokine network
results in anti-FVIII development, which may ultimately lead to prog- are considered to be very promising directions in such therapy. In order
nosis deterioration and a considerable limitation of therapeutic options. to achieve this goal it is necessary to determine the role of specific
cytokines in the immunopathological mechanisms involved in HA.
3.3. Other anti-inflammatory cytokines The present paper presents the current knowledge on inflammatory
and anti-inflammatory cytokines involved in the pathogenesis of HA.
Interleukin 11 (IL-11) deserves a particular attention as regards the The cytokines mainly associated with NF-κB signaling pathway are key
remaining anti-inflammatory cytokines [134]. No studies have analysed cytokines responsible for the clinical presentation of HA with a proven
the direct effect of IL-11 on joints in the course of HA. However, it has inflammatory and catabolic activity associated with BIJD development
been proved to have an effect on the increase of FVIII level in the blood [146]. The inflammatory cytokines whose activity has been very well
serum, Ragni et al. stated that the subcutaneous administration of re- elucidated are ones described in this paper such as IL-1β, TNFα, IL-6,
combinant human IL-11 (rhIL-11) is an effective and safe alternative to CCL2 and CXCL1. As regards the articular cartilage cells they affect the
desmopressin (DDAVP) administration [135]. Patients with mild and disintegration and reduction in the synthesis of extracellular matrix,
moderate forms of hemophilia A and von Willebrand disease (vWD) including COL2 and ACAN, they contribute to chondrocyte apoptosis.
who received 25 μg/kg of rhIL-11 subcutaneously for four subsequent They are also responsible for forming free radicals and promoting the
days demonstrated approximately 1.5–2.0-fold higher levels of FVIII in synthesis of catabolic and inflammatory factors, such as MMP-1, MMP-
the serum on the last day of the study compared to the respective 3, MMP-13, ADAM-TS4, PLA2, COX-2, PGE2, iNOS and, reciprocally,
baseline levels. According to the authors it resulted in the reduction of the synthesis of other cytokines. They also increase VEGF and TM se-
blood loss during menstruation and made the conducted surgeries safer. cretion. VEGF is responsible for the growth of endothelium in the sy-
Similar results had been obtained previously on a mouse model by novial membrane affected with an inflammation, while TM contributes
Denis et al. [136]. The expression of IL-11 in joints and periarticular to the activation of PC, a factor which degrades FVa and FVIIIa. Their
tissues has not been studied more precisely in patients with HA. The activity also manifests itself via inhibiting anti-inflammatory cytokine
data might be useful in the determination of the exact role of this cy- synthesis by numerous cell types, especially immune system cells and
tokine in the pathophysiology of HA. TGF-β and bone morphogenetic synovial membrane cells. Inflammatory cytokines may also disturb the
protein (BMP) family are indicated to be promising directions in the differentiation of progenitor cells into chondroblasts and osteoblasts
treatment of intraarticular bleeding in the course of HA [78]. They have and promote osteoclastogenesis. The reciprocal stimulation of synthesis
an anti-inflammatory and anabolic effect towards tissues which form leads to a broadened, frequently complementary catabolic activity of
the joint [137]. Until recently, their significance was demonstrated in inflammatory cytokines, which causes the development of a full-blown
non-hemophilia subjects in the treatment of bone union disorders and form of HA in a short period of time.
promotion of differentiation of articular cartilage cells [78,138]. It may On the other hand, the main anti-inflammatory cytokines which
be an interesting direction in the treatment of BIJD patients, however, it play a role in the pathogenesis of HA are IL-4 and IL-10. They inhibit
still needs to be experimentally developed. The anti-inflammatory role the degeneration of proteoglycans and the synthesis of substances with
of IL-13 should not be disregarded in HA pathophysiology [139]. It a strong inflammatory potential such as MMPs, PGE2, COX-2, PLA2,
occurs via the possibility of activating type II signaling complex which iNOS by chondrocytes and fibroblasts. They inhibit the synthesis of
triggers IL-13-specific IL-13Rα1/TYK2/STAT1/STAT6 cascade, which inflammatory cytokines and prevent blood-induced chondrocyte
is associated with anti-inflammatory and chondroprotective activity in apoptosis. They also reduce VEGF synthesis, therefore, the pathological
various arthritis-associated diseases [106] (Fig. 8). neovascularization within the synovial membrane may be limited.
Ravanbod et al. reported that the treatment of a typical HA-related
4. Summary and future perspectives deficit of anti-inflammatory cytokines with MSC led to a considerable
reduction in IL-β synthesis and inhibited the progression of degen-
The pathophysiology of HA has not been fully elucidated, especially erative lesions occurring after hemarthrosis on an animal model [120].
in terms of mechanisms responsible for the development of an in- The HA treatment with anti-inflamamtory cytokines and growth factors
flammation and catabolic processes in the joint. It is believed that cy- seems also to be promising direction. Growth factors such as TGF-β and
tokines play a key role in promoting these phenomena, as they act as a ligands from BMP family may prove helpful in the regeneration of joints
part of a complex system known as cytokine network. It is suggested and periarticular tissues in patients with HA. However, we cannot rule
that the development of an inflammation is due to the imbalance be- out their possible negative effect, such as on the development of neo-
tween the biological activity of inflammatory and anti-inflammatory plastic lesions, the stimulation of angiogenesis and synovial membrane
cytokines [140]. The main factor which triggers and causes the im- cell proliferation [147]. Additionally, the treatment with IL-10 may
balance is blood and its metabolites appearing in the joint. Recurrent contribute to the production of anti-FVIII in patients with hemophilia
joint bleeding episodes strengthen the tendency towards promoting via the stimulation of plasma cells to produce numerous types of anti-
inflammatory phenomena, which causes a gradual degeneration of bodies [124,133]. At the same time, the prolonged exposure of tissues
joints and periarticular tissues. Consequently, the patients are con- to IL-4 activity may potentially lead to the activation of macrophages
siderably impaired and the mortality rate increases [141]. The present and increased erythrophagocytosis in the liver, spleen and bone marrow
treatment methods mainly involve the use of prophylactic FRT in- intensifying the general inflammatory response [148]. But in most
cluding new bypassing agents, and in advanced cases – the use of highly cases, the supplementation of IL-4 or IL-10 directly or indirectly via
specialized orthopaedic procedures [142]. However, the methods may MSC appears to be rather safe. Taking into consideration these facts the

86
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

use of IL4-10 synerkine appears to be the most promising therapeutic Molecular models of the procoagulant factor VIIIa-factor IXa complex, J. Thromb.
option due to pharmacokinetics and bioactivity properties [132]. Time Haemost. 3 (2005) 2044–2056, http://dx.doi.org/10.1111/j.1538-7836.2005.
01527.x.
is also an important therapeutic criterion. The sooner anti-in- [10] J.P. Sheehan, H.C. Lan, Phosphorothioate oligonucleotides inhibit the intrinsic
flammatory cytokines are administered to the injured joint after tenase complex, Blood 92 (1998) 1617–1625.
bleeding occurred, the better the chondroprotective effect. Another [11] J.H. Lawson, K.G. Mann, Cooperative activation of human factor IX by the human
extrinsic pathway of blood coagulation, J. Biol. Chem. 266 (1991) 11317–11327.
promising treatment method of lesions typical of HA is the use of an- [12] M.F. Hockin, K.C. Jones, S.J. Everse, K.G. Mann, A model for the stoichiometric
tibodies against inflammatory cytokines, such as etanercept and adali- regulation of blood coagulation, J. Biol. Chem. 277 (2002) 18322–18333, http://
mumab, which block the biological activity of TNFα. Preliminary re- dx.doi.org/10.1074/jbc.M201173200.
[13] G.C. White 2nd, F. Rosendaal, L.M. Aledort, J.M. Lusher, C. Rothschild,
sults of their effectiveness in the course of HA treatment appear to be J. Ingerslev, Factor VIII and Factor IX Subcommittee. Definitions in hemophilia.
promising [77,79]. Novel approaches, currently under investigation Recommendation of the scientific subcommittee on factor VIII and factor IX of the
(phase 4), include targeted agents such as bevacizumab (anti-VEGF) scientific and standardization committee of the International Society on
Thrombosis and Haemostasis, Thromb. Haemost. 85 (2001) 560.
used in intraarticular injections (NCT02060305) which seem to be a
[14] H.M. van den Berg, P.H. De Groot, K. Fischer, Phenotypic heterogeneity in severe
potential promising direction of effective therapies for HA patients with haemophilia, J. Thromb. Haemost. 5 (Suppl. 1) (2007) 151–156, http://dx.doi.
chronic proliferative synovitis [74]. Such conclusions may not be org/10.1111/j.1538-7836.2007.02503.x.
viewed as final due to insufficient research conducted on the effect of [15] A. Srivastava, A.K. Brewer, E.P. Mauser-Bunschoten, N.S. Key, S. Kitchen,
A. Llinas, Treatment Guidelines Working Group on Behalf of The World Federation
inflammatory and anti-inflammatory cytokines on immunopathological of Hemophilia, Guidelines for the management of haemophilia, Haemophilia 19
mechanisms involved in HA. Moreover, numerous cytokines and pro- (1) (2013) e1–47, http://dx.doi.org/10.1111/j.1365-2516.2012.02909.x.
teins related to the pathomechanism of HA have not been studied yet. [16] E.C. Rodríguez-Merchán, Musculoskeletal complications of haemophilia, HSS J. 6
(2010) 37–42, http://dx.doi.org/10.1007/s11420-009-9140-9.
This group includes progranulin (PGRN), iRhom2, IL-17, CCL5, IL-11, [17] L.A. Valentino, Blood-induced joint disease: the pathophysiology of hemophilic
IL-13 and especially CX3CL1 (fractalkine). Apart from chemotactic arthropathy, J. Thromb. Haemost. 8 (2010) 1895–1902, http://dx.doi.org/10.
properties typical of chemokines, CX3CL1 has the ability to induce 1111/j.1538-7836.2010.03962.x.
[18] F. König, Die Gelenkerkrankungen bei Blutern mit besonderer Berücksichtigung
leukocyte migration through the vascular endothelium independent of der Diagnose, Klin Vorträge NF 36 (1892) 233–243.
selectins and integrins [149]. Therefore, leukocytes which have [19] M.C. Swanton, Hemophilic arthropathy in dogs, Lab. Invest. 8 (1959) 1269–1277.
CX3CR1 receptor on the surface are able to pass through the vascular [20] A. Gringeri, B. Ewenstein, A. Reininger, The burden of bleeding in haemophilia: is
one bleed too many? Haemophilia 20 (2014) 459–463, http://dx.doi.org/10.
wall to the site of inflammation in an alternative way. Overexpression 1111/hae.12375.
of CX3CL1 results with hypertrophy of the synovial membrane and the [21] K. Fischer, J.G. van der Bom, G. Mauser-Bunschoten, R. Prejs, P. de Kleijn, et al.,
proliferation of vascular endothelium in many joint diseases [150]. The effects of postponing prophylactic treatment on long-term outcome in patients
with severe haemophilia, Blood 99 (2002) 2337–2341, http://dx.doi.org/10.
Therefore, CX3CL1 impact on the development of HA need to be par-
1182/blood.V99.7.2337.
ticularly investigated. [22] W. Schramm, A. Gringeri, R. Ljung, K. Berger, A. Crispin, M. Bullinger, ESCHQOL
Exploring the role of cytokine network in HA seems to be justified, Study Group, et al., Haemophilia care in Europe: the ESCHQoL study, Haemophilia
as it shifts the center of gravity from symptomatic to causative treat- 18 (2012) 729–737, http://dx.doi.org/10.1111/j.1365-2516.2012.02847.x.
[23] E.P. Mauser-Bunschoten, D.E. Fransen Van De Putte, R.E. Schutgens, Co-morbidity
ment. It may result in a shorter duration of treatment, reduction in the in the ageing haemophilia patient: the down side of increased life expectancy,
general treatment costs. Most importantly, it may lower mortality rates Haemophilia 15 (2009) 853–863, http://dx.doi.org/10.1111/j.1365-2516.2009.
and cause a long-lasting improvement of the quality of life in patients 01987.x.
[24] M. Morfini, S. Haya, G. Tagariello, H. Pollmann, M. Quintana, B. Siegmund, et al.,
with HA. European study on orthopaedic status of haemophilia patients with inhibitors,
Haemophilia 13 (2007) 606–612, http://dx.doi.org/10.1111/j.1365-2516.2007.
Conflict of interest 01518.x.
[25] A. Srivastava, Inflammation is key to hemophilic arthropathy, Blood 126 (2015)
2175–2176, http://dx.doi.org/10.1182/blood-2015-09-665091.
None. [26] D. De Groote, P.F. Zangerle, Y. Gevaert, M.F. Fassotte, Y. Beguin, F. Noizat-
Pirenne, et al., Direct stimulation of cytokines (IL-1 beta, TNF-alpha, IL-6, IL-2,
IFN-gamma and GM-CSF) in whole blood. I. Comparison with isolated PBMC sti-
References
mulation, Cytokine 4 (1992) 239–248, http://dx.doi.org/10.1016/1043-4666(92)
90062-V.
[1] F. Peyvandi, I. Garagiola, G. Young, The past and future of haemophilia: diagnosis, [27] L.F. van Vulpen, G. Roosendaal, B.S. van Asbeck, S.C. Mastbergen, F.P. Lafeber,
treatments, and its complications, Lancet 388 (2016) 187–197, http://dx.doi.org/ R.E. Schutgens, The detrimental effects of iron on the joint: a comparison between
10.1016/S0140-6736(15)01123-X. haemochromatosis and haemophilia, J. Clin. Pathol. 68 (2015) 592–600, http://
[2] F. Peyvandi, G. Jayandharan, M. Chandy, A. Srivastava, S.M. Nakaya, dx.doi.org/10.1136/jclinpath-2015-202967.
M.J. Johnson, et al., Genetic diagnosis of haemophilia and other inherited [28] P. Aisen, C. Enns, M. Wessling-Resnick, Chemistry and biology of eukaryotic iron
bleeding disorders, Haemophilia 12 (Suppl. 3) (2006) 82–89, http://dx.doi.org/ metabolism, Int. J. Biochem. Cell Biol. 33 (2001) 940–959, http://dx.doi.org/10.
10.1111/j.1365-2516.2006.01263.x. 1016/S1357-2725(01)00063-2.
[3] J. Graw, H.H. Brackmann, J. Oldenburg, R. Schneppenheim, M. Spannagl, [29] H. Birkedal-Hansen, Role of cytokines and inflammatory mediators in tissue de-
R. Schwaab, Haemophilia A: from mutation analysis to new therapies, Nat. Rev. struction, J. Periodontal Res. 28 (1993) 500–510, http://dx.doi.org/10.1111/j.
Genet. 6 (2005) 488–501, http://dx.doi.org/10.1038/nrg1617. 1600-0765.1993.tb02113.
[4] M. Cavazza, Y. Kodra, P. Armeni, M. De Santis, J. López-Bastida, R. Linertová, [30] F.R. Balkwill, F. Burke, The cytokine network, Immunol. Today 10 (1989)
et al., BURQOL-RD research network. Social/economic costs and quality of life in 299–304, http://dx.doi.org/10.1016/0167-5699(89)90085-6.
patients with haemophilia in Europe, Eur. J. Health Econ. 17 (Suppl. 1) (2016) [31] X. Chevalier, F. Eymard, P. Richette, Biologic agents in osteoarthritis: hopes and
53–65, http://dx.doi.org/10.1007/s10198-016-0785-2. disappointments, Nat. Rev. Rheumatol. 9 (2013) 400–410, http://dx.doi.org/10.
[5] J.M. Soucie, B. Evatt, D. Jackson, Occurrence of hemophilia in the United States. 1038/nrrheum.2013.44.
The Hemophilia surveillance system project investigators, Am. J. Hematol. 59 [32] I.B. McInnes, C.D. Buckley, J.D. Isaacs, Cytokines in rheumatoid arthritis – shaping
(1998) 288–294, http://dx.doi.org/10.1002/(SICI)1096-8652(199812) the immunological landscape, Nat. Rev. Rheumatol. 12 (2016) 63–68, http://dx.
59:4<288:AID-AJH4>3.0.CO;2-I. doi.org/10.1038/nrrheum.2015.171.
[6] J.R. Baker, B. Riske, J.H. Drake, A.D. Forsberg, R. Atwood, M. Voutsis, R. Shearer, [33] X. Houard, M.B. Goldring, F. Berenbaum, Homeostatic mechanisms in articular
US Hemophilia Treatment Center population trends 1990–2010: patient diag- cartilage and role of inflammation in osteoarthritis, Curr. Rheumatol. Rep. 15
noses, demographics, health services utilization, Haemophilia 19 (2013) 21–26, (2013) 375, http://dx.doi.org/10.1007/s11926-013-0375-6.
http://dx.doi.org/10.1111/j.1365-2516.2012.02915.x. [34] Z. Szekanecz, A.E. Koch, Update on synovitis, Curr. Rheumatol. Rep. 3 (2001)
[7] J.S. Stonebraker, P.H. Bolton-Maggs, J.M. Soucie, I. Walker, M. Brooker, A study of 53–63, http://dx.doi.org/10.1007/s11926-001-0051-0.
variations in the reported haemophilia A prevalence around the world, [35] C.A. Dinarello, Overview of the interleukin-1 family of ligands and receptors,
Haemophilia 16 (2010) 20–32, http://dx.doi.org/10.1111/j.1365-2516.2009. Semin. Immunol. 25 (2013) 389–393, http://dx.doi.org/10.1016/j.smim.2013.10.
02127.x. 001.
[8] J.S. Stonebraker, P.H. Bolton-Maggs, J. Michael Soucie, I. Walker, M. Brooker, A [36] P.E. Auron, A.C. Webb, L.J. Rosenwasser, S.F. Mucci, A. Rich, S.M. Wolff, et al.,
study of variations in the reported haemophilia B prevalence around the world, Nucleotide sequence of human monocyte interleukin 1 precursor cDNA, Proc.
Haemophilia 18 (3) (2012) e91–e94, http://dx.doi.org/10.1111/j.1365-2516. Natl. Acad. Sci. U. S. A. 81 (1984) 7907–7911.
2011.02588.x. [37] F.F. Dutra, L.S. Alves, D. Rodrigues, P.L. Fernandez, R.B. de Oliveira,
[9] L. Autin, M.A. Miteva, W.H. Lee, K. Mertens, K.P. Radtke, B.O. Villoutreix, D.T. Golenbock, et al., Hemolysis-induced lethality involves inflammasome

87
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

activation by heme, Proc. Natl. Acad. Sci. U. S. A. 111 (2014) E4110–E4118, [62] S. Zhang, C. Lu, J. Ying, P. Wang, T. Xu, D. Chen, et al., Inflammatory focal bone
http://dx.doi.org/10.1073/pnas.1405023111. destruction in femoral heads with end-stage haemophilic arthropathy: a study on
[38] J.A. Symons, P.R. Young, G.W. Duff, Soluble type II interleukin 1 (IL-1) receptor clinic samples with micro-CT and histological analyses, Haemophilia 21 (6) (2015)
binds and blocks processing of IL-1 beta precursor and loses affinity for IL-1 re- e472–e478, http://dx.doi.org/10.1111/hae.12808.
ceptor antagonist, Proc. Natl. Acad. Sci. U. S. A. 92 (1995) 1714–1718. [63] M.J. Hooiveld, G. Roosendaal, H.M. Van Den Berg, J.W. Bijlsma, F.P. Lafeber,
[39] D. Boraschi, A. Tagliabue, The interleukin-1 receptor family, Semin. Immunol. 25 Haemoglobin-derived iron-dependent hydroxyl radical formation in bloodinduced
(2013) 394–407, http://dx.doi.org/10.1016/j.smim.2013.10.023. joint damage: an in vitro study, Rheumatology (Oxford) 42 (2003) 784–790,
[40] M.U. Martin, H. Wesche, Summary and comparison of the signaling mechanisms of http://dx.doi.org/10.1093/rheumatology/keg220.
the Toll/interleukin-1 receptor family, Biochim. Biophys. Acta 1592 (2002) [64] G.R. Sambrano, V. Terpstra, D. Steinberg, Independent mechanisms for macro-
265–280, http://dx.doi.org/10.1016/S0167-4889(02)00320-8. phage binding and macrophage phagocytosis of damaged erythrocytes. Evidence
[41] J.A. Roman-Blas, S.A. Jimenez, NF-kappaB as a potential therapeutic target in of receptor cooperativity, Arterioscler. Thromb Vasc. Biol. 17 (1997) 3442–3448,
osteoarthritis and rheumatoid arthritis, Osteoarthr. Cartil. 14 (2006) 839–848, http://dx.doi.org/10.1161/01.ATV.17.12.3442.
http://dx.doi.org/10.1016/j.joca.2006.04.008. [65] L.F. van Vulpen, R.E. Schutgens, K. Coeleveld, E.C. Alsema, G. Roosendaal,
[42] J.I. Torrero, C. Martínez, New developments in the treatment of osteoarthritis – S.C. Mastbergen, et al., IL-1β, in contrast to TNFα, is pivotal in blood-induced
focus on biologic agents, Open Access Rheumatol. 7 (2015) 33–43, http://dx.doi. cartilage damage and is a potential target for therapy, Blood 126 (2015)
org/10.2147/OARRR.S50058. 2239–2246, http://dx.doi.org/10.1182/blood-2015-03-635524.
[43] J.J. Nietfeld, B. Wilbrink, W. Den Otter, J. Huber, O. Huber-Bruning, The effect of [66] J.C. Shanahan, W. St Clair, Tumor necrosis factor-alpha blockade: a novel therapy
human interleukin 1 on proteoglycan metabolism in human and porcine cartilage for rheumatic disease, Clin. Immunol. 103 (2002) 231–242, http://dx.doi.org/10.
explants, J. Rheumatol. 17 (1990) 818–826. 1006/clim.2002.5191.
[44] G. Murphy, H. Nagase, Reappraising metalloproteinases in rheumatoid arthritis [67] B.B. Aggarwal, S.C. Gupta, J.H. Kim, Historical perspectives on tumor necrosis
and osteoarthritis: destruction or repair? Nat. Clin. Pract. Rheumatol. 4 (2008) factor and its superfamily: 25 years later, a golden journey, Blood 119 (2012)
128–135, http://dx.doi.org/10.1038/ncprheum0727. 651–665, http://dx.doi.org/10.1182/blood-2011-04-325225.
[45] M. Kobayashi, G.R. Squires, A. Mousa, M. Tanzer, D.J. Zukor, J. Antoniou, [68] J. Scheller, A. Chalaris, C. Garbers, S. Rose-John, ADAM17: a molecular switch to
U. Feige, A.R. Poole, Role of interleukin-1 and tumor necrosis factor alpha in control inflammation and tissue regeneration, Trends Immunol. 32 (2011)
matrix degradation of human osteoarthritic cartilage, Arthritis Rheum. 52 (2005) 380–387, http://dx.doi.org/10.1016/j.it.2011.05.005.
128–135, http://dx.doi.org/10.1002/art.20776. [69] C. Haxaire, C.P. Blobel, With blood in the joint – what happens next? Could ac-
[46] M.J. López-Armada, B. Caramés, M. Lires-Deán, B. Cillero-Pastor, C. Ruiz-Romero, tivation of a pro-inflammatory signalling axis leading to iRhom2/TNFα-con-
F. Galdo, et al., Cytokines, tumor necrosis factor-alpha and interleukin-1beta, vertase-dependent release of TNFα contribute to haemophilic arthropathy?
differentially regulate apoptosis in osteoarthritis cultured human chondrocytes, Haemophilia 20 (Suppl. 4) (2014) 11–14, http://dx.doi.org/10.1111/hae.12416.
Osteoarthr. Cartil. 14 (2006) 660–669, http://dx.doi.org/10.1016/j.joca.2006.01. [70] D.J. MacEwan, TNF receptor subtype signalling: differences and cellular con-
005. sequences, Cell. Signal. 14 (2002) 477–492, http://dx.doi.org/10.1016/S0898-
[47] Y.E. Henrotin, P. Bruckner, J.P. Pujol, The role of reactive oxygen species in 6568(01)00262-5.
homeostasis and degradation of cartilage, Osteoarthr. Cartil. 11 (2003) 747–755, [71] C.I. Westacott, A.F. Barakat, L. Wood, M.J. Perry, P. Neison, I. Bisbinas, et al.,
http://dx.doi.org/10.1016/S1063-4584(03)00150-X. Tumor necrosis factor alpha can contribute to focal loss of cartilage in osteoar-
[48] C. Jacques, M. Gosset, F. Berenbaum, C. Gabay, The role of IL-1 and IL-1Ra in joint thritis, Osteoarthr. Cartil. 8 (2000) 213–221, http://dx.doi.org/10.1053/joca.
inflammation and cartilage degradation, Vitam. Horm. 74 (2006) (2006) 371–403, 1999.0292.
http://dx.doi.org/10.1016/S0083-6729(06)74016-X. [72] E.A. Moelantis, A. Mortier, J. Van Damme, P. Proost, Regulation of TNF-alfa with a
[49] B. Nedoszytko, M. Sokołowska-Wojdyło, K. Ruckemann-Dziurdzińska, focus on rheumatoid arthritis, Immunol. Cell Biol. 91 (2013) 393–401, http://dx.
J. Roszkiewicz, R.J. Nowicki, Chemokines and cytokines network in the patho- doi.org/10.1038/icb.2013.15.
genesis of the inflammatory skin diseases: atopic dermatitis, psoriasis and skin [73] J. Karar, A. Maity, PI3K/AKT/mTOR pathway in angiogenesis, Front. Mol.
mastocytosis, Postepy Dermatol. Alergol. 31 (2014) 84–91, http://dx.doi.org/10. Neurosci. 4 (2011) 51, http://dx.doi.org/10.3389/fnmol.2011.00051.
5114/pdia.2014.40920. [74] S.S. Acharya, R.N. Kaplan, D. Macdonald, O.T. Fabiyi, D. DiMichele, D. Lyden,
[50] F.E. El Mansouri, N. Chabane, N. Zayed, M. Kapoor, M. Benderdour, J. Martel- Neoangiogenesis contributes to the development of hemophilic synovitis, Blood
Pelletier, et al., Contribution of H3K4 methylation by SET-1A to interleukin-1- 117 (2011) 2484–2493, http://dx.doi.org/10.1182/blood-2010-05-284653.
induced cyclooxygenase 2 and inducible nitric oxide synthase expression in human [75] T. Saito, A. Fukai, A. Mabuchi, T. Ikeda, F. Yano, S. Ohba, et al., Transcriptional
osteoarthritis chondrocytes, Arthritis Rheum. 63 (2011) 168–179, http://dx.doi. regulation of endochondral ossification by HIF-2alpha during skeletal growth and
org/10.1002/art.27762. osteoarthritis development, Nat. Med. 16 (2010) 678–686, http://dx.doi.org/10.
[51] S.C. Gilman, J. Chang, P.R. Zeigler, J. Uhl, E. Mochan, Interleukin-1 activates 1038/nm.2146.
phospholipase A2 in human synovial cells, Arthritis Rheum. 31 (1988) 126–130, [76] B. Henderson, E.R. Pettipher, Arthritogenic actions of recombinant IL-1 and tu-
http://dx.doi.org/10.1002/art.1780310118. mour necrosis factor alpha in the rabbit: evidence for synergistic interactions
[52] M.M. Hardy, K. Seibert, P.T. Manning, M.G. Currie, B.M. Woerner, D. Edwards, between cytokines in vivo, Clin. Exp. Immunol. 75 (1989) 306–310.
et al., Cyclooxygenase 2-dependent prostaglandin E2 modulates cartilage pro- [77] J. Sun, G. Hu, P.E. Monahan, TNF-alpha antagonists augment factor replacement
teoglycan degradation in human osteoarthritis explants, Arthritis Rheum. 46 to prevent arthropathy in hemophilic mice, J. Thromb. Haemost. 7 (2009) (2009)
(2002) (2002) 1789–1803, http://dx.doi.org/10.1002/art.10356. 225.
[53] G. Tagariello, F.S. di Giovine, Interleukin-1 in haemophilic arthritis, Thromb. [78] A.L. Forsyth, G.É. Rivard, L.A. Valentino, N. Zourikian, M. Hoffman,
Haemost. 75 (1996) 979–980. P.E. Monahan, et al., Consequences of intra-articular bleeding in haemophilia:
[54] G. Tagariello, A. Traldi, P.G. Davoli, et al., May interleukin-1 in synovial tissue science to clinical practice and beyond, Haemophilia 18 (Suppl. 4) (2012)
play a major role in the progression of haemophilic arthropathy? Proceedings of 112–119, http://dx.doi.org/10.1111/j.1365-2516.2012.02835.x.
the 4th Musculoskeletal Congress of the World Federation of Haemophilia, [79] D. Melchiorre, M. Morfini, S. Linari, A.L. Zignego, M. Innocenti, M. Matucci
Madrid, 1997, p. 21. Cerinic, Anti-TNF-α therapy prevents the recurrence of joint bleeding in haemo-
[55] G. Roosendaal, A.C. van Rinsum, M.E. Vianen, H.M. van den Berg, F.P. Lafeber, philia and arthritis, Rheumatology (Oxford) 53 (2014) 576–578, http://dx.doi.
J.W. Bijlsma, Haemophilic arthropathy resembles degenerative rather than in- org/10.1093/rheumatology/ket280.
flammatory joint disease, Histopathology 34 (1999) 144–153, http://dx.doi.org/ [80] Y. Dargaud, H. Simpson, Y. Chevalier, J.Y. Scoazec, A. Hot, O. Guyen, et al., The
10.1046/j. 1365–2559.1999.00608.x. potential role of synovial thrombomodulin in the pathophysiology of joint bleeds
[56] G. Roosendaal, M.E. Vianen, M.J. Wenting, A.C. van Rinsum, H.M. van den Berg, in haemophilia, Haemophilia 18 (2012) 818–823, http://dx.doi.org/10.1111/j.
F.P. Lafeber, et al., Iron deposits and catabolic properties of synovial tissue from 1365-2516.2012.02809.x.
patients with haemophilia, J. Bone Joint Surg. Br. 80 (1998) 540–545. [81] G. Anastasiou, A. Argyri Gialeraki, E. Merkouri, M. Politou, A. Travlou,
[57] M. Hooiveld, G. Roosendaal, M. Wenting, M. van den Berg, J. Bijlsma, F. Lafeber, Thrombomodulin as a regulator of the anticoagulant pathway: implication in the
Short-term exposure of cartilage to blood results in chondrocyte apoptosis, Am. J. development of thrombosis, Blood Coagul. Fibrinolysis 23 (2012) 1–10, http://dx.
Pathol. 162 (2003) 943–951, http://dx.doi.org/10.1016/S0002-9440(10) doi.org/10.1097/MBC.0b013e32834cb271.
63889-8. [82] Y. Dargaud, J.Y. Scoazec, S.J. Wielders, C. Trzeciak, T.M. Hackeng, C. Négrier,
[58] G. Roosendaal, M.E. Vianen, J.J. Marx, H.M. Van Den Berg, F.P. Lafeber, et al., Characterization of an autosomal dominant bleeding disorder caused by a
J.W. Bijlsma, Blood-induced joint damage: a human in vitro study, Arthritis thrombomodulin mutation, Blood 125 (2015) 1497–1501, http://dx.doi.org/10.
Rheum. 42 (1999) 1025–1032, http://dx.doi.org/10.1002/1529-0131(199905) 1182/blood-2014-10-604553.
42:5<1025:AID-ANR23>3.0.CO;2-3. [83] A. Hammacher, L.D. Ward, J. Weinstock, H. Treutlein, K. Yasukawa, R.J. Simpson,
[59] J.F. Telfer, J.H. Brock, Proinflammatory cytokines increase iron uptake into Structure-function analysis of human IL-6: identification of two distinct regions
human monocytes and synovial fibroblasts from patients with rheumatoid ar- that are important for receptor binding, Protein Sci. 3 (1994) 2280–2293, http://
thritis, Med. Sci. Monit. 10 (2004) BR91–BR95. dx.doi.org/10.1002/pro.5560031213.
[60] K. Øvlisen, A.T. Kristensen, A.L. Jensen, TranholmIL-1β, IL-6, KC and MCP-1 are [84] T. Kishimoto, IL-6: from its discovery to clinical applications, Int. Immunol. 22
elevated in synovial fluid from haemophilic mice with experimentally induced (2010) 347–352, http://dx.doi.org/10.1093/intimm/dxq030.
haemarthrosis, Haemophilia 15 (2009) 802–810, http://dx.doi.org/10.1111/j. [85] T. Taga, M. Hibi, Y. Hirata, K. Yamasaki, K. Yasukawa, T. Matsuda, et al.,
1365-2516.2008.01973.x. Interleukin-6 triggers the association of its receptor with a possible signal trans-
[61] L.A. Valentino, N. Hakobyan, T. Kazarian, K.J. Jabbar, A.A. Jabbar, Experimental ducer, gp130, Cell 58 (1989) 573–581, http://dx.doi.org/10.1016/0092-8674(89)
haemophilic synovitis: rationale and development of a murine model of human 90438-8.
factor VIII deficiency, Haemophilia 10 (2004) 280–287, http://dx.doi.org/10. [86] F. Schaper, S. Rose-John, Interleukin-6. Biology, signaling and strategies of
1111/j.1365-2516.2004.00899.x. blockade, Cytokine Growth Factor Rev. 26 (2015) 475–487, http://dx.doi.org/10.

88
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

1016/j.cytogfr.2015.07.004. Intra-articular injection of interleukin-4 decreases nitric oxide production by


[87] J.Y. Park, M.H. Pillinger, Interleukin-6 in the pathogenesis of rheumatoid arthritis, chondrocytes and ameliorates subsequent destruction of cartilage in instability-
Bull. NYU Hosp. Jt. Dis. 65 (Suppl. 1) (2007) S4–10. induced osteoarthritis in rat knee joints, Osteoarthr. Cartil. 16 (2008) 764–771,
[88] S. Srirangan, E.H. Choy, The role of interleukin 6 in the pathophysiology of http://dx.doi.org/10.1016/j.joca.2007.11.006.
rheumatoid arthritis, Ther. Adv. Musculoskelet. Dis. 2 (2010) 247–256, http://dx. [113] A.J. Schuerwegh, E.J. Dombrecht, W.J. Stevens, J.F. Van Offel, C.H. Bridts, L.S. De
doi.org/10.1177/1759720x10378372. Clerck, Influence of pro-inflammatory (IL-1 alpha, IL-6, TNF-alpha, IFN-gamma)
[89] O. Stannus, G. Jones, F. Cicuttini, V. Parameswaran, S. Quinn, J. Burgess, et al., and anti-inflammatory (IL-4) cytokines on chondrocyte function, Osteoarthr.
Circulating levels of IL-6 and TNF-α are associated with knee radiographic os- Cartil. 11 (2003) 681–687, http://dx.doi.org/10.1016/S1063-4584(03)00156-0.
teoarthritis and knee cartilage loss in older adults, Osteoarthr. Cartil. 18 (2010) [114] J. Fu, L. Zhang, S. Song, K. Sheng, Y. Li, P. Li, et al., Effect of bone marrow-derived
1441–1447, http://dx.doi.org/10.1016/j.joca.2010.08.016. CD11b + F4/80 + immature dendritic cells on the balance between pro-in-
[90] B. Porée, M. Kypriotou, C. Chadjichristos, G. Beauchef, E. Renard, F. Legendre, flammatory and anti-inflammatory cytokines in DBA/1 mice with collagen-in-
et al., Interleukin-6 (IL-6) and/or soluble IL-6 receptor down-regulation of human duced arthritis, Inflamm. Res. 63 (2014) 357–367, http://dx.doi.org/10.1007/
type II collagen gene expression in articular chondrocytes requires a decrease of s00011-014-0707-7.
Sp1. Sp3 ratio and of the binding activity of both factors to the COL2A1 promoter, [115] M.E. van Meegeren, G. Roosendaal, N.W. Jansen, M.J. Wenting, A.C. van Wesel,
J. Biol. Chem. 283 (2008) 4850–4865, http://dx.doi.org/10.1074/jbc. J.A. van Roon, et al., IL-4 alone and in combination with IL-10 protects against
M706387200. blood-induced cartilage damage, Osteoarthr. Cartil. 20 (2012) 764–772, http://
[91] A.D. Rowan, P.J. Koshy, W.D. Shingleton, B.A. Degnan, J.K. Heath, A.B. Vernallis, dx.doi.org/10.1016/j.joca.2012.04.002.
et al., Synergistic effects of glycoprotein 130 binding cytokines in combination [116] H. Niibayashi, K. Shimizu, K. Suzuki, S. Yamamoto, T. Yasuda, T. Yamamuro,
with interleukin-1 on cartilage collagen breakdown, Arthritis Rheum. 44 (2001) Proteoglycan degradation in hemarthrosis. Intraarticular, autologous blood in-
1620–1632, http://dx.doi.org/10.1002/1529-0131(200107)44:7<1620:AID- jection in rat knees, Acta Orthop. Scand. 66 (1995) 73–79, http://dx.doi.org/10.
ART285>3.0.CO;2-B. 3109/17453679508994645.
[92] S. Kwan Tat, M. Padrines, S. Théoleyre, D. Heymann, Y. Fortun, RANKL, TNF- [117] M.E. van Meegeren, G. Roosendaal, K. Coeleveld, L. Nieuwenhuizen,
alpha/IL-1: interrelations in bone resorption pathophysiology, Cytokine Growth S.C. Mastbergen, F.P. Lafeber, A single intra-articular injection with IL-4 plus IL-10
Factor Rev. 15 (2004) 49–60, http://dx.doi.org/10.1016/j.cytogfr.2003.10.005. ameliorates blood-induced cartilage degeneration in haemophilic mice, Br. J.
[93] J. Scheller, A. Chalaris, D. Schmidt-Arras, S. Rose-John, The pro- and anti-in- Haematol. 160 (2013) 515–520, http://dx.doi.org/10.1111/bjh.12148.
flammatory properties of the cytokine interleukin-6, Biochim. Biophys. Acta 1813 [118] M.E. van Meegeren, G. Roosendaal, K. van Veghel, S.C. Mastbergen, F.P. Lafeber,
(2011) 878–888, http://dx.doi.org/10.1016/j.bbamcr.2011.01.034. A short time window to profit from protection of blood-induced cartilage damage
[94] A.S. de Hooge, F.A. van de Loo, M.B. Bennink, O.J. Arntz, P. de Hooge, W.B. van by IL-4 plus IL-10, Rheumatology (Oxford) 52 (2013) 1563–1571, http://dx.doi.
den Berg, Male IL-6 gene knock out mice developed more advanced osteoarthritis org/10.1093/rheumatology/ket005.
upon aging, Osteoarthr. Cartil. 13 (2005) 66–73, http://dx.doi.org/10.1016/j. [119] T. Wyseure, L.O. Mosnier, A. von Drygalski, Advances and challenges in hemo-
joca.2004.09.011. philic arthropathy, Semin. Hematol. 53 (2016) 10–19, http://dx.doi.org/10.1053/
[95] F.A. van de Loo, S. Kuiper, F.H. van Enckevort, O.J. Arntz, W.B. van den Berg, j.seminhematol.2015.10.005.
Interleukin-6 reduces cartilage destruction during experimental arthritis. A study [120] R. Ravanbod, G. Torkaman, M. Mophid, F. Mohammadali, Experimental study on
in interleukin-6-deficient mice, Am. J. Pathol. 151 (1997) 177–191. the role of intra-articular injection of MSCs on cartilage regeneration in haemo-
[96] S. Rose-John, IL-6 trans-signaling via the soluble IL-6 receptor: importance for the philia, Haemophilia 21 (2015) 693–701, http://dx.doi.org/10.1111/hae.12659.
pro-inflammatory activities of IL-6, Int. J. Biol. Sci. 8 (2012) 1237–1247, http:// [121] E.C. Rodríguez-Merchán, Intra-articular injections of mesenchymal stem cells
dx.doi.org/10.7150/ijbs.4989. (MSCs) as a treatment for hemophilic arthropathy, Expert Rev. Hematol. 9 (2016)
[97] S. Rose-John, J. Scheller, G. Elson, S.A. Jones, Interleukin-6 biology is coordinated 737–741, http://dx.doi.org/10.1080/17474086.2016.1203780.
by membrane-bound and soluble receptors: role in inflammation and cancer, J. [122] A.C. Silveira, M.A. Santana, I.G. Ribeiro, D.G. Chaves, O.A. Martins-Filho, The IL-
Leukoc. Biol. 80 (2006) 227–236, http://dx.doi.org/10.1189/jlb.1105674. 10 polarized cytokine pattern in innate and adaptive immunity cells contribute to
[98] N. Narkbunnam, J. Sun, G. Hu, F.C. Lin, T.A. Bateman, M. Mihara, et al., IL-6 the development of FVIII inhibitors, BMC Hematol. 15 (2015) 1, http://dx.doi.
receptor antagonist as adjunctive therapy with clotting factor replacement to org/10.1186/s12878-014-0019-8.
protect against bleeding-induced arthropathy in hemophilia, J. Thromb. Haemost. [123] D. Agostini, C. Rosset, M.R. Botton, D.B. Kappel, I.A. Vieira, R.P. Gorziza, et al.,
11 (2013) 881–893, http://dx.doi.org/10.1111/jth.12176. Immune system polymorphisms and factor VIII inhibitor formation in Brazilian
[99] S.L. Deshmane, S. Kremlev, S. Amini, B.E. Sawaya, Monocyte chemoattractant haemophilia A severe patients, Haemophilia 18 (2012) e416–e418, http://dx.doi.
protein-1 (MCP-1): an overview, J. Interferon Cytokine Res. 29 (2009) 313–326, org/10.1111/hae.12015.
http://dx.doi.org/10.1089/jir.2008.0027. [124] J. Astermark, J. Oldenburg, A. Pavlova, E. Berntorp, A.K. Lefvert, Mibs Study
[100] Y. Kobayashi, The role of chemokines in neutrophil biology, Front. Biosci. 13 Group, Polymorphisms in the IL10 but not in the IL1beta and IL4 genes are as-
(2008) 2400–2407, http://dx.doi.org/10.2741/2853. sociated with inhibitor development in patients with hemophilia A, Blood 107
[101] M. Frink, Y.C. Hsieh, C.H. Hsieh, H.C. Pape, M.A. Choudhry, M.G. Schwacha, (2006) 3167–3172, http://dx.doi.org/10.1182/blood-2005-09-3918.
et al., Keratinocyte-derived chemokine plays a critical role in the induction of [125] J.C. Tan, S.R. Indelicato, S.K. Narula, P.J. Zavodny, C.C. Chou, Characterization of
systemic inflammation and tissue damage after trauma-hemorrhage, Shock 28 interleukin-10 receptors on human and mouse cells, J. Biol. Chem. 268 (1993)
(2007) 576–581, http://dx.doi.org/10.1097/shk.0b013e31814b8e0d. 21053–21059.
[102] S.M. Opal, V.A. DePalo, Anti-inflammatory cytokines, Chest 117 (2000) [126] M.R. Walter, The molecular basis of IL-10 function: from receptor structure to the
1162–1172, http://dx.doi.org/10.1378/chest.117.4.1162. onset of signalling, Curr. Top. Microbiol. Immunol. 380 (2014) 191–212, http://
[103] M.B. Goldring, M. Otero, K. Tsuchimochi, K. Ijiri, Y. Li, Defining the roles of in- dx.doi.org/10.1007/978-3-662-43492-5_9.
flammatory and anabolic cytokines in cartilage metabolism, Ann. Rheum. Dis. 67 [127] M. Saraiva, A. O'Garra, The regulation of IL-10 production by immune cells, Nat.
(Suppl. (2008)) (2008) 75–82, http://dx.doi.org/10.1136/ard.2008.098764. Rev. Immunol. 10 (2010) 170–181, http://dx.doi.org/10.1038/nri2711.
[104] J. Banchereau, F. Brière, J.P. Galizzi, P. Miossec, F. Rousset, Human interleukin 4, [128] W. Ouyang, S. Rutz, N.K. Crellin, P.A. Valdez, S.G. Hymowitz, Regulation and
J. Lipid Mediat. Cell Signal. 9 (1994) 43–53. functions of the IL-10 family of cytokines in inflammation and disease, Annu. Rev.
[105] T.D. Mueller, J.L. Zhang, W. Sebald, A. Duschl, Structure, binding, and antagonists Immunol. 29 (2011) 71–109, http://dx.doi.org/10.1146/annurev-immunol-
in the IL-4/IL-13 receptor system, Biochim. Biophys. Acta 1592 (2002) 237–250, 031210-101312.
http://dx.doi.org/10.1016/S0167-4889(02)00318-X. [129] S. Lacraz, L.P. Nicod, R. Chicheportiche, H.G. Welgus, J.M. Dayer, IL-10 inhibits
[106] A. Bhattacharjee, M. Shukla, V.P. Yakubenko, A. Mulya, S. Kundu, M.K. Cathcart, metalloproteinase and stimulates TIMP-1 production in human mononuclear
IL-4 and IL-13 employ discrete signaling pathways for target gene expression in phagocytes, J. Clin. Invest. 96 (1995) 2304–2310, http://dx.doi.org/10.1172/
alternatively activated monocytes/macrophages, Free Radic. Biol. Med. 54 (2013) JCI118286.
1–16, http://dx.doi.org/10.1016/j.freeradbiomed.2012.10.553. [130] D. Umulis, M.B. O'Connor, S.S. Blair, The extracellular regulation of bone mor-
[107] H. Jiang, M.B. Harris, P. Rothman, IL-4/IL-13 signaling beyond JAK/STAT, J. phogenetic protein signaling, Development 136 (2009) 3715–3728, http://dx.doi.
Allergy Clin. Immunol. 105 (2000) 1063–1070, http://dx.doi.org/10.1067/mai. org/10.1242/dev.031534.
2000.107604. [131] N.W. Jansen, G. Roosendaal, M.J. Hooiveld, J.W. Bijlsma, J.A. van Roon,
[108] H. Renz, Soluble interleukin-4 receptor (sIL-4R) in allergic diseases, Inflamm. Res. M. Theobald, et al., Interleukin-10 protects against blood-induced joint damage,
48 (1999) 425–431, http://dx.doi.org/10.1007/s000110050482. Br. J. Haematol. 142 (2008) 953–961, http://dx.doi.org/10.1111/j.1365-2141.
[109] H. Ishii, H. Tanaka, K. Katoh, H. Nakamura, M. Nagashima, S. Yoshino, 2008.07278.x.
Characterization of infiltrating T cells and Th1/Th2-type cytokines in the syno- [132] M.E.R. van Meegeren, Blood-induced Joint Damage: Novel Targets for Therapy,
vium of patients with osteoarthritis, Osteoarthr. Cartil. 10 (2002) 277–281, http:// Dissertation, Utrecht University, 2012, http://dspace.library.uu.nl/handle/1874/
dx.doi.org/10.1053/joca.2001.0509. 255568.
[110] J. Zhu, T helper 2 (Th2) cell differentiation, type 2 innate lymphoid cell (ILC2) [133] L.F.D. van Vulpen, J. Popov-Celeketic, M.E.R. van Meegeren, K. Coeleveld,
development and regulation of interleukin-4 (IL-4) and IL-13 production, Cytokine J.M. van Laar, C.E. Hack, R.E.G. Schutgens, S.C. Mastbergen, Lafeber FPJG, A
75 (2015) 14–24, http://dx.doi.org/10.1016/j.cyto.2015.05.010. fusion protein of interleukin-4 and interleukin-10 protects against blood-induced
[111] N. Alaaeddine, J.A. Di Battista, J.P. Pelletier, K. Kiansa, J.M. Cloutier, J. Martel- cartilage damage in vitro and in vivo, J. Thromb. Haemost. 15 (September (9))
Pelletier, Inhibition of tumor necrosis factor alpha-induced prostaglandin E2 (2017) 1788–1798, http://dx.doi.org/10.1111/jth.13778.
production by the antiinflammatory cytokines interleukin-4, interleukin-10, and [134] G. Lisignoli, A. Piacentini, S. Toneguzzi, F. Grassi, B. Cocchini, A. Ferruzzi,
interleukin-13 in osteoarthritic synovial fibroblasts: distinct targeting in the sig- Osteoblasts and stromal cells isolated from femora in rheumatoid arthritis (RA)
naling pathways, Arthritis Rheum. 42 (1999) 710–718, http://dx.doi.org/10. and osteoarthritis (OA) patients express IL-11, leukaemia inhibitory factor and
1002/1529-0131(199904)42:4<710:AID-ANR14>3.0.CO;2-4. oncostatin M, Clin. Exp. Immunol. 119 (2000) 346–353, http://dx.doi.org/10.
[112] M. Yorimitsu, K. Nishida, A. Shimizu, H. Doi, S. Miyazawa, T. Komiyama, et al., 1046/j.1365-2249.2000.01114.x.

89
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

[135] M.V. Ragni, E.M. Novelli, A. Murshed, E.P. Merricks, M.T. Kloos, T.C. Nichols, Łukasz A. Poniatowski was graduated and obtained his
Phase II prospective open-label trial of recombinant interleukin-11 in desmo- MD from the Medical University of Warsaw in 2016.
pressin-unresponsive von Willebrand disease and mild or moderate haemophilia Currently he is a PhD student in the Department of
A, Thromb. Haemost. 109 (2013) 248–254, http://dx.doi.org/10.1160/TH12-06- Experimental and Clinical Pharmacology at the Medical
0447. University of Warsaw. During his studies he received many
[136] C.V. Denis, K. Kwack, S. Saffaripour, S. Maganti, P. André, R.G. Schaub, et al., awards including the scholarship for outstanding achieve-
Interleukin 11 significantly increases plasma von Willebrand factor and factor VIII ments from the Polish Ministry of Health. His research in-
in wild type and von Willebrand disease mouse models, Blood 97 (2001) 465–472, terests are concentrated on neurosurgery and im-
http://dx.doi.org/10.1182/blood.V97.2.465. munological aspects in the pathogenesis of musculoskeletal
[137] R.N. Wang, J. Green, Z. Wang, Y. Deng, M. Qiao, M. Peabody, Bone Morphogenetic and central nervous system diseases.
Protein (BMP) signaling in development and human diseases, Genes Dis. 1 (2014)
87–105, http://dx.doi.org/10.1016/j.gendis.2014.07.005.
[138] K.R. Garrison, I. Shemilt, S. Donell, J.J. Ryder, M. Mugford, I. Harvey, et al., Bone
morphogenetic protein (BMP) for fracture healing in adults, Cochrane Database
Syst. Rev. 6 (2010) CD006950, http://dx.doi.org/10.1002/14651858.CD006950.
Paweł Nauman is head of the Department of Neurosurgery
pub2.
at the Institute of Psychiatry and Neurology in Warsaw and
[139] J.M. Woods, G.K. Haines, M.R. Shah, G. Rayan, A.E. Koch, Low-level production of
former deputy head of the Department of Neurosurgery at
interleukin-13 in synovial fluid and tissue from patients with arthritis, Clin.
the Maria Skłodowska-Curie Memorial Cancer Center and
Immunol. Immunopathol. 85 (1997) 210–220, http://dx.doi.org/10.1006/clin.
Institute of Oncology in Warsaw. He received his PhD in the
1997.4441.
field of neurosurgery in 2001 from Mossakowski Medical
[140] T. Hanada, A. Yoshimura, Regulation of cytokine signaling and inflammation,
Research Centre of the Polish Academy of Sciences and did
Cytokine Growth Factor Rev. 13 (2002) 413–421, http://dx.doi.org/10.1016/
postdoctoral work at the Lund University and Memorial
S1359-6101(02)00026-6.
Sloan Kettering Cancer Center. His research interests are
[141] A. Tagliaferri, G.F. Rivolta, A. Iorio, E. Oliovecchio, M.E. Mancuso, M. Morfini,
concentrated on molecular oncology and brain tumor
et al., Mortality and causes of death in Italian persons with haemophilia,
biology.
1990–2007, Haemophilia 16 (2009) 437–446, http://dx.doi.org/10.1111/j.1365-
2516.2009.02188.x.
[142] E. Berntorp, A.D. Shapiro, Modern haemophilia care, Lancet 379 (2012)
1447–1456, http://dx.doi.org/10.1016/S0140-6736(11)61139-2.
[143] A.D. Shapiro, S. Akins, D.L. Cooper, Long-term outcomes from orthopaedic surgery Tomasz Mandat is a professor and deputy head of the
in haemophilia: are we measuring success and documenting and assessing com- Department of Neurosurgery at the Maria Skłodowska-
plications? Haemophilia 20 (2014) e367–371, http://dx.doi.org/10.1111/hae. Curie Memorial Cancer Center and Institute of Oncology in
12504. Warsaw and senior consultant in the Department of
[144] C. Négrier, J. Goudemand, T. Lambert, Haemophilic arthropathy from A to Z. Neurosurgery at the Institute of Psychiatry and Neurology
Introduction, Haemophilia 14 (Suppl. 4) (2008) 1–2, http://dx.doi.org/10.1111/j. in Warsaw. He received his PhD in the field of neurosurgery
1365-2516.2008.01731.x. in 2002 from Central Clinical Hospital of the Military
[145] E. Berntorp, J.J. Michiels, A healthy hemophilic patient without arthropathy: from Medical Academy in Warsaw (currently Military Institute of
concept to clinical reality, Semin. Thromb. Hemost. 29 (2003) 5–10, http://dx.doi. Medicine in Warsaw) and did postdoctoral work at the
org/10.1055/s-2003-37934. University of Paris-Est and University of British Columbia.
[146] D. Sen, A. Chapla, N. Walter, V. Daniel, A. Srivastava, G.R. Jayandharan, Nuclear His research interests are concentrated on stereotactic and
factor (NF)-(B and its associated pathways are major molecular regulators of functional neurosurgery, molecular oncology and brain
blood-induced joint damage in a murine model of hemophilia, J. Thromb. tumor therapy.
Haemost. 11 (2013) 293–306, http://dx.doi.org/10.1111/jth.12101.
[147] S.B. Jakowlew, Transforming growth factor-beta in cancer and metastasis, Cancer
Metastasis Rev. 25 (2006) 435–457, http://dx.doi.org/10.1007/s10555-006-
Agnieszka Paradowska-Gorycka is head of the
9006-2.
Department of Biochemistry and Molecular Biology at the
[148] J.D. Milner, T. Orekov, J.M. Ward, L. Cheng, F. Torres-Velez, I. Junttila, et al.,
Eleonora Reicher National Institute of Geriatrics,
Sustained IL-4 exposure leads to a novel pathway for hemophagocytosis, in-
Rheumatology and Rehabilitation in Warsaw. She received
flammation, and tissue macrophage accumulation, Blood 116 (2010) 2476–2483,
her PhD in the field of medical biology in 2010 from
http://dx.doi.org/10.1182/blood-2009-11-255174.
Medical University of Warsaw. Her research interests are
[149] C.A. Haskell, M.D. Cleary, I.F. Charo, Molecular uncoupling of fractalkine-medi-
concentrated on immunogenetics of rheumatic diseases.
ated cell adhesion and signal transduction. Rapid flow arrest of CX3CR1-expres-
sing cells is independent of G-protein activation, J. Biol. Chem. 274 (1999)
10053–10058, http://dx.doi.org/10.1074/jbc.274.15.10053.
[150] J.K. Harrison, Y. Jiang, E.A. Wees, M.N. Salafranca, H.X. Liang, L. Feng,
L. Belardinelli, Inflammatory agents regulate in vivo expression of fractalkine in
endothelial cells of the rat heart, J. Leukoc. Biol. 66 (1999) 937–944.

Piotr Wojdasiewicz was graduated and obtained his MD


Katarzyna Romanowska-Próchnicka is senior consultant
from the Medical University of Warsaw in 2010. He re-
in the Department of Systemic Connective Tissue Diseases
ceived his PhD in the field of pathology and orthopaedics in
at the Eleonora Reicher National Institute of Geriatrics,
2017 from Medical University of Warsaw. Currently he is
Rheumatology and Rehabilitation in Warsaw and assistant
assistant in the Department of General and Experimental
in the Department of General and Experimental Pathology
Pathology at the Medical University of Warsaw. His re-
at the Medical University of Warsaw. She received her PhD
search interests are concentrated on neurotraumatology,
in the field of pathology and rheumatology in 2014 from
immunological aspects in the pathogenesis of arthritis-as-
Medical University of Warsaw. Her research interests are
sociated diseases and spine surgery in patients with rheu-
concentrated on immunogenetics of rheumatic diseases and
matoid and degenerative diseases.
their specific treatment including its all practical and the-
oretical aspects.

90
P. Wojdasiewicz et al. Cytokine and Growth Factor Reviews 39 (2018) 71–91

Dariusz Szukiewicz is a professor and head of the Ireneusz Kotela is a professor and head of the Department
Department of General and Experimental Pathology at the of Orthopaedics and Traumatology at the Central Clinical
Medical University of Warsaw. He received his PhD in the Hospital of the Ministry of the Interior and Administration
field of pathology in 1997 from Medical University of in Warsaw. He received his PhD in the field of orthopaedics
Warsaw and did postdoctoral work at the New York State and traumatology in 1993 from Medical University of
Institute for Basic Research in Developmental Disabilities Warsaw. His research interests are concentrated on appli-
and Erasmus University Rotterdam. His research interests cation of biomaterials and its biomechanics in the man-
are concentrated on interactions within the cytokine net- agement of joint diseases and orthopaedic treatment of
work and immunopathological mechanisms underlying patients with musculoskeletal complications of hemophilia
pathophysiology of various diseases including rheumatoid, and other inherited bleeding disorders.
neurological, endocrine and placental diseases.

Andrzej Kotela is resident in orthopaedic and trauma- Iwona Kurkowska-Jastrzębska is a professor and head of
tology in the Department of Orthopaedics and the 2nd Department of Neurology at the Institute of
Traumatology at the Medical University of Warsaw. He Psychiatry and Neurology in Warsaw. She has worked for
received his PhD in the field of orthopaedics and trauma- more than 10 years in the Department of Experimental and
tology in 2013 from Central Clinical Hospital of the Clinical Pharmacology at the Medical University of
Ministry of the Interior and Administration in Warsaw. His Warsaw, where she was involved in basic research of neu-
research interests are concentrated on joint replacement rodegenerative processes and the role of neuroin-
surgery and orthopaedic treatment of patients with mus- flammatory reaction, and of the interactions between the
culoskeletal complications of hemophilia and other in- nervous and immune systems. She spent few short post-
herited bleeding disorders. doctoral internships in the Department of Neuropathology
and in the Department of Neuroimmunology of Max Planck
Institute of Psychiatry in Martisried. Her research interest
are currently concentrated on neuroimmunology, demyeli-
nating diseases of the central nervous system, neurodegenerative diseases and epilepsy.

Łukasz Kubaszewski is head of the Department of


Spondyloorthopaedics and Biomechanics of the Spine and Robert Gasik is a professor and head of the Department of
senior consultant in the Department of Orthopaedics and Neuroorthopaedics and Neurology and former head of the
Traumatology at the Poznań University of Medical Sciences. Department of Rheumaorthopaedics at the Eleonora
He received his PhD in the field of orthopaedics and trau- Reicher National Institute of Geriatrics, Rheumatology and
matology in 2006 from Poznań University of Medical Rehabilitation in Warsaw. He received his PhD in the field
Sciences. His research interests are concentrated on spine of rheumatology in 2002 from Eleonora Reicher Institute of
surgery including complex degenerative, deformity, trau- Rheumatology in Warsaw (currently Eleonora Reicher
matic and oncologic pathology. National Institute of Geriatrics, Rheumatology and
Rehabilitation in Warsaw). His research interests are con-
centrated on spine surgery in patients with rheumatoid
diseases including specialized care for sacroiliac pain and
immunological aspects in the pathogenesis of musculoske-
letal system diseases.

91

You might also like