You are on page 1of 8

Injury, Int. J.

Care Injured 45 (2014) 1816–1823

Contents lists available at ScienceDirect

Injury
journal homepage: www.elsevier.com/locate/injury

Review

The role of pleiotrophin in bone repair


Margarita Lamprou a,1, Angelos Kaspiris a,1, Elias Panagiotopoulos b, Peter V. Giannoudis c,d,
Evangelia Papadimitriou a,*
a
Laboratory of Molecular Pharmacology, Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece
b
Academic Department of Trauma & Orthopaedics, School of Medicine, University of Patras, Greece
c
Academic Department of Trauma & Orthopaedic Surgery, University of Leeds, Clarendon Wing, Floor A, Great George Street, Leeds General Infirmary,
LS1 3EX Leeds, UK
d
NIHR Leeds Biomedical Research Unit, Chapel Allerton Hospital, LS7 4SA Leeds, West Yorkshire, UK

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

Article history: Bone has an enormous capacity for growth, regeneration, and remodelling, largely due to induction of
Accepted 7 October 2014 osteoblasts that are recruited to the site of bone formation. Although the pathways involved have not
been fully elucidated, it is well accepted that the immediate environment of the cells is likely to play a
Keywords: role via cell–matrix interactions, mediated by several growth factors. Formation of new blood vessels is
Angiogenesis also significant and interdependent to bone formation, suggesting that enhancement of angiogenesis
Bone could be beneficial during the process of bone repair. Pleiotrophin (PTN), also called osteoblast-specific
Bone repair
factor 1, is a heparin-binding angiogenic growth factor, with a well-defined and significant role in both
Osteoblasts
Osteoclasts
physiological and pathological angiogenesis. In this review we summarise the existing evidence on the
Pleiotrophin role of PTN in bone repair.
ß 2014 Elsevier Ltd. All rights reserved.

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1816
Materials and methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1817
Results . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1817
PTN receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1817
Regulation of PTN expression. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1817
PTN and angiogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1818
PTN and bone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1819
PTN pathways in clinical skeletal diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1819
Osteoporosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1819
Fracture healing. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1820
Osteoarthritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1820
Discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1820
Conflict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1821
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1821

Introduction

The formation of new blood vessels is vital to the functioning of all


tissues during development, as well as during regeneration process-
* Corresponding author at: Laboratory of Molecular Pharmacology, Department
es. In this context, at a bone fracture site, angiogenesis restores the
of Pharmacy, University of Patras, GR 26504 Patras, Greece. Tel.: +30 2610 962336.
E-mail address: epapad@upatras.gr (E. Papadimitriou). hypoxia and nutrient deprivation and facilitates osteogenesis. A
1
These authors have equally contributed to the work. number of therapeutic approaches that use angiogenic growth

http://dx.doi.org/10.1016/j.injury.2014.10.013
0020–1383/ß 2014 Elsevier Ltd. All rights reserved.
M. Lamprou et al. / Injury, Int. J. Care Injured 45 (2014) 1816–1823 1817

factors and lead to enhanced angiogenesis seem to have a beneficial examined actions and possible mechanisms of PTN [8,21–27] or its
effect on bone repair, as evidenced both in vitro and in vivo, including receptors [8,16,17,28–31] actions in bone cells. The third category
the use of vascular endothelial growth factor (VEGF), fibroblast comprised of the studies, which investigate the macroscopic
growth factors (FGFs), transforming growth factor, platelet derived effects of altered PTN expression on bone formation and/or
growth factor (PDGF) and bone morphogenetic proteins (BMPs) remodelling in animal models [8,32–40] or in clinical studies
[1]. However, clinical data are limited and further research is required [41]. Finally, the fourth category included articles on the regulation
in order to elucidate the interrelationships among the involved of PTN expression in cells of the skeletal system and/or by factors
molecules and pathways, and to apply such approaches to cases known to play a role in bone repair or remodelling [42–54], as well
where bone regeneration is desirable. as data on the PTN actions related to angiogenesis [55–78].
Pleiotrophin (PTN) is a secreted angiogenic growth factor with a
well defined regulatory role in physiological and pathological PTN receptors
angiogenesis [2,3] and a potential role in bone formation and
remodelling. The peptide sequence is highly conserved across The first identified PTN receptor was N-syndecan through its
different species, with more than 90% identity observed among the heparan sulfate chains [79,80]. N-syndecan is expressed by
sequences of chicken, rat, bovine and human [4]. Pleiotrophin was osteoblasts/osteoblast precursors and may participate in the
initially purified from bovine uterus and neonatal rat brain, while regulation of osteoblast recruitment [8]. Besides N-syndecan,
homologues have been reported in different species such as PTN also interacts with syndecans 1 and 4, which contain both
human, mouse, chicken, fish, frogs and insects [5]. The protein is heparan and chondroitin sulfate chains. PTN binds to the highly
also known as heparin-binding growth-associated molecule, sulfated chondroitin sulfate (CS) chains of syndecan-4 with higher
heparin affin regulatory peptide, heparin binding growth factor- affinity than to those of syndecan-1 and removal of the CS chains
8, protein 18 kDa, heparin binding neurotrophic factor and decreased the association and dissociation rate constants of PTN
osteoblast specific factor 1 (OSF-1) [2,3]. The term OSF-1 was for both syndecans [81].
attributed to the factor in the early 90s, due to the fact that a cDNA The best studied PTN receptor is receptor protein tyrosine
clone coding for PTN was isolated from the murine osteoblastic cell phosphatase beta/zeta (RPTPb/z), initially isolated from neural
line MC3T3-E1. The human counterpart was also found in cDNA tissue as a transmembrane protein-tyrosine-phosphatase that
libraries from the human osteosarcoma cell line MG-63 [6]. consists of a large glycosylated extracellular domain, a transmem-
Heteronuclear NMR studies have not concluded to PTN’s brane region and a cytoplasmic portion that contains two repeated
conformation but have shown that PTN contains two b-sheet tyrosine phosphatase domains. Alternative splicing and proteolytic
domains connected by a flexible linker. Each of these domains processing give rise to secreted, transmembrane, or cytoplasmic
contains three antiparallel b-strands, which are homologous to the forms of not yet fully identified biological significance [82]. RPTPb/
thrombospondin type 1 repeat (TSR-1) [4], a motif that is found in a z has a well described role in PTN-induced cell migration [83] and
wide variety of proteins that mediate interactions between cells may have a role in bone formation and remodelling based on the
and/or between cells and the extracellular matrix. Thrombospon- data presented in Table 1.
din and other TSR-containing proteins influence multiple aspects An important PTN receptor that determines the stimulatory or
of bone development and remodelling [7] supporting the the inhibitory effect of PTN on cell migration is anb3 integrin,
hypothesis that PTN may also be a significant regulator. which forms a functional complex with RPTPb/z on the cell surface
[75], and mediates PTN-induced cell surface nucleolin localization
Materials and methods [76]. PTN also interacts with several glycosaminoglycans (GAGs)
and such interactions contribute to PTN dimerization or storage
The authors explored the MEDLINE/PubMed (1946 – present) into the extracellular matrix [84,85], as well as protection of PTN
databases of the National Library of Medicine and EMBASE (1947 – from proteolytic degradation [77]. PTN has a high affinity for both
present) for appropriate articles addressing the significance of low and over-sulfated CS chains, although the binding is more
pleiotrophin in bone repair. The key words, which were searched, intense with highly sulfated CS-D and CS-E [86]. An increase of
were the terms ‘‘angiogenesis’’, ‘‘bone’’, ‘‘bone repair’’, ‘‘osteo- total sulfated GAGs was observed after supraspinatus tendon
blasts’’, ‘‘osteoclasts’’ and ‘‘pleiotrophin’’, in different combina- overuse, which correlated with an increase in PTN protein levels,
tions. Searching of the reference lists of potentially relevant origin suggesting that increased GAGs may sequester PTN, which may
was also performed. Inclusion criteria were: papers written in thus initiate a shift towards the chondrocyte phenotype [20]. Data
English, peer-reviewed journals, in vitro and in vivo studies that that correlate anb3 or GAGs with bone remodelling are presented
evaluated the implication of PTN and/or its receptors in bone in Table 1.
regeneration process or its involvement in diseases of the skeletal Nucleolin, a 100 kDa multifunctional protein, is considered to
system which are associated with bone remodelling defects. be a low affinity receptor for PTN [87,88]. Cell surface nucleolin
Exclusion criteria were: articles using language other than English, interacts with both RPTPb/z and avb3 [76] and is required for PTN
letters and expert opinion publications. Articles that conformed to [78]- and VEGF [89]-induced cell migration. Finally, anaplastic
the above mentioned criteria were retrieved and all of the studies lymphoma kinase (ALK), a 220 kDa receptor tyrosine kinase, has
related to these were extensively searched. been also mentioned as PTN receptor, although more recent
studies interrogate direct binding of PTN to ALK and favour the
Results notion that ALK is indirectly activated by PTN through PTN-
dependent inactivation of RPTPb/z [90]. There are no studies on
The search strategies yielded a total of over 200 potential the role of nucleolin or ALK in bone pathophysiology.
articles. During the selection process, the articles were excluded by
title or by abstract, because they were clearly irrelevant to the Regulation of PTN expression
study question. The papers, which fulfil the inclusion criteria, could
be distributed in four main categories. The first category comprised PTN expression has been described as regulated by several
the articles concerning the in vitro or in vivo expression of PTN factors, including growth factors, cytokines, hormones, signalling
[6,8–15] or its receptors [8,9,16–20] in cells of the bone or the molecules, transcription factors and miscellaneous conditions. One
cartilage tissue. The second category included the studies, which of the first studies showed up-regulation of PTN by PDGF-AB in
1818 M. Lamprou et al. / Injury, Int. J. Care Injured 45 (2014) 1816–1823

Table 1 all important stimulators of angiogenesis, increase PTN expression


Existing evidence on a possible role of the known PTN receptors in bone
through activation of the transcription factor activator protein-1.
pathophysiology.
Treatment of odontoblast-like M06-G3 cells with recombinant BMPs
PTN Skeletal effect 2 and 7 that are clinically available for improvement of fracture
receptor
healing, statistically increased PTN expression, while treatment with
Syndecans N-syndecan expression is increased parallel to PTN expression BMP4 had the opposite effect [53]. Finally, mechanical loading
in the periosteum accompanying the decreases PTN expression in primary osteoblasts and osteoblastic
regenerative bone response in an adjuvant-induced
SaOs-2 cells [54].
injury model [8].
Syndecan 4 is functionally involved in endochondral
ossification, and fracture healing is markedly delayed in PTN and angiogenesis
syndecan 4 knockout mice and accompanied by
increased callus formation [16].
Soluble PTN has been suggested to promote endothelial cells
RPTPb/z RPTPb/z-deficient mice display a decreased trabecular bone proliferation [55,56], although this has not been observed in all
volume at the age of 50 weeks, caused by a types of endothelial cells [57]. Besides soluble PTN, it has been also
reduced bone formation rate [17].
shown that immobilised PTN stimulates proliferation of several
RPTPb/z-deficient calvarial osteoblasts display decreased
expression of osteoblast markers and a types of endothelial cells not responding to the soluble growth
diminished potential to form osteocyte-like cellular factor [57]. On the other hand, PTN down-regulates the stimulatory
extensions on Matrigel-coated surfaces [17]. effect of VEGF on endothelial cell proliferation through direct
PTPRZ1 gene is over-expressed in samples of binding of the two growth factors [58].
osteosarcoma patients versus normal bone, but with
no correlation between PTPRZ1 gene expression profile,
PTN has been also shown to promote migration of endothelial
clinicopathological parameters and survival [18]. cells [56,59,60,75,76], and to mediate the stimulatory effect of
It is expressed by chondrocytes and subchondral hydrogen peroxide [52], endothelial nitric oxide synthase/nitric
osteocytes in osteoarthritis patients [9]. oxide [52] and aprotinin [70] on human endothelial cell
anb3 Mediates the anabolic effects of mechanical stimulation migration, through its receptor RPTPb/z. On the other hand,
in osteoblasts [28]. PTN decreases VEGF-induced migration of human endothelial
It is critical to osteoclast function and development cells to the levels of its own migratory effect [58,62,63,77],
[19,29].
suggesting that it may also play a regulatory role to limit
It is a central molecule for osteoclastic bone resorption
[30]. excessive angiogenic response.
Besides the effect of PTN on endothelial cells proliferation and
GAGs Artificial extracellular matrices composed of collagen I
migration, PTN has been shown to induce embryoid body
and synthetically over-sulfated GAGs reveal significant pro-
osteogenic effect, as determined by tissue angiogenesis [64] and promote in vitro formation of tube-like
nonspecific alkaline phosphatase activity and calcium structures in collagen gels [56,59], fibrin gels or matrigel [59] and
deposition, especially when tested in early osteoblasts to stimulate in vivo angiogenesis of the chick embryo chorioallan-
[31].
toic membrane (CAM) [59,78] and in matrigel implants in mice
An increase of total sulfated GAGs was observed 4 weeks
after supraspinatus tendon overuse, which remained elevated
[65]. In the latter assay, PTN also limits VEGF-induced angiogenic
up to 16 weeks [20]. response to the levels of its own smaller stimulatory effect [58],
further supporting its regulatory effect.
Nucleolin No studies available.
It is also possible that besides a direct effect on endothelial cells,
ALK No studies available. PTN has an indirect effect, potentiating the angiogenic effect of other
molecules. For example, PTN up-regulates the mRNA levels of VEGF
isoforms 165 and 190, as well as activates metalloproteinase 2 in the
chick embryo CAM [78] and promotes VEGF expression and
NIH-3T3 cells [42]. PDGF-BB was also found to increase PTN mRNA cooperates with VEGF in promoting colorectal cancer angiogenesis
levels in hepatic stellate cells, one of the major effector cell types [66]. PTN induces proliferation of human peripheral blood
during the repair process of liver tissue injury [43]. Basic FGF mononuclear cells [67] and increases the mRNA expression of the
(bFGF) also increases PTN mRNA levels in NIH3T3 cells [42], as well VEGF receptor 1 in endothelial cell cultures [62]. Flt-1 is expressed
as ptn gene transcription and PTN protein levels in human prostate by monocytes and its expression is increased after monocyte
cancer LNCaP cells, through NAD(P)H oxidase-dependent hydro- activation [68]. In the same line, PTN induces transformation of
gen peroxide production, phosphorylation of ERK1/2 and p38, and monocytes into functional endothelial cells, thus supporting
activation of AP-1. Interestingly, in these cells PTN seems to angiogenesis [69,70].
mediate bFGF-induced cell proliferation and migration [44]. The role of PTN in tumour angiogenesis has been initially
1a,25-Dihydroxyvitamin D3 decreases PTN mRNA levels in the suggested by the observation that culture supernatants derived
osteoblast-like cell lines MC3T3-E1 and ROS17/2.8, whereas in the from PTN transfected human adrenal carcinoma cells, lung cancer
non-osteoblastic cell line, ROS 25/1, it increases PTN mRNA levels. cells or PTN transfected MCF-7 human breast cancer cells possess
This effect was found specific to 1a,25-dihydroxyvitamin D3, mitogenic activities for endothelial cells. PTN also increases the
suggesting that PTN may play a role in vitamin D-dependent angiogenic potential of multiple myeloma. Ribozyme targeting of
regulation of calcium and phosphate homeostasis and for bone PTN in a human melanoma cell line decreases vessel formation in
remodelling [45]. Androgens and estrogens, which have direct the primary tumour, as well as metastases [2]. Antisense PTN
beneficial effects on bone, also increase PTN expression in prostate expression in human prostate LNCaP cells decreases prostate
cells [46–48]. In cultured mouse fibroblasts, PTN expression was cancer cell-induced angiogenesis in vitro and in vivo [71,72]. On
found to be up-regulated by tumour necrosis factor alpha (TNF-a) the other hand, antisense PTN expression in rat glioma C6 cells
and to a smaller extent by epidermal growth factor [49]. TNF-a decreases glioma cell-induced angiogenesis in vitro and in vivo
seems to accelerate bone fracture healing through the coordinate [73] and PTN seems to act as an angiostatic factor in an in vivo
regulation of the expression of specific matrix metalloproteinases neuroblastoma model that is resistant to irinotecan [74]. In both
and angiogenic factors [50]. Hypoxia [43], signalling levels of cases, its regulatory effect on VEGF actions has been discussed as
hydrogen peroxide [51] and endothelial nitric oxide synthase [52], the most likely mechanism.
M. Lamprou et al. / Injury, Int. J. Care Injured 45 (2014) 1816–1823 1819

PTN and bone Osteoporosis


The highest expression of PTN in PTN transgenic mice is
There are several studies that support a potential role of PTN in maintained in the periosteum, and the PTN transgenic mice
the skeletal system, outlined in Table 2. Initially, PTN mRNA was develop a phenotype characterised by an increased bone thickness
found during development in bone and cartilage progenitors and [8]. In transgenic mice over-expressing the human PTN gene driven
in dental pulp [6,10]. Later, PTN was detected within the by the osteocalcin promoter, femoral bone mineral content was
epiphyseal plate of mouse suggesting a possible role in the increased compared with wild-type controls. In ovariectomised
longitudinal bone growth and bone formation [11]. One biological mice, bone mass loss due to oestrogen deficiency was observed in
function that was early attributed to PTN is the promotion of both transgenic and control mice but bone mass was higher in
osteoblast attachment to the extracellular bone matrix through its transgenic mice [32]. In a later study using the same transgenic
carboxy-terminal domain [21]. This was further verified in several line, the bone mass increase was found evident in female, but not
in vivo models, in which PTN was found to be prominently male mice, although no direct evidence supporting female-specific
expressed in the cell matrices that act as target substrates for bone mRNA synthesis of the transgene was obtained [34]. In both cases,
formation and to possibly play an important role in bone the mechanism seems to be through activation and recruitment of
formation, probably by mediating recruitment and attachment osteoblasts. In a more detailed analysis, it was shown that PTN
of osteoblasts/osteoblast precursors to the appropriate substrates over-expression enhanced intramembranous bone formation and
for deposition of new bone [8]. Unpublished observations of had multiple effects on long-term bone growth. The pubertal
our research team in an animal model suggest that PTN is growth spurt did not take place in transgenic mice, in which the
expressed by chondrocytes in the hypertrophic and proliferating growth trajectory was steady and continuous until 25 weeks. By
zones of the growth plate and its expression depends on 30 weeks, transgenic and control mice were of the same size, but
mechanical loading. the calcium content/mg bone was approximately 10% higher in the
transgenic group. PTN was also localised in growth plate and
PTN pathways in clinical skeletal diseases articular chondrocytes, but only in transgenic mice. In the latter, an
encroachment of subchondral bone into the articular cartilage was
The relevance of PTN implication in skeletal homeostasis and also observed [33].
diseases is currently under investigation. PTN is thought to affect PTN-deficient mice seem to have normal bone formation [37],
osteoblasts’ rather than osteoclasts’ functions; however, the but they show growth retardation in the weight-bearing bones by
mechanisms involved are not yet fully elucidated. two months of age and low bone formation and osteopenia, as well

Table 2
Outline of evidence supporting a possible role for PTN in bone remodelling/repair.

Species Findings

PTN expression
Mouse PTN mRNA in calvarial osteoblast-enriched cells [6].
Human PTN mRNA in cDNA libraries from osteosarcoma cell line MG63 [6].
Rat PTN mRNA in bone and cartilage progenitors and in dental pulp during development [10].
Rat Abundant PTN expression in the secondary ossification centre, in the growth plate, and in the periosteum [8].
Rat Up-regulated PTN expression on the surface of adult damaged bone in a post arthritic ossification model [8].
Mouse Increased PTN expression in the periosteum of adult PTN transgenic mice and by the osteocytes dispersed in the
cortical bone [8].
Rat Immunolocalised PTN on both osteoblasts and endothelial cells in the well vascularised newly formed woven bone [11].
Mouse Increased PTN expression in the tibias of female animals after four days of mechanical stimulation [12].
Mouse Greater PTN expression in juvenile compared with the adult dura mater [13].
Human Increased PTN in serum of patients during fracture healing. Not observed in non union patients [14].
Human Over-expressed PTN in the synovial fluid of patients with osteoarthritis [15].
Human PTN in chondrocytes, osteocytes and bone lining cells in the joints of osteoarthritis patients [9].

In vitro cells/cell cultures


Human PTN binds on the surface of the osteoblast-like osteosarcoma cell lines HOS(TE85) and MG-63, and promotes osteoblast
attachment to the extracellular bone matrix [21].
Rat, Human PTN induces migration of osteoblast-type cells [8].
Human PTN promotes adhesion, migration, expansion and differentiation of osteoprogenitor cells [22].
Human PTN has chemotactic effects on both osteoblastic and endothelial cells [23].
Human PTN is an autocrine growth factor involved in the clustering and proliferation of chondrocytes [24].
Mouse PTN induces heparin-binding epidermal growth factor release to trans-activate epidermal growth factor receptor in primary
osteoblasts and osteoblast-like MC3T3-E1 cells, and through this pathway, it increases alkaline phosphatase activity and
inhibits dexamethazone-induced cell death in both cell lines [25].
Human PTN induces hypertrophy during chondrogenic differentiation of bone marrow stem cells [26].

Animal models
PTN transgenic mouse Increased thickness of the cortical bone, increased cortical and cancellous bone volume, and increased osteoblast surface.
No alteration in the bone marrow area [8].
Increased femoral bone mineral content compared with wild-type controls.
In ovariectomised mice, bone mass loss was lower in transgenic mice [32].
Enhanced intramembranous bone formation and multiple effects on long-term bone growth [33].
Bone mass increase in female but not male mice [34].
Bone healing process is impaired in the adult PTN mice [35].

PTN knockout mouse Growth retardation in the weight-bearing bones by two months of age and low bone formation and osteopenia, as well as
resistance to immobilization-dependent bone remodelling during adulthood [36].
The number, morphology, and function of osteoclasts, osteoblasts, and osteocytes are not altered in PTN-deficient mice
compared with wild-type littermates [37].
1820 M. Lamprou et al. / Injury, Int. J. Care Injured 45 (2014) 1816–1823

as resistance to immobilization-dependent bone remodelling [13]. In the same line, PTN serum levels increased asymptotically in
during adulthood [36]. timely fracture healing, while no such increase was detected
A microarray based identification of osteoporosis-related genes during delayed healing [14]. On the other hand, it has been also
in primary culture of human osteoblasts suggested that PTN is shown that the bone healing process was impaired in adult mice
among the candidate genes for further studies in the assessment of over-expressing PTN only in bone and this has been attributed to
the genetic susceptibility to osteoporosis [27]. In a recent clinical inhibitory effects of PTN over-expression on BMP-2 mediated bone
study of 530 postmenopausal women, with and without hip induction [35].
fractures, it was shown that the PTN gene promoter polymorphism
1227C > T and CT haplotype could contribute to the genetic Osteoarthritis
background of osteoporosis, affecting the bone mass density values Studies on the possible involvement of PTN in the pathogenesis of
in the lumbar spine and in the femoral neck [41]. osteoarthritis are limited. PTN is over-expressed in the synovial fluid
Interestingly, the microgravity-induced bone loss is found to be [15], the serum, as well as in chondrocytes and subchondral bone
due to an increased bone resorption and a decreased bone osteocytes of patients with osteoarthritis [9]. Specifically, PTN
deposition. PTN over-expressing mice exposed to normal gravity expression was increased in OA patients graded Ahlback II and III,
present a poorer trabecular organization than wild type mice but the localised in superficial chondrocyte clusters and chondrocytes in the
expression of PTN during the flight offers some protection against upper radial and tidemark zones. PTN was also immunolocalised in
microgravity’s negative effects, as a result of a higher osteoblast the osteocytes, bone lining cells and the extracellular matrix of the
activity in the flight mice [38]. A subsequent study showed that the subchondral bone, as well as in the osteocytes of deeper trabeculae
reduction in the expression of collagen type I and osteocalcin in PTN [9]. These data make PTN a promising candidate for further studies
transgenic mice was less than in the samples from wild type mice, as far as developing therapeutic schemes for osteoarthritis.
possibly due to the higher level calcitonin expression that could
participate to the protective effect of PTN over-expression on the Discussion
bone damage [39].
Bone is characterised by an increased potential for growth,
Fracture healing regeneration and remodelling throughout life. Bone repair is a
Fracture healing is a complex physiological process in which complex process of biological events regulated by specific cells, the
PTN may play an important role. During fracture healing, PTN is extracellular matrix, distinct growth factors and a variety of
immunolocalised on both osteoblasts and endothelial cells in the hormones [92–100]. Despite the increased intrinsic capacity of
well vascularised, newly formed woven bone [40] and recombi- bone to restore fracture healing without scar formation, non-union
nant human PTN has chemotactic effects on both human may still occur in 5–10% of the cases [101]. This impaired fracture
osteoblastic and endothelial cells [23]. A role for PTN in bone healing phenomenon has been attributed to a number of factors,
regeneration is also supported by data showing that PTN induces including poor mechanical stability, extensive soft tissue and
hypertrophy during chondrogenic differentiation of human bone periosteal disruption, patient related co-morbidities, non-steroidal
marrow stem cells, as evidenced by increased expression of anti-inflammatory drugs, vitamin D deficiency and malnutrition
collagen 2 protein and increased transcription of hypertrophic amongst others [102,103]. Current research to understand better
chondrocyte markers, such as MMP13, collagen 10 and alkaline and to treat successfully fracture non-union is focusing on
phosphatase and enhanced calcification and content of collagen biological based approaches and involves the implantation of
10 protein [26]. Additionally, our unpublished data demonstrate growth factors, osteoprogenitor cells, biophysical stimuli, and the
that PTN is expressed in chondrocytes during callus formation promotion of local angiogenesis [104].
(Fig. 1). PTN expression is greater in juvenile compared with the Angiogenesis is a critical step of bone healing. During the bone
adult dura mater and may be related with the reossification of repair process in humans, several angiogenic factors are expressed
large calvarial defects that is possible in children but not adults and play key regulatory roles during the early phase of fracture
healing. PTN is a potent angiogenic factor that may also affect
several aspects of bone remodelling. Its importance in fracture
healing has been supported by the study of Weiss et al. (2009) in
patients with and without fracture union [14]. Compared to the
relatively low serum concentrations in non-injured adults,
systemic PTN values show a prolonged increase during physiolog-
ical fracture healing and remodelling, while no such increase could
be detected in non-union patients. Therefore, PTN merits further
investigation as a possible powerful bone-inducing component to
promote healing in orthopaedic and spinal surgery by affecting
bone cells themselves or indirectly, by regulating angiogenesis.
In all cases, since PTN may have multiple effects in other tissues,
as well, the perspective of exclusively targeting the bone tissue is
critical, so as to prevent potentially side effects at non skeletal sites.
PTN could be locally administered in implanted carriers such as
scaffolds, membranes for tissue-guided regeneration or hydrogels,
providing different schemes for release, in cases such as long bone
open fractures treated with intramedullary fixation, non-united
Fig. 1. Expression of PTN by chondrocytes during healing process, five weeks after a fractures, and in spine surgery for spinal fusion. Concerning the
rat’s vertebrae fracture. PTN shows both cytoplasmic (red arrows) and nuclear required dose, clinical trials are desirable in order to monitor
(black arrows) localization (brown staining), with the latter being dominant. carefully the appropriate local PTN concentration leading to a
Although there is no known physiological significance for the nuclear localization of
PTN, it has been also observed in endothelial and glioma cells [78], cardiomyocytes
sufficient bone formation. Alternatively, apart from direct admin-
[91] and osteoarthritic chondrocytes [9]. (For interpretation of the references to istration of PTN, indirect ways of endogenous PTN regulation could
color in this text, the reader is referred to the web version of the article.) be another possible target.
M. Lamprou et al. / Injury, Int. J. Care Injured 45 (2014) 1816–1823 1821

An important issue that should be taken into consideration is the [12] Xing W, Baylink D, Kesavan C, Hu Y, Kapoor S, Chadwick RB, et al. Global gene
expression analysis in the bones reveals involvement of several novel genes
fact that it is becoming increasingly apparent that growth factors act and pathways in mediating an anabolic response of mechanical loading in
in a coordinated cascade of events to restore bone; therefore, mice. J Cell Biochem 2005;96:1049–60.
delivering combinations of growth factors may be of greater [13] Wan DC, Aalami OO, Wang Z, Nacamuli RP, Lorget F, Derynck R, et al.
Differential gene expression between juvenile and adult dura mater: a
significance in the clinic. Concerning PTN, its use in combination window into what genes play a role in the regeneration of membranous
with osteogenic growth factors, such as BMPs, may prove beneficial. bone. Plast Reconstr Surg 2006;118:851–61.
It has been shown that fracture healing was impaired in the adult [14] Weiss S, Zimmermann G, Pufe T, Varoga D, Henle P. The systemic angiogenic
response during bone healing. Arch Orthop Trauma Surg 2009;129:989–97.
mouse with targeted PTN over-expression in bone, and this was [15] Pufe T, Bartscher M, Petersen W, Tillmann B, Mentlein R. Pleiotrophin an
attributed to the inhibitory effects of PTN over-expression on BMP-2 embryonic differentiation and growth factor, is expressed in osteoarthritis.
mediated bone induction [35]. Taking into account that BMP-2 up- Osteoarthritis Cartilage 2003;11:260–4.
[16] Bertrand J, Stange R, Hidding H, Echtermeyer F, Nalesso G, Godmann L, et al.
regulates PTN expression [53], these data favour the notion of a
Syndecan 4 supports bone fracture repair, but not fetal skeletal development,
regulatory role of PTN in the pathways of primary signalling in mice. Arthritis Rheum 2013;65:743–52.
molecules, such as BMPs, leading to the hypothesis that it may [17] Schinke T, Gebauer M, Schilling AF, Lamprianou S, Priemel M, Mueldner C,
regulate complications of BMPs, such as heterotopic ossifications or et al. The protein tyrosine phosphatase Rptpzeta is expressed in differen-
tiated osteoblasts and affects bone formation in mice. Bone 2008;42:
hardware failure secondary to non-union. A similar regulatory role 524–34.
of PTN has been shown for the key angiogenic growth factor VEGF [18] Toledo SR, Oliveira ID, Okamoto OK, Zago MA, de Seixas Alves MT, Filho RJ,
[2,3,58,63], further supporting a potential regulatory role of PTN for et al. Bone deposition, bone resorption, and osteosarcoma. J Orthop Res
2010;28:1142–8.
both angiogenesis and bone formation and/or repair. A future [19] Zou W, Teitelbaum SL. Integrins, growth factors, and the osteoclast cytoskel-
application of PTN in combination with other growth factors, e.g. eton. Ann N Y Acad Sci 2010;1192:27–31.
BMPs, may thus contribute to the clinically desired bone formation, [20] Attia M, Scott A, Duchesnay A, Carpentier G, Soslowsky LJ, Huynh MB, et al.
Alterations of overused supraspinatus tendon: a possible role of glycosami-
which has not been fully attained up to date with the application of a noglycans and HARP/pleiotrophin in early tendon pathology. J Orthop Res
single growth factor. Although the issues to be unravelled are 2012;30:61–71.
multiple in such a case and are related to the appropriate and safe [21] Gieffers C, Engelhardt W, Brenzel G, Matsuishi T, Frey J. Receptor binding of
osteoblast-specific factor 1 (OSF-1/HB-GAM) to human osteosarcoma cells
concentration of each growth factor, rate of release and timing of promotes cell attachment. Eur J Cell Biol 1993;62:352–61.
application, this approach may better mimic the physiological [22] Yang X, Tare RS, Partridge KA, Roach HI, Clarke NM, Howdle SM, et al.
situation and deserves further consideration and appropriate Induction of human osteoprogenitor chemotaxis, proliferation, differentia-
tion, and bone formation by osteoblast stimulating factor-1/pleiotrophin:
studies.
osteoconductive biomimetic scaffolds for tissue engineering. J Bone Miner
In conclusion, the heparin-binding angiogenic growth factor Res 2003;18:47–57.
PTN appears to play a well-defined and significant role in both [23] Li G, Cui Y, McIlmurray L, Allen WE, Wang H. rhBMP-2, rhVEGF(165), rhPTN
physiological and pathological angiogenesis. Emerging evidence and thrombin-related peptide, TP508 induce chemotaxis of human osteo-
blasts and microvascular endothelial cells. J Orthop Res 2005;23:680–5.
supports the view that it plays a crucial role in bone repair. Based [24] Pufe T, Groth G, Goldring MB, Tillmann B, Mentlein R. Effects of pleiotrophin,
on its molecular properties, future studies must investigate the a heparin-binding growth factor, on human primary and immortalized
potential of its application in clinical situations were bone chondrocytes. Osteoarthritis Cartilage 2007;15:155–62.
[25] Fan JB, Liu W, Yuan K, Zhu XH, Xu DW, Chen JJ, et al. EGFR trans-activation
restoration is desirable. mediates pleiotrophin-induced activation of Akt and Erk in cultured osteo-
blasts. Biochem Biophys Res Commun 2014;447:425–30.
Conflict of interest statement [26] Bouderlique T, Henault E, Lebouvier A, Frescaline G, Bierling P, Rouard H, et al.
Pleiotrophin commits human bone marrow mesenchymal stromal cells
towards hypertrophy during chondrogenesis. PLOS ONE 2014;9:e88287.
The authors report no conflict of interest. [27] Trost Z, Trebse R, Prezelj J, Komadina R, Logar DB, Marc J. A microarray based
identification of osteoporosis-related genes in primary culture of human
References osteoblasts. Bone 2010;46:72–80.
[28] Rangaswami H, Schwappacher R, Marathe N, Zhuang S, Casteel DE, Haas B,
et al. Cyclic GMP and protein kinase G control a Src-containing mechanosome
[1] Pountos I, Panteli M, Panagiotopoulos E, Jones E, Giannoudis PV. Can we enhance in osteoblasts. Sci Signal 2010;3:ra91.
fracture vascularity: what is the evidence? Injury 2014;45(Suppl. 2):S49–57. [29] Schneider JG, Amend SR, Weilbaecher KN. Integrins and bone metastasis:
[2] Papadimitriou E, Mikelis C, Lampropoulou E, Koutsioumpa M, Theochari K, integrating tumor cell and stromal cell interactions. Bone 2011;48:54–65.
Tsirmoula S, et al. Roles of pleiotrophin in tumor growth and angiogenesis. [30] Nakamura I, Duong le T, Rodan SB, Rodan GA. Involvement of alpha(v)beta3
Eur Cytokine Netw 2009;20:180–90. integrins in osteoclast function. J Bone Miner Metab 2007;25:337–44.
[3] Pantazaka E, Papadimitriou E. PTN (pleiotrophin). Atlas Genet Cytogenet [31] Hempel U, Preissler C, Vogel S, Möller S, Hintze V, Becher J, et al. Artificial
Oncol Haematol 2012;16:821–37. extracellular matrices with oversulfated glycosaminoglycan derivatives pro-
[4] Kilpelainen I, Kaksonen M, Kinnunen T, Avikainen H, Fath M, Linhardt RJ, et al.
mote the differentiation of osteoblast-precursor cells and premature osteo-
Heparin-binding growth-associated molecule contains two heparin-binding
blasts. Biomed Res Int 2014;2014:938368.
beta-sheet domains that are homologous to the thrombospondin type I
[32] Masuda H, Tsujimura A, Yoshioka M, Arai Y, Kuboki Y, Mukai T, et al. Bone
repeat. J Biol Chem 2000;275:13564–70.
mass loss due to estrogen deficiency is compensated in transgenic mice
[5] Englund C, Birve A, Falileeva L, Grabbe C, Palmer RH. Miple1 and miple2
overexpressing human osteoblast stimulating factor-1. Biochem Biophys Res
encode a family of MK/PTN homologues in Drosophila melanogaster. Dev
Commun 1997;238:528–33.
Genes Evol 2006;216:10–8.
[33] Tare RS, Oreffo RO, Sato K, Rauvala H, Clarke NM, Roach HI. Effects of targeted
[6] Tezuka K, Takeshita S, Hakeda Y, Kumegawa M, Kikuno R, Hashimoto-Gotoh
overexpression of pleiotrophin on postnatal bone development. Biochem
T. Isolation of mouse and human cDNA clones encoding a protein expressed
Biophys Res Commun 2002;298:324–32.
specifically in osteoblasts and brain tissues. Biochem Biophys Res Commun
[34] Hashimoto-Gotoh T, Ohnishi H, Tsujimura A, Tsunezuka H, Imai K, Masuda H,
1990;173:246–51.
et al. Bone mass increase specific to the female in a line of transgenic mice
[7] Hankenson KD, Sweetwyne MT, Shitaye H, Posey KL. Thrombospondins and
overexpressing human osteoblast stimulating factor-1. J Bone Miner Metab
novel TSR-containing proteins, R-spondins, regulate bone formation and
2004;22:278–82.
remodeling. Curr Osteoporos Rep 2010;8:68–76.
[35] Li G, Bunn JR, Mushipe MT, He Q, Chen X. Effects of pleiotrophin (PTN) over-
[8] Imai S, Kaksonen M, Raulo E, Kinnunen T, Fages C, Meng X, et al. Osteoblast
expression on mouse long bone development, fracture healing and bone
recruitment and bone formation enhanced by cell matrix-associated heparin-
repair. Calcif Tissue Int 2005;76:299–306.
binding growth-associated molecule (HB-GAM). J Cell Biol 1998;143:1113–28.
[36] Imai S, Heino TJ, Hienola A, Kurata K, Buki K, Matsusue Y, et al. Osteocyte-
[9] Kaspiris A, Mikelis C, Heroult M, Khaldi L, Grivas TB, Kouvaras I, et al.
derived HB-GAM (pleiotrophin) is associated with bone formation and
Expression of the growth factor pleiotrophin and its receptor protein tyrosine
mechanical loading. Bone 2009;44:785–94.
phosphatase beta/zeta in the serum, cartilage and subchondral bone of
[37] Lehmann W, Schinke T, Schilling AF, Catala-Lehnen P, Gebauer M, Pogoda P,
patients with osteoarthritis. Joint Bone Spine 2013;80:407–13.
et al. Absence of mouse pleiotrophin does not affect bone formation in vivo.
[10] Vanderwinden JM, Mailleux P, Schiffmann SN, Vanderhaeghen JJ. Cellular
Bone 2004;35:1247–55.
distribution of the new growth factor pleiotrophin (HB-GAM) mRNA in
[38] Tavella S, Ruggiu A, Giuliani A, Brun F, Canciani B, Manescu A, et al. Bone
developing and adult rat tissues. Anat Embryol (Berl) 1992;186:387–406.
turnover in wild type and pleiotrophin-transgenic mice housed for three
[11] Petersen W, Rafii M. Immunolocalization of the angiogenetic factor pleiotrophin
months in the International Space Station (ISS). PLoS ONE 2012;7:e33179.
(PTN) in the growth plate of mice. Arch Orthop Trauma Surg 2001;121:414–6.
1822 M. Lamprou et al. / Injury, Int. J. Care Injured 45 (2014) 1816–1823

[39] Albi E, Curcio F, Spelat R, Lazzarini A, Lazzarini R, Cataldi S, et al. Loss of beta/zeta in vascular endothelial growth factor-induced endothelial cell
parafollicular cells during gravitational changes (microgravity, hypergravity) migration (Abstract). Angiogenesis 2014;17:754.
and the secret effect of pleiotrophin. PLoS ONE 2012;7:e48518. [64] Magnusson PU, Dimberg A, Mellberg S, Lukinius A, Claesson-Welsh L. FGFR-1
[40] Petersen W, Wildemann B, Pufe T, Raschke M, Schmidmaier G. The angiogenic regulates angiogenesis through cytokines interleukin-4 and pleiotrophin.
peptide pleiotrophin (PTN/HB-GAM) is expressed in fracture healing: an Blood 2007;110:4214–22.
immunohistochemical study in rats. Arch Orthop Trauma Surg 2004;124: [65] Bernard-Pierrot I, Delbe J, Caruelle D, Barritault D, Courty J, Milhiet PE. The
603–7. lysine-rich C-terminal tail of heparin affin regulatory peptide is required for
[41] Mencej-Bedrac S, Prezelj J, Komadina R, Vindisar F, Marc J. 1227C > T mitogenic and tumor formation activities. J Biol Chem 2001;276:12228–34.
polymorphism in the pleiotrophin gene promoter influences bone mineral [66] Kong Y, Bai PS, Nan KJ, Sun H, Chen NZ, Qi XG. Pleiotrophin is a potential
density in postmenopausal women. Mol Genet Metab 2011;103:76–80. colorectal cancer prognostic factor that promotes VEGF expression and
[42] Li YS, Gurrieri M, Deuel TF. Pleiotrophin gene expression is highly restricted induces angiogenesis in colorectal cancer. Int J Colorectal Dis 2012;27:
and is regulated by platelet-derived growth factor. Biochem Biophys Res 287–98.
Commun 1992;184:427–33. [67] Achour A, Laaroubi D, Caruelle D, Barritault D, Courty J. The angiogenic factor
[43] Antoine M, Tag CG, Wirz W, Borkham-Kamphorst E, Sawitza I, Gressner AM, heparin affin regulatory peptide (HARP) induces proliferation of human
et al. Upregulation of pleiotrophin expression in rat hepatic stellate cells by peripheral blood mononuclear cells. Cell Mol Biol (Noisy-le-grand)
PDGF and hypoxia: implications for its role in experimental biliary liver 2001;47 Online Pub:OL73–7.
fibrogenesis. Biochem Biophys Res Commun 2005;337:1153–64. [68] Shibuya M. Structure and dual function of vascular endothelial growth factor
[44] Hatziapostolou M, Polytarchou C, Katsoris P, Courty J, Papadimitriou E. receptor-1 (Flt-1). Int J Biochem Cell Biol 2001;33:409–20.
Heparin affin regulatory peptide/pleiotrophin mediates fibroblast growth [69] Sharifi BG, Zeng Z, Wang L, Song L, Chen H, Qin M, et al. Pleiotrophin induces
factor 2 stimulatory effects on human prostate cancer cells. J Biol Chem transdifferentiation of monocytes into functional endothelial cells. Arterios-
2006;281:32217–26. cler Thromb Vasc Biol 2006;26:1273–80.
[45] Tamura M, Ichikawa F, Guillerman RP, Deuel TF. Nodal M.1a,25-dihydrox- [70] Chen H, Campbell RA, Chang Y, Li M, Wang CS, Li J, et al. Pleiotrophin
yvitamin D(3) down-regulates pleiotrophin messenger RNA expression in produced by multiple myeloma induces transdifferentiation of monocytes
osteoblast-like cells. Endocrine 1995;3:21–4. into vascular endothelial cells: a novel mechanism of tumor-induced vascu-
[46] Vacherot F, Laaroubi K, Caruelle D, Delbe J, Barritault D, Caruelle JP, et al. logenesis. Blood 2009;113:1992–2002.
Upregulation of heparin-affin regulatory peptide by androgen. In Vitro Cell [71] Hatziapostolou M, Delbe J, Katsoris P, Polytarchou C, Courty J, Papadimitriou
Dev Biol Anim 1995;31:647–8. E. Heparin affin regulatory peptide is a key player in prostate cancer cell
[47] Orr B, Vanpoucke G, Grace OC, Smith L, Anderson RA, Riddick AC, et al. growth and angiogenicity. Prostate 2005;65:151–8.
Expression of pleiotrophin in the prostate is androgen regulated and it [72] Tsirmoula S, Dimas K, Hatziapostolou M, Lamprou M, Ravazoula P, Papadi-
functions as an autocrine regulator of mesenchyme and cancer associated mitriou E. Implications of pleiotrophin in human PC3 prostate cancer cell
fibroblasts and as a paracrine regulator of epithelia. Prostate 2011;71: growth in vivo. Cancer Sci 2012;103:1826–32.
305–317. [73] Parthymou A, Lampropoulou E, Mikelis C, Drosou G, Papadimitriou E. Heparin
[48] Lee JY, Jeong W, Lim W, Kim J, Bazer FW, Han JY, et al. Chicken pleiotrophin: affin regulatory peptide/pleiotrophin negatively affects diverse biological
regulation of tissue specific expression by estrogen in the oviduct and distinct activities in C6 glioma cells. Eur J Cell Biol 2008;87:17–29.
expression pattern in the ovarian carcinomas. PLoS ONE 2012;7:e34215. [74] Calvet L, Geoerger B, Regairaz M, Opolon P, Machet L, Morizet J, et al.
[49] Pufe T, Bartscher M, Petersen W, Tillmann B, Mentlein R. Expression of Pleiotrophin a candidate gene for poor tumor vasculature and in vivo
pleiotrophin, an embryonic growth and differentiation factor, in rheumatoid neuroblastoma sensitivity to irinotecan. Oncogene 2006;25:3150–9.
arthritis. Arthritis Rheum 2003;48:660–7. [75] Mikelis C, Sfaelou E, Koutsioumpa M, Kieffer N, Papadimitriou E. Integrin
[50] Lehmann W, Edgar CM, Wang K, Cho TJ, Barnes GL, Kakar S, et al. Tumor alpha(v)beta(3) is a pleiotrophin receptor required for pleiotrophin-induced
necrosis factor alpha (TNF-alpha) coordinately regulates the expression of endothelial cell migration through receptor protein tyrosine phosphatase
specific matrix metalloproteinases (MMPS) and angiogenic factors during beta/zeta. FASEB J 2009;23:1459–69.
fracture healing. Bone 2005;36:300–10. [76] Koutsioumpa M, Polytarchou C, Courty J, Zhang Y, Kieffer N, Mikelis C, et al.
[51] Polytarchou C, Hatziapostolou M, Papadimitriou E. Hydrogen peroxide sti- Interplay between alphavbeta3 integrin and nucleolin regulates human
mulates proliferation and migration of human prostate cancer cells through endothelial and glioma cell migration. J Biol Chem 2013;288:343–54.
activation of activator protein-1 and up-regulation of the heparin affin [77] Polykratis A, Delbé J, Courty J, Papadimitriou E, Katsoris P. Identification of
regulatory peptide gene. J Biol Chem 2005;280:40428–35. heparin affin regulatory peptide domains with potential role on angiogenesis.
[52] Polytarchou C, Hatziapostolou M, Poimenidi E, Mikelis C, Papadopoulou A, Int J Biochem Cell Biol 2004;36:1954–66.
Parthymou A, et al. Nitric oxide stimulates migration of human endothelial [78] Koutsioumpa M, Drosou G, Mikelis C, Theochari K, Vourtsis D, Katsoris P, et al.
and prostate cancer cells through up-regulation of pleiotrophin expression Pleiotrophin expression and role in physiological angiogenesis in vivo:
and its receptor protein tyrosine phosphatase b/z. Int J Cancer potential involvement of nucleolin. Vasc Cell 2012;4:4.
2009;124:1785–93. [79] Kinnunen T, Raulo E, Nolo R, Maccarana M, Lindahl U, Rauvala H. Neurite
[53] Erlandsen H, Ames JE, Tamkenath A, Mamaeva O, Stidham K, Wilson ME, et al. outgrowth in brain neurons induced by heparin-binding growth-associated
Pleiotrophin expression during odontogenesis. J Histochem Cytochem molecule (HB-GAM) depends on the specific interaction of HB-GAM with
2012;60:366–75. heparan sulfate at the cell surface. J Biol Chem 1996;271:2243–8.
[54] Liedert A, Augat P, Ignatius A, Hausser HJ, Claes L. Mechanical regulation [80] Raulo E, Chernousov MA, Carey DJ, Nolo R, Rauvala H. Isolation of a neuronal
of HB-GAM expression in bone cells. Biochem Biophys Res Commun cell surface receptor of heparin binding growth-associated molecule (HB-
2004;319:951–8. GAM), identification as N-syndecan (syndecan-3). J Biol Chem 1994;269:
[55] Courty J, Dauchel MC, Caruelle D, Perderiset M, Barritault D. Mitogenic 12999–3004.
properties of a new endothelial cell growth factor related to pleiotrophin. [81] Deepa SS, Yamada S, Zako M, Goldberger O, Sugahara K. Chondroitin sulfate
Biochem Biophys Res Commun 1991;180:145–51. chains on syndecan-1 and syndecan-4 from normal murine mammary gland
[56] Souttou B, Raulais D, Vigny M. Pleiotrophin induces angiogenesis: involve- epithelial cells are structurally and functionally distinct and cooperate with
ment of the phosphoinositide-3 kinase but not the nitric oxide synthase heparin sulfate chains to bind growth factors. A novel function to control
pathways. J Cell Physiol 2001;187:59–64. binding of midkine, pleiotrophin, and basic fibroblast growth factor. J Biol
[57] Papadimitriou E, Heroult M, Courty J, Polykratis A, Stergiou C, Katsoris P. Chem 2004;279:37368–76.
Endothelial cell proliferation induced by HARP: implication of N or C terminal [82] Pantazaka E, Papadimitriou E. Chondroitin sulfate-cell membrane effectors as
peptides. Biochem Biophys Res Commun 2000;274:242–8. regulators of growth factor-mediated vascular and cancer cell migration.
[58] Héroult M, Bernard-Pierrot I, Delbé J, Hamma-Kourbali Y, Katsoris P, Barri- Biochim Biophys Acta 2014;1840:2643–50.
tault D, et al. Heparin affin regulatory peptide binds to vascular endothelial [83] Koutsioumpa M, Papadimitriou E. PG receptors with phosphatase action in
growth factor (VEGF) and inhibits VEGF-induced angiogenesis. Oncogene cancer and angiogenesis. In: Karamanos N, editor. Extracellular matrix:
2004;23:1745–53. pathobiology and signalling. Berlin/Boston: Walter de Gruyter GmbH &
[59] Papadimitriou E, Polykratis A, Courty J, Koolwijk P, Heroult M, Katsoris P. Co. KG; 2012. p. 813–23.
HARP induces angiogenesis in vivo and in vitro: implication of N or C terminal [84] Bernard-Pierrot I, Héroult M, Lemaı̂tre G, Barritault D, Courty J, Milhiet PE.
peptides. Biochem Biophys Res Commun 2001;282:306–13. Glycosaminoglycans promote HARP/PTN dimerization. Biochem Biophys Res
[60] Polykratis A, Katsoris P, Courty J, Papadimitriou E. Characterization of heparin Commun 1999;266:437–42.
affin regulatory peptide signaling in human endothelial cells. J Biol Chem [85] Vacherot F, Delbé J, Heroult M, Barritault D, Fernig DG, Courty J. Glycosami-
2005;280:22454–61. noglycans differentially bind HARP and modulate its biological activity. J Biol
[61] Koutsioumpa M, Hatziapostolou M, Mikelis C, Koolwijk P, Papadimitriou E. Chem 1999;274:7741–7.
Aprotinin stimulates angiogenesis and human endothelial cell migration [86] Mizumoto S, Fongmoon D, Sugahara K. Interaction of chondroitin sulfate and
through the growth factor pleiotrophin and its receptor protein tyrosine dermatan sulfate from various biological sources with heparin-binding
phosphatase beta/zeta. Eur J Pharmacol 2009;602:245–9. growth factors and cytokines. Glycoconj J 2013;30:619–32.
[62] Kokolakis G, Mikelis C, Papadimitriou E, Courty J, Karetsou E, Katsoris P. [87] Take M, Tsutsui J, Obama M, Ozawa M, Nakayama T, Maruyama I, et al. Identifi-
Effect of heparin affin regulatory peptide on the expression of vascular cation of nucleolin as a binding protein for midkine (MK) and heparin-binding
endothelial growth factor receptors in endothelial cells. In Vivo growth associated molecule (HB-GAM). J Biochem 1994;116:1063–8.
2006;20:629–35. [88] Said EA, Courty J, Svab J, Delbe J, Krust B, Hovanessian AG. Pleiotrophin
[63] Koutsioumpa M, Poimenidi E, Theodoropoulou C, Pantazaka E, Skoura A, inhibits HIV infection by binding the cell surface-expressed nucleolin. FEBS J
Megalooikonomou V, et al. A role of receptor protein tyrosine phosphatase 2005;272:4646–59.
M. Lamprou et al. / Injury, Int. J. Care Injured 45 (2014) 1816–1823 1823

[89] Destouches D, El Khoury D, Hamma-Kourbali Y, Krust B, Albanese P, [96] Papanna MC, Al-Hadithy N, Somanchi BV, Sewell MD, Robinson PM, Khan SA,
Katsoris P, et al. Suppression of tumor growth and angiogenesis by a et al. The use of bone morphogenic protein-7 (OP-1) in the management of
specific antagonist of the cell-surface expressed nucleolin. PLoS ONE resistant non-unions in the upper and lower limb. Injury 2012;43:1135–40.
2008;3:e2518. [97] Reyes R, De la Riva B, Delgado A, Hernández A, Sánchez E, Évora C. Effect of
[90] Deuel TF. Anaplastic lymphoma kinase: ligand independent activation me- triple growth factor controlled delivery by a brushite-PLGA system on a bone
diated by the PTN/RPTPb/z signaling pathway. Biochim Biophys Acta defect. Injury 2012;43:334–42.
2013;1834:2219–23. [98] Jungbluth P, Hakimi AR, Grassmann JP, Schneppendahl J, Betsch M, Kröpil P,
[91] Li J, Wei H, Chesley A, Moon C, Krawczyk M, Volkova M, et al. The pro- et al. The early phase influence of bone marrow concentrate on metaphyseal
angiogenic cytokine pleiotrophin potentiates cardiomyocyte apoptosis bone healing. Injury 2013;44:1285–94.
through inhibition of endogenous AKT/PKB activity. J Biol Chem [99] Neunaber C, Yesilkaya P, Pütz C, Krettek C, Hildebrand F. Differentiation
2007;282:34984–93. of osteoprogenitor cells is affected by trauma-haemorrhage. Injury
[92] Buschmann J, Härter L, Gao S, Hemmi S, Welti M, Hild N, et al. Tissue 2013;44:1279–84.
engineered bone grafts based on biomimetic nanocomposite PLGA/amor- [100] Giannoudis PV, Ahmad MA, Mineo GV, Tosounidis TI, Calori GM, Kanakaris
phous calcium phosphate scaffold and human adipose-derived stem cells. NK. Subtrochanteric fracture non-unions with implant failure managed with
Injury 2012;43:1689–97. the diamond concept. Injury 2013;44(Suppl. 1):S76–81.
[93] Calori GM, Colombo M, Mazza E, Ripamonti C, Mazzola S, Marelli N, et al. [101] Tzioupis C, Giannoudis PV. Prevalence of long-bone non-unions. Injury
Monotherapy vs. polytherapy in the treatment of forearm non-unions and 2007;38(Suppl. 2):S3–9.
bone defects. Injury 2013;44(Suppl. 1):S63–9. [102] Copuroglu C, Calori GM, Giannoudis PV. Fracture non-union: who is at risk?
[94] Blokhuis TJ, Calori GM, Schmidmaier G. Autograft versus BMPs for the Injury 2013;44:1379–82.
treatment of non-unions: what is the evidence? Injury 2013;44(Suppl. [103] Pountos I, Georgouli T, Blokhuis TJ, Pape HC, Giannoudis PV. Pharmacological
1):S40–2. agents and impairment of fracture healing: what is the evidence? Injury
[95] Giannoudis PV, Calori GM, Begue T, Schmidmaier G. Bone regeneration strate- 2008;39:384–94.
gies: current trends but what the future holds? Injury 2013;44(Suppl. 1): [104] Pountos I, Georgouli T, Pneumaticos S, Giannoudis PV. Fracture non-union:
S1–2. can biomarkers predict outcome? Injury 2013;44:1725–32.

You might also like