ArticlePDF Available

Osteoblast-specific overexpression of complement receptor C5aR1 impairs fracture healing

PLOS
PLOS ONE
Authors:
  • University of Um, Germany

Abstract and Figures

The anaphylatoxin receptor C5aR1 plays an important role not only in innate immune responses, but also in bone metabolism and fracture healing, being highly expressed on immune and bone cells, including osteoblasts and osteoclasts. C5aR1 induces osteoblast migration, cytokine generation and osteoclastogenesis, however, the exact role of C5aR1-mediated signaling in osteoblasts is not entirely known. Therefore, we hypothesized that osteoblasts are essential target cells for C5a and that fracture healing should be disturbed in mice with an osteoblast-specific C5aR1 overexpression (Col1a1-C5aR1). Osteoblast activity in vitro, bone phenotype and fracture healing after isolated osteotomy and after combined osteotomy with additional thoracic trauma were analyzed. The systemic and local inflammatory reactions were analyzed by determining C5a and IL-6 concentrations in blood, bronchoalveolar lavage fluid and fracture callus and the recruitment of immune cells. In vitro, osteoblast proliferation and differentiation were similar to wildtype cells, and phosphorylation of p38 and expression of IL-6 and RANKL were increased in osteoblasts derived from Col1a1-C5aR1 mice. Bone phenotype and the inflammatory reaction were unaffected in Col1a1-C5aR1 mice. Fracture healing was significantly impaired as demonstrated by significantly reduced bone content, bone mineral density and flexural rigidity, possibly due to significantly increased osteoclast numbers. C5aR1 signaling in osteoblasts might possibly affect RANKL/OPG balance, leading to increased bone resorption. Additional trauma significantly impaired fracture healing, particularly in Col1a1-C5aR1 mice. In conclusion, the data indicate that C5aR1 signaling in osteoblasts plays a detrimental role in bone regeneration after fracture.
Content may be subject to copyright.
RESEARCH ARTICLE
Osteoblast-specific overexpression of
complement receptor C5aR1 impairs fracture
healing
Stephanie Bergdolt
1
,Anna Kovtun
1
,Yvonne Ha
¨gele
1
,Astrid Liedert
1
,Thorsten Schinke
2
,
Michael Amling
2
,Markus Huber-Lang
3
,Anita Ignatius
1
*
1Institute of OrthopedicResearch and Biomechanics, University of Ulm, Ulm, Germany, 2Department of
Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,
3Institute of Clinical and Experimental Traumaimmunology, University of Ulm, Ulm, Germany
*anita.ignatius@uni-ulm.de
Abstract
The anaphylatoxin receptor C5aR1 plays an important role not only in innate immune
responses, but also in bone metabolism and fracture healing, being highly expressed on
immune and bone cells, including osteoblasts and osteoclasts. C5aR1 induces osteoblast
migration, cytokine generation and osteoclastogenesis, however, the exact role of C5aR1-
mediated signaling in osteoblasts is not entirely known. Therefore, we hypothesized that
osteoblasts are essential target cells for C5a and that fracture healing should be disturbed
in mice with an osteoblast-specific C5aR1 overexpression (Col1a1-C5aR1). Osteoblast
activity in vitro,bone phenotype and fracture healing after isolated osteotomy and after com-
bined osteotomy with additional thoracic trauma were analyzed. The systemic and local
inflammatory reactions were analyzed by determining C5a and IL-6 concentrations in blood,
bronchoalveolar lavage fluid and fracture callus and the recruitment of immune cells. In
vitro,osteoblast proliferation and differentiation were similar to wildtype cells, and phosphor-
ylation of p38 and expression of IL-6 and RANKL were increased in osteoblasts derived
from Col1a1-C5aR1 mice. Bone phenotype and the inflammatory reaction were unaffected
in Col1a1-C5aR1 mice. Fracture healing was significantly impaired as demonstrated by sig-
nificantly reduced bone content, bone mineral density and flexural rigidity, possibly due to
significantly increased osteoclast numbers. C5aR1 signaling in osteoblasts might possibly
affect RANKL/OPG balance, leading to increased bone resorption. Additional trauma signifi-
cantly impaired fracture healing, particularly in Col1a1-C5aR1 mice. In conclusion, the data
indicate that C5aR1 signaling in osteoblasts plays adetrimentalrole in bone regeneration
after fracture.
Introduction
In recent years, evidence has accumulated that the complement system, an essential part of
innate immunity, is involved in the regulation of bone turnover and in the pathogenesis of
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 1 / 17
a1111111111
a1111111111
a1111111111
a1111111111
a1111111111
OPEN ACCESS
Citation: Bergdolt S, Kovtun A, Ha¨gele Y, Liedert A,
Schinke T, Amling M, et al. (2017) Osteoblast-
specific overexpression of complement receptor
C5aR1 impairs fracture healing. PLoS ONE 12(6):
e0179512. https://doi.org/10.1371/journal.
pone.0179512
Editor: Jose Manuel Garcia Aznar, University of
Zaragoza, SPAIN
Received: February 17, 2017
Accepted: May 30, 2017
Published: June 14, 2017
Copyright: ©2017 Bergdolt et al. This is an open
access article distributed under the terms of the
Creative Commons Attribution License,which
permits unrestricteduse, distribution, and
reproduction in any medium, provided the original
author and source are credited.
Data AvailabilityStatement: All relevant data are
within the paper.
Funding: The study was funded by the German
Research Foundation (DFG) in the framework of
the Clinical Research Group KFO200 (IG18/14-2)
and the Collaborative Research Center CRC1149
(INST 40/491-1). The funders had no role in study
design, data collection and analysis, decision to
publish, or preparation of the manuscript.
Competing interests:The authors have declared
that no competing interests exist.
bone disorders, including fracture healing. However, the underlying mechanisms are currently
poorly understood [13].
The complement system, anetwork of more than 30 soluble and membrane-bound mole-
cules, serves as apowerful danger-sensing system, which is activated by exogenous and en-
dogenous danger molecules [4]. It can be activated intrinsically by the alternative, lectin or
classical pathway or extrinsically by serine proteases, including thrombin of the coagulation
system [4,5]. All pathways lead to the generation of the anaphylatoxin C5a, which crucially
triggers the inflammatory response of immune cells. For example, C5a induces the migration
of inflammatory cells, the degranulation of mast cells and the release of inflammatory cyto-
kines and regulates apoptosis [68]. The pro-inflammatory actions of C5a are mediated by its
receptor C5aR1, aclassical Gprotein-coupled receptor, expressed on the plasma membrane of
avariety of cells, particularly immune cells, including neutrophils, eosinophils and basophils
[6,9]. However, C5aR1 is also expressed on non-immune cells, including, among others, endo-
thelial cells and neurons, suggesting that complement-mediated signaling is not limited to
inflammatory cells [8]. We and others have demonstrated that C5aR1 is also strongly upregu-
lated during osteogenic differentiation of mesenchymal stem cells (MSCs) and induced osteo-
blast migration and the production of pro-inflammatory cytokines, including interleukin (IL)-
6and IL-8 in vitro [1,10,11]. Notably, C5a also enhanced the expression of receptor activator
of nuclear factor-κBligand (RANKL) in osteoblasts, akey stimulator of osteoclastogenesis,
indicating that C5a may indirectly induce osteoclastic bone resorption [12]. C5aR1 is also
expressed on osteoclasts and can directly regulate their differentiation and activity [1,13].
Taken together, these data indicate that the activation of the C5a/C5aR1 axis could modulate
bone cell activity and their immune-modulatory response [3]. The C5aR1 axis in bone cells
might be particularly important under inflammatory conditions after bone injury. Accord-
ingly, we demonstrated that C5aR1 was strongly upregulated in the periosteum, where precur-
sor cells already start to proliferate and undergo osteogenic differentiation during the early
phase after bone fracture [1]. These osteoblasts may modulate the local immune response after
stimulation with C5a, which is locally and, depending on trauma severity, systemically gener-
ated as aresponse to damage-associated molecular patterns. Furthermore, C5aR1 was strongly
expressed by bone-forming osteoblasts in the fracture callus over the entire healing period [1],
indicating that osteoblasts may serve as target cells for C5a not only in the early inflammatory
phase but also later during bone repair. Confirming this, C5-deficient mice displayed dis-
turbed bone formation in the fracture callus [2]. However, the specific role of the C5a-
C5aR1-interaction in osteoblasts during bone healing is currently poorly understood.
Therefore, we investigated the function of the C5a/C5aR1 axis in osteoblasts during bone
repair using transgenic mice with an osteoblast-specific C5aR1 overexpression. We evaluated
fracture healing in amodel of uncomplicated isolated fracture and in amodel of impaired
healing by combining the fracture with an additional thoracic trauma, which is associated with
local and systemic complement activation and severe inflammation [14,15]. We hypothesize
that if osteoblasts were important target cells for C5a, fracture healing would be disturbed.
Materials and methods
Mouse model
The mouse experiments were performed according to the international regulations for the
care and use of laboratory animals and approved by the Local Ethical Committee (Regierung-
spra¨sidium Tu¨bingen, GER, Reg.Nr. 1096). All mice were housed in groups of up to four mice
per cage with a14 hlight and 10 hdark cycle and received astandard mouse feed (ssniff
1
R/
M-H, V1535-300, Ssniff, Soest, Germany) and water ad libitum.Mice with an osteoblast-
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 2 / 17
specific overexpression of C5aR1 (Col1a1-C5aR1) were generated as follows: The open reading
frame encoding C5aR1 was placed under the control of the osteoblast-specific 2.3 kb Col1a1
promotor fragment. To confirm C5aR1 overexpression, Col1a1-C5aR1 mice were genotyped
using the forward primer 5-ACTACATCCTGGTCATCATCCTGC-3 and the reverse primer
5-CCAGCAGGAAACGGTCGGCA-3.Wildtype littermates (WT) were used as controls.
Cultivation of primary mouse osteoblasts
Primary osteoblasts were isolated from long bones of 8- to 12-week-old WT and Col1a1--
C5aR1 mice. Briefly, long bones were harvested and bone marrow was flushed out using phos-
phate-buffered saline (PBS, Biochrom, Berlin, Germany). The diaphyses were cut into small
pieces, which were digested with collagenase (1 mg/ml collagenase type II (125 U/mg, Sigma-
Aldrich, Taufkirchen, Germany)) for 2 h at 37˚C and then washed twice with PBS. For osteo-
blast expansion, bone fragments were cultivated in culture medium (α-MEM, Biochrom) sup-
plemented with 10% heat-inactivated fetal calf serum (Gibco, Darmstadt, Germany), 100 U/ml
penicillin/streptomycin (Gibco), 1% L-glutamine (PAN-Biotech, Aidenbach, Germany) and
0.5% Fungizone
TM
(Amphotericin B, Gibco) at 37˚C and 5% CO
2
atmosphere.
To induce osteogenic differentiation, osteoblasts were seeded in 24-well plates (CELL-
STAR1,Greiner Bio-One) at adensity of 0.5 ×10
4
cells/cm
2
in the presence of 0.2 mM ascor-
bate-2-phosphate (Sigma-Aldrich) and 10 mM β-glycerophosphate (Sigma-Aldrich) for 14 d.
Alkaline-phosphatase staining (Sigma-Aldrich) was performed to confirm osteogenic differen-
tiation. To investigate whether the differentiation capacity was influenced by C5a, 0.1 μg/ml of
recombinant mouse C5a (R&D Systems, Minneapolis, USA) was added to the differentiation
medium. To investigate whether C5a can induce alocal inflammatory response in osteoblasts
after osteogenic differentiation, cells were stimulated with 0.1 μg/ml C5a for 6or 24 hand ana-
lyzed by western blot and real-time quantitative PCR (RT-qPCR), respectively.
Osteoblast proliferation was assessed using the BrdU Cell Proliferation Kit (Cell Signaling
Technology, Danvers, MA, USA) according to manufacturer’s recommendations. Briefly, oste-
oblasts were seeded in 96-well plates at adensity of 1.6 ×10
4
cells/cm
2
.BrdU detection anti-
body (1:100) was added to culture medium for 24 hat 37˚C. Cells were then incubated with a
horseradish peroxidase (HRP)-conjugated secondary antibody for 1 h at RT, before adding
3,3,5,5-Tetramethylbenzidin substrate for 25 min at RT. The cells were analyzed by spectro-
photometry at 450 nm.
Reverse transcription and real-time quantitative PCR
Total RNA was isolated from cells using the RNeasy Mini Kit (Qiagen, Hilden, Germany)
according to the manufacturer’s instructions and cDNA was transcribed from 1μgRNA using
the Omniscript RT Kit (Qiagen). The StepOnePlus
TM
Real-Time PCR System (Applied Bio-
systems, Darmstadt, Germany) and the Platinum
1
SYBR
1
Green qPCR SuperMix-UDG
(Invitrogen ThermoFisher Scientific, Waltham, MA, USA) were used for cDNA amplification
and quantification according to the manufacturer’s instructions. Samples were initially incu-
bated at 50˚C and at 95˚C, for 2min each, followed by 40 cycles with the following cycling
conditions: 95˚C for 15 sand 60˚C for 1min. The amplification protocol was terminated
with amelting-curve step at 95˚C for 15 s, followed by 60˚C for 1min. Gene expression was
analyzed relative to the housekeeping gene glyceraldehyde-3-phosphate dehydrogenase
(GAPDH) using the ΔΔC
t
method with PCR-efficiency correction using LinRegPCR software
as described previously [16]. All primers were purchased from Invitrogen or Thermo Electron
(Ulm, Germany) (Table 1).
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 3 / 17
Western blot
Osteoblasts were lysed in Pierce1RIPA buffer (ThermoFisher Scientific, Waltham, MA,
USA), containing protease and phosphatase inhibitors (ThermoFisher Scientific). We resolved
10 μgof total protein on a10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis
and transferred it to anitrocellulose membrane (Amersham
TM
Protran
TM
0.2 mNC, GE
Healthcare, Chicago, IL, USA). Antibody blocking was performed in Tris-buffered saline with
Tween20 (TBS-T) with 3% bovine serum albumin for 1 h at room temperature (RT). Primary
rat anti-mouse C5aR (CD88) (MA1-81761, 1:1000, ThermoFisher Scientific), rabbit anti-
mouse p-Erk1/2 (4370P, 1:1000, Cell Signaling Technology), rabbit anti-mouse Erk1/2 (4695P,
1:1000, Cell Signaling Technology), rabbit anti-mouse p-p38 (4511P, 1:1000, Cell Signaling
Technology), rabbit anti-mouse p38 (8690P, 1:1000, Cell Signaling Technology) and arabbit
anti-mouse GAPDH antibody (2118, 1:2000, Cell Signaling) were incubated overnight at
4˚C, followed by incubation with asecondary HRP-coupled anti-rat or anti-rabbit antibody
(1:15,000, Cell Signaling Technology), respectively, at RT for 1h. WesternBright
TM
Quantum
or WesternBright
TM
ECL chemiluminescent HRP substrate (both Advansta, CA, USA) were
added to the membranes for 2min at RT and the luminescent signal was captured using the
Fusion Molecular Imaging System (Vilber Lourmat, Eberhardzell, Germany) or by membrane
exposure to X-ray film.
Animal studies
To investigate whether osteoblast-specific C5aR1 overexpression influences bone development
and bone mass, the skeleton of 12- and 55-week-old male WT and Col1a1-C5aR1 mice was
analyzed by micro-computed tomography (μCT) and histomorphometry as described below.
Mice received subcutaneous injections of alizarin red and calcein green (both 30 mg/kg) (both
Sigma, Steinheim, Germany) 3and 12 days before bone harvesting to determine dynamic
bone formation.
To study fracture healing, 12-week-old male Col1a1-C5aR1 mice and WT littermates
received astandardized femur osteotomy as described previously [17]. Briefly, mice were anes-
thetized with 2% isoflurane (Forene, Abbott, Wiesbaden, Germany) and an external fixator
(axial stiffness 3N/mm, RISystem Ind., Davos, Switzerland) was attached to the femur using
four Schanz-screws. An osteotomy gap was created between the two inner pins using awire
saw of 0.4 mm diameter (RISystem Ind.). Immediately after the osteotomy, half of the mice
received an additional thoracic trauma to induce abilateral, isolated lung contusion via a
Table 1. Primer sequences.
Target Gene Forward Primer Sequence (5’–3’) Reverse Primer Sequence (5’–3’)
AP (Alpl) GCT GAT CAT TCC CAC GTT TT GAG CCA GAC CAA AGA TGG AG
OCN (Bglap) GCG CTC TGT CTC TCT GAC CT ACC TTA TTG CCC TCC TGC TT
BSP (Ibsp) GAA GCA GGT GCA GAA GGA AC GAA ACC CGT TCA GAA GGA CA
GAPDH (Gapdh) ACC CAG AAG ACT GTG GAT GG GGA TGC AGG GAT GAT GTT CT
IL-6 (Il6)TCC TTC CTA CCC CAA TTT CC GCC ACT CCT TCT GTG ACT CC
RANKL (Tnfsf11) ATC ATG AAA CAT CGG GAA GC CTT GGG ATT TTG ATG CTG GT
OPG (Tnfrsf11b) CTG CCT GGG AAG AAG ATC AG GCT CGA TTT GCA GGT CTT TC
Protein name with gene name in bracket. AP: alkaline phosphatase, OCN: bone gamma carboxyglutamate
protein/osteocalcin, BSP: bone sialoprotein, GAPDH: glyceraldehyde-3-phosphate dehydrogenase, IL-6:
interleukin 6, RANKL: receptor activator of nuclear factor-κB ligand, OPG: osteoprotegerin.
https://doi.org/10.1371/journal.pone.0179512.t001
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 4 / 17
blast-wave generator centered 2cm above the middle of the thorax as described previously
[14,18]. For pain medication, all mice received 25 mg/l tramadol hydrochloride (Tramal1,
Gruenenthal GmbH, Aachen, Germany) in the drinking water 1 d before and up to 3 d after
surgery. Mice were sacrificed 3h, 1d, 3d, 14 d, 21 dand 25 dafter surgery using an overdose
of CO
2
and subsequent heart puncture with blood withdrawal. Blood for serum/plasma,
bronchoalveolar lavage (BAL) fluid and femur were harvested for further analyses.
Analysis of inflammatory mediators in blood, BAL fluids and callus
homogenates
Blood was harvested 3 h after surgery in microvettes (Sarstedt AG &Co., Nu¨mbrecht, Ger-
many) and centrifuged at 10,000 ×gfor 10 min for plasma collection or at 2800 ×gfor 10 min
for serum collection and stored at 80˚C. At the same time point, BAL fluid was collected by
flushing the lungs with 500 μlof ice-cold PBS. After centrifugation at 4000 ×gfor 10 min, the
supernatant was stored at 80˚C [19]. Fractured femurs were explanted 3 h after surgery,
shock-frozen in liquid nitrogen and stored at 80˚C. The fracture hematoma was collected in
150 μllysis buffer (10 mM Tris pH 7.5, 10 mM NaCl, 0.1 mM ethylenediaminetetraacetic acid,
0.5 mM Triton X-100, 0.02% NaN
3
,0.2 mM phenylmethylsulfonyl fluoride) with protease
inhibitors (1:100 vol/vol Sigma-Aldrich) and homogenized. After incubation for 30 min on
ice, the samples were centrifuged for 30 min at 10,000 ×gat 4˚C. The protein concentration in
the supernatant was determined using the Pierce
TM
BCA Protein Assay Kit (Fisher Scientific
GmbH, Schwerte, Germany) according to manufacturer’s protocol and lysates were stored at
80˚C. C5a concentrations were determined in plasma, callus lysates and BAL fluid and IL-6
was determined in serum using mouse enzyme-linked immunosorbent assay (ELISA) kits
(C5a: R&D Systems, 1:100, 1:50 and 1:100; respectively; IL-6: BD Biosciences, Singapore, 1:2).
IL-6 concentrations in callus lysates and BAL fluid were determined using amouse Multiplex-
ELISA kit (Bio-Plex Pro Cytokine Assay, Bio-Rad, Hercules, CA, USA). Data were analyzed
using the standard curve of cytokine standards and values below the detection limit of the
assay were set to zero.
Micro-computed tomography
Following fixation in 4% formalin, the femurs were scanned by micro-computed tomography
(μCT) using aSkyscan 1172, (Kontich, Belgium) at 50 kV, 200 μAand aresolution of 8μm
[14]. Analysis of μCT data was performed according to the guidelines of Bouxsein and col-
leagues [20]and the standard American Society for Bone and Mineral Research (ASBMR)
guidelines for μCT analysis [21]. For bone phenotyping, the volumes of interest (VOIs) were
defined as follows: 168 μmheight in the mid-diaphysis of the cortical bone and for the trabecu-
lar bone in the distal part of the femur 200 μmabove the growth plate and 280 μmheight. For
analysis of fracture healing, the fracture gap was defined as the VOI. Global thresholding
(641.9 mg hydroxyapatite/cm
3
)was used to distinguish between mineralized and non-miner-
alized tissue to calculate tissue mineral density and trabecular thickness, number and separa-
tion (CTAnalyser, Skyscan) [22]. Bone mineral density was determined without thresholding.
Histology and immunohistochemistry
Femurs were fixed in 4% buffered formaldehyde and embedded in paraffin for Safranin-O
staining (14 dafter surgery) or in polymethylmethacrylate for Giemsa staining (21 dand 25 d
after surgery) and bone phenotyping. Longitudinal sections of the fractured femur were cut
in the anterior-posterior direction. Bone, cartilage and fibrous tissue areas were assessed
using image-analyzing software (MMAF Version 1.4.0 MetaMorph1,Leica, Heerbrugg,
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 5 / 17
Switzerland) and OsteoMeasure1image-analysis software (OsteoMetrics, Inc., GA, USA) was
used to determine the number of osteoblasts in toluidine blue-stained sections, the number of
osteoclasts in tartrate-resistant acid phosphatase (TRAP)-stained sections and the bone forma-
tion rate according to the guidelines of the ASBMR [21].
Sections of the fractured femurs were immunostained 1and 3 d after surgery for neutro-
phils (Ly-6G
+
,#127632, BioLegend, San Diego, CA, USA), macrophages (F4/80
+
,Bio-Rad
AbD Serotec GmbH, Puchheim, Germany) and T-lymphocytes (CD8
+
,AM11118PU-S, Acris,
Herford, Germany), and 14 dafter surgery for Runx2 (cs8486, Cell Signaling), osteocalcin
(orb77248, Biorbyt Ltd, Cambridge, UK), RANKL (sc-7628, Santa Cruz, Dellas, Texas, USA)
and osteoprotegerin (OPG, APO6631PU-N, Acris). After deparaffinizing the 4% buffered
formaldehyde-fixed femur sections in xylene, they were rehydrated and blocked (F4/80
+
,
Runx2 and osteocalcin with 5% goat serum; Ly6-G
+
,CD8
+
and OPG with 10% goat serum) for
1 h at RT. Sections were incubated with primary antibodies (Ly6-G
+
1:300, F4/80
+
1:500,
CD8
+
1:500, Runx2 1:50, osteocalcin 1:200, RANKL 1:100, OPG 1:400) at 4˚C overnight except
for CD8
+
for 2 h at RT and then all for 1 h at RT with respective secondary antibodies (goat
anti-rat or goat anti-rabbit, biotinylated, 1:200, Life Technologies, Carlsbad, CA, USA). To
detect the antibodies, ABC kit and NovaRED substrate (both Vector laboratories Inc., Burlin-
game, CA, USA) were applied according to the manufacturer’s protocol, followed by counter-
staining with hematoxylin. For analysis under alight microscope (Leica DMI6000 B, Leica,
Heerbrugg, Switzerland), the region of interest was defined as the periosteal callus between the
inner pins of the fixator. Macrophage staining was analyzed in the marrow cavity in direct
proximity to the fracture gap.
Biomechanical testing
Osteotomized and intact femurs were explanted 21 dand 25 dafter surgery and the flexural
rigidity was analyzed by anon-destructive, three-point bending test using amaterial testing
machine (Z10, Zwick GmbH, Ulm, Germany) [17]. Briefly, an axial bending load with aforce
of up to 4 N was applied on top of the callus or on the mid-point of the diaphysis of the intact
femur. The flexural rigidity (EI) was calculated from the slope of the force deflection curve.
The EI of the fractured femur was related to the contralateral femur of the same animal [17].
Statistical analysis
Proliferation and osteogenic differentiation of primary osteoblasts was analyzed in 5–6 inde-
pendent experiments. Bone phenotype and fracture healing were determined in 5–8 mice per
genotype and time point.
Groups were tested for normal distribution using the Shapiro-Wilk test and then compared
by either Kruskall-Wallis and Dunn’s post-hoc test or by one-way analysis of variance and Fish-
ers LSD post-hoc test using GraphPad Prism 6(GraphPad Software Inc., La Jolla, CA, USA).
Data are presented as mean ±standard deviation. The level of significance was set at p<0.05.
Results
Osteoblast-specific overexpression of C5aR1 did not cause abone
phenotype
To investigate whether osteoblast-specific overexpression of C5aR1 influenced osteoblast
activity and bone remodeling, we performed in vitro and in vivo analyses. Western blot analysis
confirmed considerably increased C5aR1 expression in primary osteoblasts derived from
Col1a1-C5aR1 mice after inducing osteogenic differentiation (Fig 1A). Osteoblasts from WT
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 6 / 17
Fig 1. In vitro analysis of WT and Col1a1-C5aR1 osteoblasts. (A) Western blot showing C5aR1 expression in primary osteoblasts of wildtype (WT) and
Col1a1-C5aR1 mice after cultivating for 14 din normal proliferation medium (NM) and osteogenic differentiation medium (ODM). (B) Cell proliferationof WT
and Col1a1-C5aR1 osteoblastsdeterminedby BrdU incorporation. (C) Osteogenic differentiation capacity ofosteoblasts assessed by alkaline-phosphatase
staining after cultivating for 14 din ODM with or without C5a and (D) by the expression of the osteogenic marker genes Alpl (alkaline phosphatase), Ibsp
(bone sialoprotein) and Bglap (bone gamma carboxyglutamate protein/osteocalcin). (E) Western blot showing increased expression of phospho (p)-ERK 1/2
and phospho (p)-p38 after osteoblast stimulation with C5a for 6h. F) Relative gene expression of interleukin (IL)-6, receptor activator of NF-κBligand
(RANKL) and osteoprotegerin (OPG).
*
p<0.05;
***
p<0.001, n = 5–6 per group and treatment.
https://doi.org/10.1371/journal.pone.0179512.g001
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 7 / 17
and Col1a1-C5aR1 mice did not display any difference in proliferation (Fig 1B)or differentia-
tion capacity as demonstrated by alkaline phosphatase staining (Fig 1C)and the expression of
osteogenic differentiation markers (Fig 1D), also in the presence of C5a. Stimulation of WT and
Col1a1-C5aR1 osteoblasts with C5a increased phosphorylation of extracellular signal-regulated
kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinase (MAPK) (Fig 1E). In Col1a1--
C5aR1 osteoblasts, phosphorylation of p38 was increased compared to WT osteoblasts, indicat-
ing that the receptor was biologically active. This was confirmed by significantly increased IL-6
and RANKL expression after stimulation of Col1a1-C5aR1 cells with C5a (Fig 1F).
Cross inspection and X-ray examination of the skeleton did not reveal any difference
between 12-week-old and 55-week-old WT and Col1a1-C5aR1 mice (data not shown). μCT
and histomorphometric analyses of the femurs demonstrated that C5aR1 overexpression on
osteoblasts did not influence the bone mass, structural or cellular bone parameters or the bone
formation rate neither in the cortical nor trabecular compartments, indicating that bone devel-
opment and remodeling were not significantly affected (Table 2).
Osteoblast-specific overexpression of C5aR1 did not affect inflammation
in response to fracture
To investigate whether osteoblasts modulate the early inflammation via C5aR1 at the fracture
site, we analyzed the key inflammatory mediator IL-6 and the presence of immune cells locally
in the fracture hematoma. C5a was locally and systemically generated after fracture in WT and
Col1a1-C5aR1 mice without significant differences between the mouse strains (Table 3). In
the isolated fracture model, IL-6 concentration (Table 3)and the numbers of neutrophils
(Ly6G
+
cells), macrophages (F4/80
+
cells) and T-lymphocytes (CD8
+
cells) (Fig 2)were not
significantly altered in Col1a1-C5aR1 mice compared to WT mice.
Because C5a has been reported to trigger posttraumatic systemic inflammation after severe
injury [23], we combined the fracture with an additional thoracic trauma. The combined
trauma significantly enhanced serum IL-6 concentrations in WT and Col1a1-C5aR1 mice,
indicating systemic inflammation (Table 3). In the BAL fluid, C5a and IL-6 were significantly
enhanced in both genotypes (Table 3). The additional thoracic trauma did not influence the
concentration of inflammatory mediators in the fracture hematoma (Table 3)but, according
Table 2. Bone phenotype of WT and Col1a1-C5aR1 mice aged 12 and 55 weeks.
Parameter WT
12 weeks
Col1a1-C5aR1
12 weeks
WT
55 weeks
Col1a1-C5aR1
55 weeks
cortex TMD (g/mm
3
)1282 (±14) 1239 (±7) 1446 (±14) 1409 (±48)
C.Th (mm) 0.20 (±0.01) 0.24 (±0.02) 0.21 (±0.02) 0.21 (±0.03)
trabecular bone BMD (g/mm
3
)200 (±27) 257 (±49) 193 (±12) 179 (±46)
Tb.N (1/mm) 2.8 (±0.5) 3.4 (±0.8) 1.7 (±0.3) 1.8 (±0.3)
Tb.Th (mm) 0.06 (±0.00) 0.06 (±0.01) 0.06 (±0.01) 0.06 (±0.01)
Tb.Sp (mm) 0.19 (±0.02) 0.18 (±0.03) 0.24 (±0.02) 0.25 (±0.01)
Nb.Ob/B.Pm (1/mm) 18.9 (±3.2) 21.5 (±2.6) 15.6 (±6.2) 11.0 (±3.3)
#
Nb.Oc/B.Pm (1/mm) 3.7 (±1.7) 3.1 (±1.9) 2.3 (±0.6) 2.3 (±0.7)
BFR/BS (μm
3
/μm
2
/t) 0.5 (±0.04) 0.7 (±0.16) 0.2 (±0.22) 0.1 (±0.12)
#
WT: wildtype, TMD: tissue mineral density, C.Th: cortical thickness, BMD: bone mineral density, Tb.N: trabecular number, Tb.Th: trabecular thickness, Tb.
Sp: trabecular separation, Nb.Ob/B.Pm: number of osteoblasts per bone perimeter, Nb.Oc/B.Pm: number of osteoclasts per bone perimeter, BFR/BS: bone
formation rate per bone surface. Data are shown as mean ±standard deviation. No data were found to be significantly different (p <0.05) between strains.
#
p<0.05 vs. 12-week-old Col1a1-C5aR1 mice, n = 5–8 per group.
https://doi.org/10.1371/journal.pone.0179512.t002
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 8 / 17
Fig 2. Numberof inflammatory cells in the fracture callus of WT and Col1a1-C5aR mice.(A) Quantification of neutrophils (Ly-6G
+
)at 1d, (B)
representative immunohistological images of neutrophils 1 d after surgery and (C) quantification of neutrophils (Ly-6G
+
) 3 d after surgery. (D) Quantification
of macrophages (F4/80
+
) 3 d and E) T-lymphocytes (CD8
+
) 1 d after surgery of wildtype (WT) and Col1a1-C5aR1 mice. Fx: mice with isolated fracture, Fx
+TXT: mice with combined fracture and thoracic trauma (TXT). Arrows indicate some of the positively Ly-6G
+
stained cells, C: cortex, scale bar 100 μm.
*
p<0.05, n = 6 per group.
https://doi.org/10.1371/journal.pone.0179512.g002
Table 3. C5a and IL-6 concentrations in blood, BAL fluid and fracture hematoma 3 h after surgery.
untreated mice Fracture fracture + TXT
WT Col1a1-C5aR1 WT Col1a1-C5aR1
blood C5a (ng/ml) 0.8 (±0.6) 5.7 (±1.0) 5.3 (±1.5) 6.5 (±1.1) 6.3 (±1.6)
IL-6 (pg/ml) 8.9 (±3.0) 42.8 (±20.3) 57.1 (±28.6) 154.2 (±59.0)*188.2 (±38.4)*
BAL fluid C5a (ng/ml) 8.6 (±1.2) 9.0 (±3.0) 8.6 (±1.4) 13.9 (±2.1)*13.6 (±2.0)*
IL-6 (pg/ml) 3.5 (±0.6) 3.2 (±3.9) 3.1 (±2.8) 123.7 (±55.6)*218.7 (±161.0)*
fracture hematoma C5a (ng/mg protein) - 1.4 (±0.6) 1.2 (±0.6) 1.6 (±0.7) 2.0 (±1.0)
IL-6 (pg/mg protein) - 81.9 (±49.2) 80.4 (±51.8) 53.2 (±27.0) 35.3 (±26.4)
BAL: bronchoalveolar lavage, WT, wildtype, TXT: thorax trauma. Data are shown as mean ±standard deviation. C5a was analyzed in plasma and IL-6 was
analyzed in serum.
*
p<0.05 vs. group without TXT, n = 5–6 per group.
https://doi.org/10.1371/journal.pone.0179512.t003
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 9 / 17
to our previous studies [14,15,19,24], increased the recruitment of neutrophils to the fracture
site in WT mice (Fig 2). However, the osteoblast-specific overexpression of C5aR1 did not
affect inflammatory parameters in the combined trauma model (Table 3,Fig 2).
Osteoblast-specific overexpression of C5aR1 significantly impaired
bone formation in the fracture callus
To investigate callus formation, we analyzed the tissue composition in the repair phase on day
14, when callus tissue is abundantly formed by intramembranous and endochondral ossifica-
tion. In the isolated fracture model, we did not find significant differences in the content of
cartilage and bone between the genotypes (Fig 3A–3D). Notably, the number of osteoblasts
was significantly decreased whereas the number of osteoclasts increased in the callus of mice
with an osteoblast-specific C5aR1 overexpression (Fig 3E–3G). The additional thoracic trauma
Fig 3. Fracturehealing in WT and Col1a1-C5aR1 mice 14 dafter surgery. (A) Representative histological images of wildtype (WT) and Col1a1-C5aR1
mice with isolated fracture and fracture with additional thoracic trauma (TXT). (B) Callus area, (C) amount of osseous tissue and (D) amount of cartilage. (E)
Number of osteoblasts per bone perimeter(N.Ob/B.Pm)and (F) number of osteoclasts per bone perimeter (N.Oc/B.Pm) of WT and Col1a1-C5aR1 mice. (G)
Representative tartrate-resistant acid phosphatase (TRAP) staining of fractured calli of WT and Col1a1-C5aR1 mice with isolated fracture. Fx: mice with
isolated fracture, Fx+TXT: mice with combined fracture and thoracic trauma. C: cortex, scale bar 100 μm,
*
p<0.05, n=6per group.
https://doi.org/10.1371/journal.pone.0179512.g003
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 10 /17
slightly reduced the amount of bone and decreased the number of osteoblasts in the callus
without significant differences between both genotypes (Fig 3C and 3E). Immunostaining
revealed intense staining for Runx2 and osteocalcin of osteoblasts on newly formed bone tra-
beculae in all groups without significant differences between genotypes, indicating undis-
turbed osteogenic differentiation (Fig 4). We did not detect significant differences in the
expression of RANKL, but found less OPG-positive osteoblasts in mice with Col1a1-C5aR1
overexpression.
In WT mice with isolated fracture, most of the cartilage was transformed to bone after 21
days. However, in Col1a1-C5aR1 mice, bone content, BMD and the flexural rigidity were sig-
nificantly reduced (Fig 5). We found more osteoclasts in the callus of Col1a1-C5aR1 mice,
whereas osteoblast numbers were not different compared to WT mice (Fig 5G–5I). In Col1a1--
C5aR1 mice, the additional thoracic trauma induced significantly more pronounced effects
compared to WT mice as demonstrated by significantly reduced bone content and flexural
rigidity (Fig 5C and 5F).
Fig 4. Representative immunohistological images of bone markersin the fracture callus of WT and Col1a1-C5aR1mice 14 dafter surgery. (A)
Runx2,(B) osteocalcin, (C) receptor activator of nuclear factor-κBligand (RANKL) and (D) osteoprotegerin (OPG). WT: wildtype, Fx: mice with isolated
fracture,Fx+TXT: mice with combined fracture and thoracic trauma (TXT). Arrows indicate some of the positively stained cells, C: cortex, scale bar 100 μm.
https://doi.org/10.1371/journal.pone.0179512.g004
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 11 /17
After 25 days, fractured bones were completely healed in both genotypes with isolated
fracture (Fig 6). However, at this time point, the strong effect of the thoracic trauma in
Fig 5. Fracturehealing in WT and Col1a1-C5aR1 mice 21 dafter surgery. (A) Representative histological images of wildtype (WT) and Col1a1-C5aR1
mice with isolated fracture and fracture with additional thoracic trauma (TXT). (B) Callus area, (C) amount of osseous tissue and (D) amount of cartilage. (E)
Bone mineral density (BMD). (F) Relative flexural rigidity. (G) Number of osteoblasts per bone perimeter(N.Ob/B.Pm) and (H) number of osteoclastsper
bone perimeter (N.Oc/B.Pm) of WT and Col1a1-C5aRmice. (I) Representative tartrate-resistant acid phosphatase (TRAP) staining of fracturedcalli of WT
and Col1a1-C5aR1 mice with isolated fracture.Fx: mice with isolated fracture, Fx+TXT: mice with combined fracture and thoracic trauma. C: cortex, scale
bar 100 μm,
*
p<0.05;
**
p<0.005;
***
p<0.001, n = 6–7 per group.
https://doi.org/10.1371/journal.pone.0179512.g005
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 12 /17
Col1a1-C5aR1 mice was still clear as demonstrated by significantly reduced flexural rigidity of
the fractured femurs, increased callus area and atendential increase in the cartilaginous tissue
area (Fig 6).
Discussion
The objective of the present study was to unravel the role of the C5aR1 in osteoblasts regarding
inflammation and bone formation post fracture using mice with an osteoblast-specific overex-
pression of C5aR1. Transgenic Col1a1-C5aR1 mice displayed normal bone development and
turnover. However, fracture healing both in amodel of uncomplicated fracture and in amodel
after severe additional trauma was considerably impaired in transgenic mice, indicating that
C5aR1 signaling in osteoblasts may be crucially involved in bone formation under pathological
conditions.
We first characterized the bone phenotype of Col1a1-C5aR1 mice to investigate whether
C5aR1 overexpression affects bone development and turnover. The morphology of the skele-
ton and the density and structural parameters of cortical and trabecular bone were unaffected
in both young and elderly Col1a1-C5aR1 mice. Col1a1-C5aR1 osteoblasts derived from these
mice exhibited undisturbed proliferation and differentiation capacities, including after stimu-
lation of the receptor with its ligand C5a. These results demonstrated that osteoblast-specific
C5aR1 overexpression did not alter bone development or turnover. However, this did not
exclude an important role for the C5aR1 under physiological conditions. It was demonstrated
that osteoblasts produce the key complement protein C5 also in the absence of inflammatory
Fig 6. Fracture healing in WT and Col1a1-C5aR1 mice 25 dafter surgery. (A) Callus area, (B) amount of osseous
tissue, (C) amount of cartilageand (D) relative flexural rigidity of wildtype (WT) and Col1a1-C5aR1 mice. Fx: mice with
isolated fracture, Fx+TXT: mice with combined fracture and thoracic trauma (TXT).
*
p<0.05, n = 6 per group.
https://doi.org/10.1371/journal.pone.0179512.g006
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 13 /17
stimuli and that osteoclasts can effectively cleave C5 to biologically active C5a [12]. Confirm-
ing this, Andrades et al. found C5 together with other complement components expressed in
the bone growth plate and suggested that complement might contribute to the transformation
of cartilage to bone during endochondral ossification [25]. Indeed, previous examination of
the bone phenotype of C5-deficient mice demonstrated that the absence of C5 had an impact
on cortical bone [2]. Twelve-week-old C5-deficient mice exhibited decreased bending stiffness
and increased growth plates, confirming that C5a signaling could play aregulatory role in
bone development, however, further studies are necessary to dissect the molecular mecha-
nisms [2].
Confirming previous data that C5aR1 signaling induces apro-inflammatory and osteoclas-
togenic response in osteoblasts [10,12], our results revealed that IL-6 and RANKL expression
were significantly increased in Col1a1-C5aR1 osteoblasts in response to C5a. In agreement
with findings in immune cells [2628]and MSCs [11], we showed that the C5aR1 downstream
signaling in osteoblasts involved ERK1/2 and p38 phosphorylation, which is known to regulate
the expression of inflammatory cytokines, including IL-6 [27,29]. These data suggest that the
C5aR1 axis in osteoblasts may be particularly important under inflammatory conditions. In
accordance with this, it was shown that C5aR1-knockout mice were protected against ost-
eoarthritis [30]and that bone loss in bacteria-induced periodontitis was associated with an
increased C5aR1 activity in osteoblasts [31]. We previously demonstrated that osteoblasts
strongly upregulated C5aR1 during the early phase after bone fracture [1]. Therefore, we
hypothesized, that C5a, which is known to be abundantly generated locally and systemically in
response to tissue injury [3234], may induce an immune-modulatory response in osteoblasts
during fracture healing. Our results revealed that the concentration of IL-6, akey cytokine in
the local immune response after bone fracture and in posttraumatic systemic inflammation
[3538], was neither locally nor systemically altered in Col1a1-C5aR1 mice. Additionally, the
recruitment of innate and adaptive immune cells to the fracture site was not significantly influ-
enced, suggesting that C5aR1 signaling in osteoblasts did not modulate the early immune
response after fracture. Apossible reason may be the low number of osteoblasts compared to
the high number of active immune cells during this early stage. However, bone healing was
considerably disturbed in the later healing stages in Col1a1-C5aR1 mice. The bone content
and mineral density and the mechanical properties of the fracture callus were significantly
reduced. The reduced bone fraction may be primarily caused by asignificantly increased num-
ber of osteoclasts observed after 14 and 21 days in the fracture callus of Col1a1-C5aR1 mice.
These observations are in agreement with the present and previous in vitro data, demonstrat-
ing that C5a significantly enhanced the RANKL/OPG ratio in osteoblasts, thereby promoting
osteoclastogenesis [12]. Immunohistochemistry did not reveal obvious differences in RANKL
expression, but OPG expression was clearly reduced in the callus of Col1a1-C5aR1 mice.
These results are confirmed by other authors demonstrating that C5aR1 is important for osteo-
clastogenesis [1,10,12]. Bone marrow cells from C5aR1-deficient mice and abrogating C5aR1
signaling in WT cells with aC5aR1 antibody resulted in decreased osteoclastogenesis, whereas
stimulation of WT or C3-deficent bone marrow cells with C5a enhanced osteoclastogenesis
[13]. Moreover, IL-6 is probably involved in the differentiation of osteoclasts regulated by
complement, because IL-6 expression was decreased in bone marrow cells from C3aR/C5aR1-
deficent mice [13]. In Col1a1-C5aR1 mice, osteoblast numbers in the fracture callus were
reduced on day 14 but were unchanged on day 21 and the expression of the osteoblastic differ-
entiation markers Runx2 and osteocalcin was unaltered, suggesting that osteoblast activity
might not be considerably affected, which was confirmed by our in vitro data. However, this
needs further investigation. Moreover, to the best of our knowledge, no literature data exist
about the role of the second C5a receptor, C5aR2, in bone metabolism and fracture healing.
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 14 /17
Concluding, the data suggest that C5aR1 signaling in osteoblasts may crucially modulate oste-
oclast formation and activity and thereby increase bone resorption during fracture healing by
inducing the expression of osteoclastic factors, including RANKL and IL-6.
We also investigated healing in acombined model of fracture and thoracic trauma, which
induces aposttraumatic systemic inflammatory response, resulting in impaired fracture heal-
ing [14,15,19,24]. Previously, we demonstrated that blockade of the C5aR1 by aspecific antag-
onist during the early inflammatory phase abolished the negative effects of the severe trauma
on fracture healing, indicating that C5a abundantly generated during posttraumatic inflamma-
tion may crucially contribute to trauma-induced disturbed fracture healing [39]. The com-
bined fracture and thoracic trauma induced asystemic posttraumatic inflammatory response
and impaired bone healing in both WT and Col1a1-C5aR1 mice, as demonstrated by increased
IL-6 serum levels and by significantly reduced bone content and mechanical properties of the
fracture callus, respectively, in both mouse strains. The impairment of fracture healing by the
additional thoracic trauma was more pronounced in mice with the osteoblast-specific overex-
pression of C5aR1, confirming the significance of the C5a/C5aR1 axis in osteoblasts during
fracture healing. After 25 days, when the fractures were completely healed in WT mice,
Col1a1-C5aR1 mice still exhibited asignificantly reduced bending stiffness, suggesting adetri-
mental impact of C5aR1 signaling in trauma-induced impaired bone regeneration.
In conclusion, the present data together with our previous experiments [1,12,15,24,39,40]
suggest acrucial role of the C5aR1 in fracture healing and implies that C5aR1 inhibition
may be asuitable strategy to prevent fracture-healing complications particularly after severe
trauma.
Acknowledgments
The authors acknowledge the excellent technical assistance of Sonja Braunmu¨ller, Bettina
Stahl, Sevil Essig, Marion Tomo, Ursula Maile, Stefanie Riesemann and Helga Bach.
Author Contributions
Conceptualization: MHL AI.
Data curation: AI.
Formal analysis: SB AK YH.
Funding acquisition: TS MA MHL AI.
Investigation: SB AK YH AL TS.
Methodology: AL TS MA MHL AI.
Project administration: SB AK AI.
Resources: TS MA MHL AI.
Supervision: AK MHL AI.
Validation: SB AK YH AL AI.
Visualization: SB AK YH.
Writing original draft: SB AK YH AI.
Writing review &editing: SB AK YH AL TS MA MHL AI.
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 15 /17
References
1. Ignatius A, EhrnthallerC, Brenner RE, Kreja L, Schoengraf P, Lisson P, et al. The anaphylatoxin recep-
tor C5aR is present during fracture healing in rats and mediates osteoblast migration in vitro. JTrauma.
2011 Oct; 71(4):952–60. PMID: 21460748.https://doi.org/10.1097/TA.0b013e3181f8aa2d
2. EhrnthallerC, Huber-Lang M, Nilsson P, Bindl R, Redeker S, Recknagel S, et al. Complement C3 and
C5 deficiency affects fracture healing. PLoS One. 2013 Nov 18; 8(11):e81341. https://doi.org/10.1371/
journal.pone.0081341 PMID: 24260573.
3. Scho
¨ngraf P, Lambris JD, Recknagel S, Kreja L, Liedert A, Brenner RE, et al. Does complement play a
role in bone development and regeneration? Immunobiology. 2013 Jan; 218(1):1–9.PMID: 22464814.
https://doi.org/10.1016/j.imbio.2012.01.020
4. Ricklin D, Hajishengallis G, Yang K, Lambris JD. Complement: a key system for immune surveillance and
homeostasis. Nat Immunol. 2010 Sep; 11(9):785–97.PMID: 20720586.https://doi.org/10.1038/ni.1923
5. Amara U, RittirschD, Flierl M, Bruckner U, Klos A, Gebhard F, et al. Interactionbetween the coagulation
and complement system. Adv Exp Med Biol. 2008; 632: 71–9. PMID: 19025115.
6. Klos A, Tenner AJ, Johswich KO, Ager RR, Reis ES, Ko
¨hl J. The role of the anaphylatoxins in health
and disease. Mol Immunol. 2009; Sep; 46(14):2753–66. PMID: 19477527.https://doi.org/10.1016/j.
molimm.2009.04.027
7. Manthey HD, Woodruff TM, Taylor SM, Monk PN. Complement component 5a (C5a). Int JBiochem
Cell Biol. 2009 Nov; 41(11):2114–7. PMID: 19464229.https://doi.org/10.1016/j.biocel.2009.04.005
8. Monk PN, Scola AM, Madala P, Fairlie DP. Function, structure and therapeutic potential of complement
C5a receptors. Br JPharmacol. 2007 Oct; 152(4):429–48. PMID: 17603557.https://doi.org/10.1038/sj.
bjp.0707332
9. Gerard NP, Hodges MK, Drazen JM, Weller PF, Gerard C. Characterization of areceptor for C5a ana-
phylatoxin on human eosinophils. JBiol Chem. 1989 Jan 25; 264(3):1760–6. PMID: 2912983.
10. Pobanz JM, Reinhardt RA, Koka S, Sanderson SD. C5a modulation of interleukin-1 beta-induced inter-
leukin-6 production by human osteoblast-like cells. JPeriodontalRes. 2000 Jun; 35(3):137–45. PMID:
10929868.
11. Schraufstatter IU, Discipio RG, Zhao M, Khaldoyanidi SK. C3a and C5a are chemotactic factors for
human MSCs, which cause prolonged ERK1/2 phosphorylation. JImmunol. 2009 Mar 15; 182
(6):3827–36. PMID: 19265162.https://doi.org/10.4049/jimmunol.0803055
12. Ignatius A, Schoengraf P, Kreja L, Liedert A, Recknagel S, Kandert S, et al. Complement C3a and C5a
modulate osteoclast formation and inflammatoryresponse of osteoblastsin synergism with IL-1β. J Cell
Biochem. 2011 Sep; 112(9):2594–605. PMID: 21598302.https://doi.org/10.1002/jcb.23186
13. Tu Z, Bu H, Dennis JE, Lin F. Efficient osteoclast differentiation requires local complement activation.
Blood. 2010 Nov 25; 116(22):4456–63. PMID: 20709903 https://doi.org/10.1182/blood-2010-01-263590
14. Kemmler J, Bindl R, McCook O, Wagner F, Gro
¨ger M, Wagner K, et al. Exposure to 100% Oxygen abol-
ishes the impairment of fracture healing after thoracic trauma. PLoS One. 2015Jul 6; 10(7):e0131194.
https://doi.org/10.1371/journal.pone.0131194 PMID: 26147725.
15. Recknagel S, Bindl R, Brochhausen C, Go
¨ckelmann M, Wehner T, Schoengraf P, et al. Systemic
inflammationinduced by athoracic trauma alters the cellular composition of the early fracture callus. J
Trauma Acute Care Surg. 2013 Feb; 74(2):531–7. PMID: 23354247.https://doi.org/10.1097/TA.
0b013e318278956d
16. Ramakers C, Ruijter JM, Deprez RH, Moorman AF. Assumption-free analysis of quantitativereal-time
polymerase chain reaction (PCR) data. Neurosci Lett. 2003 Mar 13; 339(1):62–6. PMID: 12618301.
17. Ro
¨ntgen V, Blakytny R, Matthys R, Landauer M, Wehner T, Go
¨ckelmann M, et al. Fracture healing in
mice under controlled rigid and flexible conditionsusing an adjustable external fixator. JOrthop Res.
2010 Nov; 28(11):1456–62. PMID: 20872581.https://doi.org/10.1002/jor.21148
18. Kno
¨ferl MW, Liener UC, Seitz DH, Perl M, Bru¨ckner UB, Kinzl L, et al. Cardiopulmonary, histological,
and inflammatoryalterations after lung contusion in anovel mouse model of blunt chest trauma. Shock.
2003 Jun; 19(6):519–25. PMID: 12785006.https://doi.org/10.1097/01.shk.0000070739.34700.f6
19. Kovtun A, Bergdolt S, Wiegner R, Radermacher P, Huber-Lang M, Ignatius A. The crucial role of neutro-
phil granulocytes in bone fracture healing. Eur Cell Mater. 2016 Jul 25; 32:152–62. PMID: 27452963.
20. Bouxsein ML, Boyd SK, Christiansen BA, Guldberg RE, Jepsen KJ, Mu¨ller R. Guidelines for assess-
ment of bone microstructure in rodents using micro-computed tomography. JBone Miner Res. 2010
Jul; 25(7):1468–86. PMID: 20533309.https://doi.org/10.1002/jbmr.141
21. Parfitt AM, Drezner MK, Glorieux FH, Kanis JA, Malluche H, Meunier PJ, et al. Bone histomorphometry:
Standardization of nomenclature, symbols and units. JBone Miner Res. 1987 Dec; 2(6):595–610.
PMID: 3455637.https://doi.org/10.1002/jbmr.5650020617
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 16 /17
22. Morgan EF, Mason ZD, Chien KB, Pfeiffer AJ, Barnes GL, Einhorn TA, et al. Micro-computed tomogra-
phy assessment of fracture healing: Relationships among callus structure, composition, and mechani-
cal function. Bone. 2009 Feb; 44(2):335–44. PMID: 19013264.https://doi.org/10.1016/j.bone.2008.10.
039
23. Huber-Lang M, Gebhard F, Schmidt CQ, Palmer A, Denk S, Wiegner R. Complement therapeutic strat-
egies in trauma, hemorrhagic shock and systemic inflammation—closingPandora’s box? Semin Immu-
nol. 2016 Jun; 28(3):278–84. PMID: 27157701.https://doi.org/10.1016/j.smim.2016.04.005
24. Recknagel S, Bindl R, Kurz J, Wehner T, EhrnthallerC, Knoferl MW, et al. Experimental blunt chest
trauma impairs fracture healing in rats. JOrthop Res. 2011 May; 29(5):734–9. PMID: 21437953.https://
doi.org/10.1002/jor.21299
25. Andrades JA1, Nimni ME, Becerra J, Eisenstein R, Davis M, Sorgente N. Complement proteins are
present in developing endochondral bone and may mediate cartilagecell death and vascularization.
Exp Cell Res. 1996 Sep 15; 227(2):208–13. PMID: 8831558 https://doi.org/10.1006/excr.1996.0269
26. Chiou WF, Tsai HR, Yang LM, Tsai WJ. C5a differentially stimulates the ERK1/2 and p38 MAPK phos-
phorylation throughindependent signaling pathwaysto induced chemotactic migration in RAW264.7
macrophages. Int Immunopharmacol. 2004 Oct; 4(10–11):1329–41. PMID: 15313431.https://doi.org/
10.1016/j.intimp.2004.05.017
27. Riedemann NC, Guo RF, Hollmann TJ, Gao H, Neff TA, Reuben JS, et al. Regulatoryrole of C5a in
LPS-induced IL-6 production by neutrophils during sepsis.FASEB J. 2004 Feb; 18(2):370–2. PMID:
14688199.https://doi.org/10.1096/fj.03-0708fje
28. RousseauS, Dolado I, Beardmore V, Shpiro N, MarquezR, Nebreda AR, et al. CXCL12 and C5a trigger
cell migrationvia aPAK1/2-p38alpha MAPK-MAPKAP-K2-HSP27 pathway.Cell Signal. 2006 Nov; 18
(11):1897–905. PMID: 16574378.https://doi.org/10.1016/j.cellsig.2006.02.006
29. Tuyt LM, Dokter WH, BirkenkampK, Koopmans SB, Lummen C, Kruijer W, et al. Extracellular-regulated
kinase 1/2, Jun N-terminal kinase, and c-Jun are involved in NF-kappa B-dependent IL-6 expression in
human monocytes. J Immunol. 1999 Apr 15; 162(8):4893–902. PMID: 10202034.
30. Banda NK, Hyatt S, Antonioli AH, White JT, Glogowska M, Takahashi K, et al. Role of C3a receptors,
C5a receptors, and complement protein C6 deficiency in collagen antibody-induced arthritis in mice. J
Immunol. 2012 Feb 1; 188(3):1469–78. PMID: 22205026.https://doi.org/10.4049/jimmunol.1102310
31. Liang S, Krauss JK, Domon H, McIntosh ML, Hosur KB, Qu H, et al. The C5a receptor impairs IL-12–
dependent clearance of Porphyromonas gingivalis and is required for induction of periodontal bone
loss. JImmunol. 2011 Jan 15; 186(2):869–77. PMID: 21149611.https://doi.org/10.4049/jimmunol.
1003252
32. Dong Q, Sun L, Peng L, Yan B, Lv J, Wang G, et al. Expressionof C5a and its receptor following spinal
cord ischemia reperfusion injury in the rat. Spinal Cord. 2015 Aug; 53(8):581–4. PMID: 25917954.
https://doi.org/10.1038/sc.2015.65
33. Kanse SM, Gallenmueller A, Zeerleder S, Stephan F, Rannou O, Denk S, et al. Factor VII-activating
protease is activated in multiple trauma patients and generates anaphylatoxin C5a. JImmunol. 2012
Mar 15; 188(6):2858–65. PMID: 22308306.https://doi.org/10.4049/jimmunol.1103029
34. LeinhaseI, Holers VM, Thurman JM, Harhausen D, Schmidt OI, Pietzcker M, et al. Reduced neuronal
cell death after experimental brain injury in mice lacking afunctional alternative pathway of complement
activation. BMC Neurosci. 2006 Jul 14; 7:55. PMID: 16842619.https://doi.org/10.1186/1471-2202-7-55
35. Gebhard F, Pfetsch H, Steinbach G, Strecker W, Kinzl L, Bruckner UB. Is interleukin 6an early marker
of injury severity following major trauma in humans? Archives of surgery. 2000 Mar; 135(3):291–5.
PMID: 10722030.
36. Kolar P, Schmidt-Bleek K, Schell H, Gaber T, Toben D, SchmidmaierG, et al. The early fracture hema-
toma and its potentialrole in fracture healing. Tissue Eng Part BRev. 2010 Aug; 16(4):427–34. PMID:
20196645.https://doi.org/10.1089/ten.TEB.2009.0687
37. Marsell R, Einhorn TA. The biology of fracture healing. Injury. 2011 Jun; 42(6):551–5. PMID: 21489527.
https://doi.org/10.1016/j.injury.2011.03.031
38. Yang X, Ricciardi BF, Hernandez-Soria A, Shi Y, Pleshko Camacho N, Bostrom MP. Callus mineraliza-
tion and maturationare delayed during fracture healing in interleukin-6 knockout mice. Bone. 2007 Dec;
41(6):928–36. PMID: 17921078.https://doi.org/10.1016/j.bone.2007.07.022
39. Recknagel S, Bindl R, Kurz J, Wehner T, Schoengraf P, EhrnthallerC, et al. C5aR-antagonist signifi-
cantly reduces the deleterious effect of ablunt chest trauma on fracture healing. JOrthop Res. 2012
Apr; 30(4):581–6. PMID: 21922535.https://doi.org/10.1002/jor.21561
40. EhrnthallerC, Ignatius A, Gebhard F, Huber-Lang M. New insights of an old defense system: structure,
function, and clinical relevance of the complement system. Mol Med. 2011 Mar-Apr; 17(3–4):317–29.
PMID: 21046060.https://doi.org/10.2119/molmed.2010.00149
Osteoblast-specific overexpression of C5aR1 impairs fracture healing
PLOS ONE |https://doi.org/10.1371/journal.pone.0179512 June 14, 2017 17 /17
... While global C3 − /− and C5 − /− mice displayed no differences in bone volume fraction at age 12 weeks (Ehrnthaller et al., 2013), C5aR1 null mice have increased bone mass accrual . Thus, prior osteoimmunology studies have focused on C5aR in fracture healing of the adult skeleton Ehrnthaller et al., 2013;Recknagel et al., 2012;Ehrnthaller et al., 2016;Bergdolt et al., 2017). However, the role of C3a/C3aR signaling in the growing skeleton is currently unknown. ...
... Complement involvement in bone has included homeostasis Ehrnthaller et al., 2011;Andrades et al., 1996), regeneration (Ehrnthaller et al., 2016), inflammation Banda et al., 2012;Murayama et al., 2015), alveolar bone (Hajishengallis et al., 2011;Liang et al., 2011), and more. Though the anaphylatoxin receptor C5aR has been primarily described in bone fracture and repair Kovtun et al., 2017;Ehrnthaller et al., 2016;Bergdolt et al., 2017), C3a/C3aR signaling and expression have not been as well-characterized in health and pathological skeletal conditions. ...
Article
Full-text available
Osteoimmune studies have identified complement signaling as an important regulator of the skeleton. Specifically, complement anaphylatoxin receptors (i.e., C3aR, C5aR) are expressed on osteoblasts and osteoclasts, implying that C3a and/or C5a may be candidate mediators of skeletal homeostasis. The study aimed to determine how complement signaling influences bone modeling/remodeling in the young skeleton. Female C57BL/6J C3aR-/-C5aR-/- vs. wildtype and C3aR-/- vs. wildtype mice were examined at age 10 weeks. Trabecular and cortical bone parameters were analyzed by micro-CT. In situ osteoblast and osteoclast outcomes were determined by histomorphometry. Osteoblast and osteoclast precursors were assessed in vitro. C3aR-/-C5aR-/- mice displayed an increased trabecular bone phenotype at age 10 weeks. In vitro studies revealed C3aR-/-C5aR-/- vs. wildtype cultures had less bone-resorbing osteoclasts and increased bone-forming osteoblasts, which were validated in vivo. To determine whether C3aR alone was critical for the enhanced skeletal outcomes, wildtype vs. C3aR-/- mice were evaluated for osseous tissue outcomes. Paralleling skeletal findings in C3aR-/-C5aR-/- mice, C3aR-/- vs. wildtype mice had an enhanced trabecular bone volume fraction, which was attributed to increased trabecular number. There was elevated osteoblast activity and suppressed osteoclastic cells in C3aR-/- vs. wildtype mice. Furthermore, primary osteoblasts derived from wildtype mice were stimulated with exogenous C3a, which more profoundly upregulated C3ar1 and the pro-osteoclastic chemokine Cxcl1. This study introduces the C3a/C3aR signaling axis as a novel regulator of the young skeleton.
... 21 The authors showed that both, C5aR1 overexpression in osteoblasts as well as a global knockout of C5aR1 resulted in impaired fracture healing in mice. 47,48 Thus, a balanced C5aR1 expression seems important during bone regeneration. During fracture healing, bone develops partly from a cartilage template, but this cartilage-to-bone transition also takes place at the growth plate. ...
Article
Full-text available
Objective: The complement cascade as major fluid phase innate immune system is activated during progression of osteoarthritis (OA). Generated anaphylatoxins and the corresponding receptors C3aR and C5aR1 are associated with the calcification of blood vessels and involved in osteogenic differentiation. This study aims on elucidating whether complement activation products contribute to cartilage calcification of OA cartilage. Method: Human articular chondrocytes were osteogenically differentiated in vitro in the presence or absence of C3a, C5a, and bone morphogenetic protein (BMP) 2. Furthermore, macroscopically intact (OARSI grade ≤ 1) and highly degenerated human cartilage (OARSI grade ≥ 3) was used for C3aR and C5aR1 histochemistry. Calcification of the cartilage was assessed by Alizarin Red S and von Kossa staining. Results: C3a and C5a amplified matrix mineralization during in vitro osteogenesis, while inhibition of the corresponding receptors impaired calcium deposition. Moreover, C3aR and C5aR1 expression was upregulated during osteogenic differentiation and also in degenerated cartilage. Additionally, anaphylatoxin receptor expression was positively associated with calcification of native cartilage tissue and calcium deposition during osteogenic differentiation. Finally, the pro-hypertrophic growth factor BMP2 induced the expression of C5aR1. Conclusions: Our findings indicate that anaphylatoxins and their receptors play a decisive role in cartilage calcification processes during OA progression.
... Activation of the complement system leads to the release of pro-inflammatory peptide fragments C3a and C5a from C3 and C5 molecules, respectively, which are involved in immune responses in human cells [35]. A recent study demonstrated that disruption of C5AR1 reduces tumor-associated osteoclastogenesis and impairs osseous colonization in various cancer [36]. Additionally, C5aR1 activation in LysM-expressing cells is crucial for their recruitment and activation [31]. ...
Article
Full-text available
Background Accumulating evidence suggests that polo-like kinase 3 (PLK3) plays an essential role in tumor cells and induces cell proliferation and may have implications for the prognosis of various cancers. We sought to define the role of PLK3-dependent proneural–mesenchymal transition (PMT) in the glioblastoma (GBM) therapy.Methods and resultsWe analyzed the expression data for PLK3 by using the TCGA database. PLK3 expression in GBM cell lines was determined by qRT-PCR and Western blotting. PLK3 levels were modulated using Lentivirus infection, and the effects on symptoms, tumor volume, and survival in mice intracranial xenograft models were determined. Irradiation (IR) was performed to induce PMT. PLK3 expression was significantly elevated in mesenchymal subtype GBM and promoted tumor proliferation in GBM. Additionally enriched PLK3 expression could be associated with poor prognosis in GBM patients compared with those who have lower PLK3 expression. Mechanically, PLK3-dependent PMT induced radioresistance in GBM cells via transcriptional regulation of complement C5a receptor 1 (C5AR1). In therapeutic experiments conducted in vitro, targeting PLK3 by using small molecule inhibitor decreased tumor growth and radioresistance of GBM cells both in vitro and in vivo.ConclusionsPLK3-C5AR1 axis induced PMT thus enhanced radioresistance in GBM and could become a novel potential therapeutic target for GBM.
... Total RNA isolation was performed using the FFPE RNEasy kit from Qiagen and RT-PCR was performed as described previously (46). Quantitative RT-PCR was performed using the SensiFAST SYBR Hi-ROX One-Step Kit (Bioline, Memphis, TN, USA). ...
Article
Full-text available
Introduction Clinical and preclinical data suggest accelerated bone fracture healing in subjects with an additional traumatic brain injury (TBI). Mechanistically, altered metabolism and neuro-endocrine regulations have been shown to influence bone formation after combined fracture and TBI, thereby increasing the bone content in the fracture callus. However, the early inflammatory response towards fracture and TBI has not been investigated in detail so far. This is of great importance, since the early inflammatory phase of fracture healing is known to be essential for the initiation of downstream regenerative processes for adequate fracture repair. Methods Therefore, we analyzed systemic and local inflammatory mediators and immune cells in mice which were exposed to fracture only or fracture + TBI 6h and 24h after injury. Results We found a dysregulated systemic immune response and significantly fewer neutrophils and mast cells locally in the fracture hematoma. Further, local CXCL10 expression was significantly decreased in the animals with combined trauma, which correlated significantly with the reduced mast cell numbers. Discussion Since mast cells and mast cell-derived CXCL10 have been shown to increase osteoclastogenesis, the reduced mast cell numbers might contribute to higher bone content in the fracture callus of fracture + TBI mice due to decreased callus remodeling.
... Interestingly, Ignatius et al. showed that osteoblasts and osteoclasts were able to activate complement components C3 and C5. These activated peptides, especially C5a, triggered osteoclast activity (called osteoclastogenesis) in a pro-inflammatory environment, even in the absence of the main osteoclast activators, receptor activator of nuclear factor-kappa B ligand (RANKL) and macrophage-colony stimulating factor (M-CSF) [81,83,84] (Fig. 1A). Intra-articular ligaments and tendons are known to be predisposed to OA mostly due to aberrant mechanical stress [85]. ...
Article
Osteoarthritis (OA) affects more than 500 million people worldwide and is among the five diseases in Germany causing the highest suffering of the patients and cost for the society. The quality of life of OA patients is severely compromised, and adequate therapy is lacking owing to a knowledge gap that acts as a major barrier to finding safe and effective solutions. Chronic, low-grade inflammation plays a central role in OA pathogenesis and is associated with both OA pain and disease progression. Innate immune pathways, such as the complement- and pattern-recognition receptor pathways, are pivotal to the inflammation in OA and key components of the innate immune system implicated in OA include DAMP-TLR signaling, the complement system, carboxypeptidase B (CPB), and mononuclear cells. Anaphylatoxins C3a and C5a are small polypeptides (77 and 74 amino acids, respectively) which are released by proteolytic cleavage of the complement components C3 and C5. The alternative complement pathway seems to play a crucial role in OA pathogenesis as these complement components, mostly C3 and its activation peptide C3a, were detected at high levels in osteoarthritic cartilage, synovial membrane, and cultured chondrocytes. Targeting the complement system by using anti-complement drugs as a therapeutic option bears the risk of major side effects such as increasing the risk of infection, interfering with cell regeneration and metabolism, and suppressing the clearance of immune complexes. Despite those adverse effects, several synthetic complement peptide antagonists show promising effects in ameliorating inflammatory cell responses also in joint tissues.
Article
Myocardial infarction (MI) and osteoporotic fracture (Fx) are two of the leading causes of mortality and morbidity worldwide. Although these traumatic injuries are treated as if they are independent, there is epidemiological evidence linking the incidence of Fx and MI, thus raising the question whether each of these events can actively influence the risk of the other. Atherosclerotic cardiovascular disease and osteoporosis, the chronic conditions leading to MI and Fx, are known to have shared pathoetiology. Furthermore, sustained systemic inflammation after traumas such as MI and Fx has been shown to exacerbate both of these underlying chronic conditions. However, the systemic effects of MI and Fx outside their own system have not been well studied. The sympathetic nervous system (SNS) and the complement system have been shown to initiate a systemic response after MI that could lead to subsequent changes in bone remodeling through osteoclasts. Similarly, SNS and complement system activation following fracture could lead to heart tissue damage and exacerbate atherosclerosis. To determine whether damaging bone-heart crosstalk may be an important co-morbidity following Fx or MI, this review details the current understanding of bone loss after MI, cardiovascular damage after Fx, and possible shared underlying mechanisms of these processes.
Article
Osteoimmunology is at full display during endosseous implant osseointegration. Bone formation, maintenance and resorption at the implant surface is a result of bidirectional and dynamic reciprocal communication between the bone and immune cells that extends beyond the well-defined osteoblast-osteoclast signaling. Implant surface topography informs adherent progenitor and immune cell function and their cross-talk to modulate the process of bone accrual. Integrating titanium surface engineering with the principles of immunology is utilized to harness the power of immune system to improve osseointegration in healthy and diseased microenvironments. This review summarizes current information regarding immune cell–titanium implant surface interactions and places these events in the context of surface-mediated immunomodulation and bone regeneration. A mechanistic approach is directed in demonstrating the central role of osteoimmunology in the process of osseointegration and exploring how regulation of immune cell function at the implant-bone interface may be used in future control of clinical therapies. The process of peri-implant bone loss is also informed by immunomodulation at the implant surface. How surface topography is exploited to prevent osteoclastogenesis is considered herein with respect to peri-implant inflammation, osteoclastic precursor-surface interactions, and the upstream/downstream effects of surface topography on immune and progenitor cell function.
Article
Full-text available
In recent years, evidence has accumulated that the complement system, an integral part of innate immunity, may be involved in the regulation of bone homeostasis as well as inflammatory bone loss, for example, in rheumatoid arthritis and periodontitis. Complement may also contribute to osteoporosis development, but investigation of the mechanism is limited. Using mice with a conditional deletion of the complement anaphylatoxin receptor C5aR1, we here demonstrated that C5aR1 in osteoblasts (C5aR1 Runx2-Cre mice) or osteoclasts (C5aR1 LysM-Cre mice) did not affect physiological bone turnover or age-related bone loss in either sex, as confirmed by micro-computed tomography, histomorphometry, and biomechanical analyses of the bone and by the measurement of bone turnover markers in the blood serum. When female mice were subjected to ovariectomy (OVX), a common model for postmenopausal osteoporosis, significant bone loss was induced in C5aR1 fl/fl and C5aR1 LysM-Cre mice, as demonstrated by a significantly reduced bone volume fraction, trabecular number and thickness as well as an increased trabecular separation in the trabecular bone compartment. Confirming this, the osteoclast number and the receptor activator of nuclear factor k-B (RANK) ligand (RANKL) serum level were significantly elevated in these mouse lines. By contrast, C5aR1 Runx2-Cre mice were protected from bone loss after OVX and the serum RANKL concentration was not increased after OVX. These data suggested that bone cell-specific C5aR1 may be redundant in bone homeostasis regulation under physiological conditions. However, C5aR1 on osteoblasts was crucial for the induction of bone resorption under osteoporotic conditions by stimulating RANKL release, whereas C5aR1 on osteoclasts did not regulate OVX-induced bone loss. Therefore, our results implicate C5aR1 on osteoblasts as a potential target for treating postmenopausal osteoporosis.
Article
Full-text available
Bone remodeling is a physiological, dynamic process that mainly depends on the functions of 2 cell types: osteoblasts and osteoclasts. Emerging evidence suggests that complement system is crucially involved in the regulation of functions of these cells, especially during inflammatory states. In this context, complement component 5a (C5a), a powerful pro-inflammatory anaphylatoxin that binds the receptor C5aR1, is known to regulate osteoclast formation and osteoblast inflammatory responses, and has thus been proposed as potential therapeutic target for the treatment of inflammatory bone diseases. In this review, we will analyze the role of C5a-C5aR1 axis in bone physiology and pathophysiology, describing its involvement in the pathogenesis of some of the most frequent inflammatory bone diseases such as rheumatoid arthritis, and also in osteoporosis and bone cancer and metastasis. Moreover, we will examine C5aR1-based pharmacological approaches that are available and have been tested so far for the treatment of these conditions. Given the growing interest of the scientific community on osteoimmunology, and the scarcity of data regarding the role of C5a-C5aR1 axis in bone pathophysiology, we will highlight the importance of this axis in mediating the interactions between skeletal and immune systems and its potential use as a therapeutic target.
Article
Full-text available
Delayed bone fracture healing and the formation of non- unions represent an important clinical problem, particularly in polytrauma patients who suffer from posttraumatic systemic inflammation. However, the underlying pathomechanisms remain unclear. Neutrophil granulocytes are crucial effector cells in the systemic immune response and represent the most abundant immune cell population in the early fracture haematoma. Here we investigated the role of neutrophils in a mouse model of uncomplicated fracture healing and compromised fracture healing induced by an additional thoracic trauma. Twenty four hours before injury, 50 % of the mice were systemically treated with an anti-Ly-6G-antibody to reduce neutrophil numbers. In the isolated fracture model, Ly-6G-Ab treatment significantly increased the concentration of both pro- and anti-inflammatory cytokines, including interleukin (IL)-6 and IL-10, and chemokines, for example, C-X-C motif ligand 1 (CXCL1) and monocyte chemotactic protein-1 (MCP-1), in the fracture haematoma. Monocyte/ macrophage recruitment was also significantly enhanced. After 21 d, bone regeneration was considerably impaired as demonstrated by significantly diminished bone content and impaired mechanical properties of the fracture callus. These results indicate that undisturbed neutrophil recruitment and function in the inflammatory phase after fracture is crucial to initiate downstream responses leading to bone regeneration. In the combined trauma model, the reduction of neutrophil numbers ameliorated pulmonary inflammation but did not provoke any significant effect on bone regeneration, suggesting that neutrophils may not play a crucial pathomechanistic role in compromised fracture healing induced by an additional thoracic trauma.
Article
Full-text available
In polytrauma patients a thoracic trauma is one of the most critical injuries and an important trigger of post-traumatic inflammation. About 50% of patients with thoracic trauma are additionally affected by bone fractures. The risk for fracture malunion is considerably increased in such patients, the pathomechanisms being poorly understood. Thoracic trauma causes regional alveolar hypoxia and, subsequently, hypoxemia, which in turn triggers local and systemic inflammation. Therefore, we aimed to unravel the role of oxygen in impaired bone regeneration after thoracic trauma. We hypothesized that short-term breathing of 100% oxygen in the early post-traumatic phase ameliorates inflammation and improves bone regeneration. Mice underwent a femur osteotomy alone or combined with blunt chest trauma 100% oxygen was administered immediately after trauma for two separate 3 hour intervals. Arterial blood gas tensions, microcirculatory perfusion and oxygenation were assessed at 3, 9 and 24 hours after injury. Inflammatory cytokines and markers of oxidative/nitrosative stress were measured in plasma, lung and fracture hematoma. Bone healing was assessed on day 7, 14 and 21. Thoracic trauma induced pulmonary and systemic inflammation and impaired bone healing. Short-term exposure to 100% oxygen in the acute post-traumatic phase significantly attenuated systemic and local inflammatory responses and improved fracture healing without provoking toxic side effects, suggesting that hyperoxia could induce anti-inflammatory and pro-regenerative effects after severe injury. These results suggest that breathing of 100% oxygen in the acute post-traumatic phase might reduce the risk of poorly healing fractures in severely injured patients.
Article
Full-text available
There is increasing evidence that complement may play a role in bone development. Our previous studies demonstrated that the key complement receptor C5aR was strongly expressed in the fracture callus not only by immune cells but also by bone cells and chondroblasts, indicating a function in bone repair. To further elucidate the role of complement in bone healing, this study investigated fracture healing in mice in the absence of the key complement molecules C3 and C5. C3(-/-) and C5(-/-) as well as the corresponding wildtype mice received a standardized femur osteotomy, which was stabilized using an external fixator. Fracture healing was investigated after 7 and 21 days using histological, micro-computed tomography and biomechanical measurements. In the early phase of fracture healing, reduced callus area (C3(-/-): -25%, p=0.02; C5(-/-): -20% p=0.052) and newly formed bone (C3(-/-): -38%, p=0.01; C5(-/-): -52%, p=0.009) was found in both C3- and C5-deficient mice. After 21 days, healing was successful in the absence of C3, whereas in C5-deficient mice fracture repair was significantly reduced, which was confirmed by a reduced bending stiffness (-45%; p=0.029) and a smaller callus volume (-17%; p=0.039). We further demonstrated that C5a was activated in C3(-/-) mice, suggesting cleavage via extrinsic pathways. Our results suggest that the activation of the terminal complement cascade in particular may be crucial for successful fracture healing.
Article
Full-text available
Severe tissue injury results in early activation of serine protease systems including the coagulation and complement cascade. In this context, little is known about factor VII-activating protease (FSAP), which is activated by substances released from damaged cells such as histones and nucleosomes. Therefore, we have measured FSAP activation in trauma patients and have identified novel FSAP substrates in human plasma. Mass spectrometry-based methods were used to identify FSAP binding proteins in plasma. Anaphylatoxin generation was measured by ELISA, Western blotting, protein sequencing, and chemotaxis assays. Plasma samples from trauma patients were analyzed for FSAP Ag and activity, nucleosomes, C5a, and C3a. Among others, we found complement components C3 and C5 in FSAP coimmunoprecipitates. C3 and C5 were cleaved by FSAP in a dose- and time-dependent manner generating functional C3a and C5a anaphylatoxins. Activation of endogenous FSAP in plasma led to increased C5a generation, but this was not the case in plasma of a homozygous carrier of Marburg I single nucleotide polymorphism with lower FSAP activity. In multiple trauma patients there was a large increase in circulating FSAP activity and nucleosomes immediately after the injury. A high correlation between FSAP activity and C5a was found. These data suggest that activation of FSAP by tissue injury triggers anaphylatoxin generation and thereby modulates the posttraumatic inflammatory response in vivo. A strong link between C5a, nucleosomes, and FSAP activity indicates that this new principle might be important in the regulation of inflammation.
Article
After severe trauma, the immune system is challenged with a multitude of endogenous and exogenous danger molecules. The recognition of released danger patterns is one of the prime tasks of the innate immune system. In the last two decades, numerous studies have established the complement cascade as a major effector system that detects and processes such danger signals. Animal models with engineered deficiencies in certain complement proteins have demonstrated that widespread complement activation after severe injury culminates in complement dysregulation and excessive generation of complement activation fragments. Such exuberant pro-inflammatory signaling evokes systemic inflammation, causes increased susceptibility to infections and is associated with a detrimental course of the disease after injury. We discuss the underlying processes of such complementopathy and recapitulate different intervention strategies within the complement cascade. So far, several orthogonal anti-complement approaches have been tested with varying success in a large number of rodent, in several porcine and few simian studies. We illustrate the different features among those intervention strategies and highlight those that hold the greatest promise to become potential therapeutic options for the intricate disease of traumatic injury.
Article
Experimental study. To investigate the expression of C5a and its receptor after spinal cord ischemia reperfusion injury (SIRI) in rat. Department of Neurosurgery, the Second Affiliated Hospital, Xi'an Jiaotong University, Shaanxi Province, Xi'an, ChinaMethods:Sprague Dawley rats were subjected to 1 h of infrarenal aorta occlusion to induce spinal cord ischemia. Spinal cord was reperfused for 12, 24, 48 h or 3 days separately after ischemia, respectively. Enzyme-linked immunosorbent assay was used to detect C5a in rat serum. Immunofluorescent staining was performed to detect the expression and cellular localization of C5aR in spinal cord following SIRI. Following SIRI, the motor behavior of the rats was significantly compromised. The serum concentration of C5a in the rat was elevated after SIRI and peaked at 24 h. C5aR was significantly upregulated in the lumbar spinal cord following SIRI and expressed on motor neurons and the microglia, but not astrocyte. There was no significant difference in the expression level of C5aR localized on motor neurons after SIRI. Remarkable upregulation of the C5aR may be associated with increased number of C5aR-positive microglia and elevated cellular expression level. This study provides the first evidence of the expression of C5a and its receptors in SIRI and suggests their possible contribution to SIRI in a rat model.Spinal Cord advance online publication, 28 April 2015; doi:10.1038/sc.2015.65.
Article
Periodontal bone resorption is controlled by osteoblast products, including interleukin (IL)-6, which are stimulated by other cytokines and complement components in the pro-inflammatory milieu. This study demonstrated that human osteoblast-like osteosarcoma cells (MG-63) responded to human recombinant (hr) C5a by releasing significant amounts of the bone-resorbing cytokine IL-6. C5a-induced release of IL-6 was enhanced 330% when cells were exposed to IL-1β prior to C5a challenge at optimal concentrations (1.0 μg/ml C5a, 0.1 ng/ml IL-1β). Cells simultaneously challenged with these concentrations of C5a and IL-1β produced a 700% increase in IL-6 release relative to cells challenged with IL-1β alone. Incubation of IL-1β-treated cells with anti-human C5a receptor (C5aR) Ab resulted in a 78% suppression of the C5a-induced release of IL-6, but C5aR neutralization did not affect C5a/IL-1β co-stimulation of IL-6. In addition, neither IL-1β nor C5a significantly altered the other's cell-surface receptor relative to binding affinity or density. These results indicate that while MG-63 cells express functional C5aRs, the synergistic effect of C5a and IL-1β on osteoblast IL-6 production is probably controlled by post-receptor signaling events. C5a agonists and antagonist used to alter critical C5a concentrations may present a new point of therapeutic intervention for the treatment of inflammatory bone resorption such as is found in periodontitis.
Article
We recently demonstrated that a blunt chest trauma, a strong inducer of the posttraumatic systemic inflammatory response and one of the most critical injuries in polytrauma patients, significantly delayed fracture healing in rats, possibly by the interaction of the systemic inflammation with early regeneration processes locally at the fracture site. The underlying cellular mechanisms, however, have as yet remained unknown. Therefore, the aim of this study was to analyze the cellular and morphologic composition of the early fracture callus after a blunt chest trauma. Rats received an osteotomy of the right femur stabilized by an external fixator in combination with a blunt chest trauma or not. The animals were killed after 3, 7, and 35 days, and the fracture calli were analyzed histologically for new tissue formation, polymorphonuclear leucocytes, macrophages, osteoclasts, and the presence of the proinflammatory cytokine interleukin 6. The blunt chest trauma considerably increased the number of polymorphonuclear leucocytes in the callus by Day 3 compared with animals with isolated fractures. The number of macrophages was significantly reduced by the thoracic trauma at Days 3 and 7. The number of osteoclasts was not changed at any postoperative time point. After 3 days, the blunt chest trauma led to a significantly stronger interleukin 6 staining within the periosteal callus in zones of intramembranous ossification. During the time of cortical bridging at Day 35, the amount of newly formed bone was significantly decreased after blunt chest trauma. Our results suggest that the systemic posttraumatic inflammation induced by a thoracic trauma disturbed the inflammatory balance during the early healing stage by altering the recruitment of inflammatory cells and cytokine expression locally at the fracture site and thus impaired fracture healing. These findings provide new insights in the pathomechanisms of impaired fracture healing in patients experiencing severe trauma.
Article
There is a tight interaction of the bone and the immune system. However, little is known about the relevance of the complement system, an important part of innate immunity and a crucial trigger for inflammation. The aim of this study was, therefore, to investigate the presence and function of complement in bone cells including osteoblasts, mesenchymal stem cells (MSC), and osteoclasts. qRT-PCR and immunostaining revealed that the central complement receptors C3aR and C5aR, complement C3 and C5, and membrane-bound regulatory proteins CD46, CD55, and CD59 were expressed in human MSC, osteoblasts, and osteoclasts. Furthermore, osteoblasts and particularly osteoclasts were able to activate complement by cleaving C5 to its active form C5a as measured by ELISA. Both C3a and C5a alone were unable to trigger the release of inflammatory cytokines interleukin (IL)-6 and IL-8 from osteoblasts. However, co-stimulation with the pro-inflammatory cytokine IL-1β significantly induced IL-6 and IL-8 expression as well as the expression of receptor activator of nuclear factor-kappaB ligand (RANKL) and osteoprotegerin (OPG) indicating that complement may modulate the inflammatory response of osteoblastic cells in a pro-inflammatory environment as well as osteoblast–osteoclast interaction. While C3a and C5a did not affect osteogenic differentiation, osteoclastogenesis was significantly induced even in the absence of RANKL and macrophage-colony stimulating factor (M-CSF) suggesting that complement could directly regulate osteoclast formation. It can therefore be proposed that complement may enhance the inflammatory response of osteoblasts and increase osteoclast formation, particularly in a pro-inflammatory environment, for example, during bone healing or in inflammatory bone disorders. J. Cell. Biochem. 112: 2594–2605, 2011. © 2011 Wiley-Liss, Inc.