SlideShare une entreprise Scribd logo
1  sur  12
Télécharger pour lire hors ligne
Acta Biomaterialia 10 (2014) 4730–4741 
Contents lists available at ScienceDirect 
Acta Biomaterialia 
journal homepage: www.elsevier.com/locate/actabiomat 
Mesenchymal stroma cells trigger early attraction of M1 macrophages 
and endothelial cells into fibrin hydrogels, stimulating long bone healing 
without long-term engraftment 
Elisabeth Seebach, Holger Freischmidt, Jeannine Holschbach 1, Jörg Fellenberg, Wiltrud Richter ⇑ 
Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany 
a r t i c l e i n f o 
Article history: 
Received 22 May 2014 
Received in revised form 10 July 2014 
Accepted 14 July 2014 
Available online 22 July 2014 
Keywords: 
Bone healing 
Mesenchymal stroma cells 
Fibrin hydrogel 
Host cell recruitment 
Macrophage 
a b s t r a c t 
Implantation of mesenchymal stroma cells (MSCs) is an attractive approach to stimulate closure of large 
bone defects but an optimal carrier has yet to be defined. MSCs may display trophic and/or immunomod-ulatory 
features or stimulate bone healing by their osteogenic activity. The aim of this study was to unra-vel 
whether fibrin hydrogel supports early actions of implanted MSCs, such as host cell recruitment, 
immunomodulation and tissue regeneration, in long bone defects. Female rats received cell-free fibrin 
or male MSCs embedded in a fibrin carrier into plate-stabilized femoral bone defects. Removed callus 
was analyzed for host cell invasion (day 6), local cytokine expression (days 3 and 6) and persistence of 
male MSCs (days 3, 6, 14 and 28). Fibrin–MSC composites triggered fast attraction of host cells into 
the hydrogel while cell-free fibrin implants were not invaded. A migration front dominated by M1 
macrophages and endothelial progenitor cells formed while M2 macrophages remained sparse. Only 
MSC-seeded fibrin hydrogel stimulated early tissue maturation and primitive vessel formation at day 6 
in line with significantly higher VEGF mRNA levels recorded at day 3. Local TNF-a, IL-1b and IL-10 expres-sion 
indicated a balanced immune cell activity independent of MSC implantation. Implanted MSCs 
persisted until day 14 but not day 28. Our results demonstrate that fibrin hydrogel is an attractive carrier 
for MSC implantation into long bone defects, supporting host cell attraction and pro-angiogenic activity. 
By this angiogenesis, implant integration and tissue maturation was stimulated in long bone healing 
independent of long-term engraftment of implanted MSCs. 
 2014 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved. 
1. Introduction 
The closure of large bone defects secondary to trauma or tumor 
resection remains a challenge in orthopedic surgery. One promis-ing 
approach to support natural bone healing is the implantation 
of bone marrow-derived mesenchymal stroma cells (MSCs) in a 
suitable carrier [1]. So far, sponge-like scaffolds have mainly been 
used for cell implantation although these have disadvantages, such 
as limited cell seeding, unequal cell distribution and non-homoge-neous 
matrix deposition. Hydrogels are attractive vehicles for cell 
transplantation because of their easy adaptation to any shape of 
defect, homogeneous cell distribution, even extracellular matrix 
deposition and beneficial interaction with the host tissue [2–4]. 
High biocompatibility, biodegradation and the adhesiveness of 
fibrin glue [5] have rendered fibrin hydrogel a common carrier 
for autologous cell transplantation in cartilage repair [6,7]. Surpris-ingly, 
fibrin has so far attracted little attention as a carrier for MSC-based 
treatment of bone defects and little is known as to whether 
fibrin hydrogels support early actions of MSCs in stimulating long 
bone healing. MSCs can easily be isolated from bone marrow [8], 
are characterized by fast proliferation in culture, and express 
CD90, CD105 and vascular cell adhesion molecule 1 (VCAM1, 
CD106) on their surface in the absence of CD34 and CD45 [9,10]. 
After expansion culture, MSCs can be induced to differentiate into 
chondrogenic, adipogenic and osteogenic lineages in vitro [11]. 
Moreover, MSCs provide a supportive cellular microenvironment 
by secreting a variety of molecules including growth factors and 
cytokines which potentially mediate host cell recruitment, angio-genesis 
and immune modulation [12,13]. MSCs further possess a 
significant osteogenic potential in vivo and have successfully been 
used to induce repair of bone defects in animals [14,15] and in 
humans [16,17]. 
⇑ Corresponding author. Tel.: +49 (0) 6221 5629254; fax: +49 (0) 6221 5629288. 
E-mail address: wiltrud.richter@med.uni-heidelberg.de (W. Richter). 
1 Current address: Merck KGaA, Frankfurter Strasse 250, 64293 Darmstadt, 
Germany. 
http://dx.doi.org/10.1016/j.actbio.2014.07.017 
1742-7061/ 2014 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 4731 
According to the concept of stem cell-based bone regeneration, 
implanted MSCs contribute physically to the repair tissue by gen-erating 
new bone tissue, as has been reported in ectopic implanta-tion 
models in immune-deficient mice in which fibrin hydrogel 
formed part of the carrier [18,19]. The role and fate of MSCs 
implanted into a bone defect is, however, unclear and long-term 
persistence, especially in immune competent animals, has been 
discussed [20–22]. Fast attraction of a large number of diverse host 
cells to the defect site, acceleration of new vessel formation and 
modulation of the early inflammatory reaction may be important 
trophic roles of MSCs in stimulation of bone healing which should 
be supported by the carrier used for implantation. In an orthotopic 
setting, these mechanisms are potentially more important for 
improved bone regeneration than enhancing the number of local 
bone-forming cells by implantation of additional osteogenic pro-genitors. 
Whether fibrin hydrogel is an attractive carrier for MSC 
implantation in this respect remains to be determined. 
A highly potent MSC population has to be produced in a stan-dardized 
fashion from each donor to allow optimal healing results 
in the patient. In view of the changing paradigm of MSC function 
[23], the therapeutically most potent MSC subpopulations may, 
however, be characterized by pronounced trophic and/or immuno-modulatory 
features displayed in the early callus instead of a high 
osteogenic differentiation capacity. Thus, it is essential to better 
understand relevant actions of MSC composites in a bone defect 
in order to optimize MSC-based bone tissue engineering 
approaches and to design an optimal application strategy for clin-ical 
regeneration of large bone defects. 
The aim of this study was to assess whether fibrin hydrogel is 
an attractive carrier for MSC implantation supporting early bone 
regeneration with regard to host cell attraction, angiogenesis, 
immunomodulation and physical contribution of implanted MSCs 
to newly formed bone tissue. In order to follow up implanted MSCs 
without extra manipulation by a labelling reaction, our approach 
was to implant sex-mismatched MSCs in a fibrin hydrogel into a 
long bone defect in rats. Callus was removed from female recipi-ents 
at distinct time points after surgery and analyzed for host cell 
recruitment, the presence of inflammatory cytokines and the per-sistence 
of male MSCs to pinpoint the dominant actions of 
fibrin–MSC composites in early bone regeneration. 
2. Materials and methods 
2.1. Animals and study design 
Sprague–Dawley rats (n = 46; 17–23 weeks of age, Charles 
River, Sulzfeld, Germany) were included in the animal study. The 
hind limbs in the growing rat are skeletally mature at around 
15 weeks of age [24]. The experimental protocol was approved 
by the local animal experimental ethics committee and all proce-dures 
were performed according to the European Laboratory Ani-mal 
Science Guidelines. Male rats were used for MSC donation 
and female rats served as MSC recipients. A total of 43 animals 
received a non-critical 2 mm bone defect in the right femur. Two 
experimental groups were defined. Rats of group 1 received an 
empty fibrin clot and served as controls (n = 20), while rats of 
group 2 were treated with 1  106 MSCs in a fibrin clot (n = 23). 
Animals were killed 3, 6, 14 or 28 days after surgery. Histological 
evaluation of callus was performed at day 6 (five animals per 
group). Gene expression analysis and detection of the sex-deter-mining 
region of Y-Gen (SRY)-specific DNA were assessed in the 
same sample at days 3 and 6 from the callus of five animals per 
condition. Visualization of new bone formation by micro-com-puted 
tomography (lCT) and SRY-DNA detection was done on 
the same animals with n = 3 animals for the cell-free and n = 4 
animals for the MSC-treated group at day 14, and with n = 2 
animals for the cell-free and n = 4 animals for the MSC-treated 
group at day 28. 
2.2. MSC isolation and expansion 
MSCs were isolated from bone marrow of three male rats using a 
modified centrifugation method [25]. Animals were killed by 
asphyxiation with CO2 and MSCs were extracted from tibiae and 
femora by centrifugation at 750g for 2 min. All cells were pooled, 
washed with phosphate-buffered saline (PBS) and seeded into 
0.1% gelatin-coated flasks in a high-glucose Dulbecco’s modified 
Eagle’s medium (DMEM) containing 12.5% fetal calf serum (FCS), 
2mM L-glutamine, 1% non-essential amino acids, 0.1% 2-mercap-toethanol 
(Invitrogen, Karlsruhe, Germany), 100 U ml–1 penicillin, 
100 lgml–1 streptomycin (Biochrom, Berlin, Germany) and 
4 ngml–1 recombinant human fibroblast growth factor-2 (Active 
Bioscience, Hamburg, Germany) at a density of 1.25  105 
cells cm–2. After 5 days under standard culturing conditions (37 C 
and 6% CO2) non-adherent cells were removed by washing. Medium 
was changed every 2 days and cells were split at 80% confluency and 
replated at a density of 4–6  103 cells cm–2. All cells were frozen 
in passage 1 in liquid nitrogen and recultivated at a density of 
6  103 cells cm–2 3 days before implantation. 
2.3. Flow cytometry 
At the time of implantation cells were harvested with trypsin 
and 1  105 cells were labelled with anti-CD45 (1:250), anti- 
CD68 (1:100), anti-CD90-FITC (1:25; Abcam, Cambridge, UK) and 
anti-CD106-PE (1:50; BD, Heidelberg, Germany) antibodies, 
respectively. For CD45 and CD68 staining cells were incubated 
with a secondary anti-mouse FITC-conjugated antibody (1:100; 
Jackson ImmunoResearch Laboratories, Suffolk, UK). Surface mar-ker 
positive cells were quantified in a FACSCalibur (BD, Heidelberg, 
Germany). 
2.4. Osteogenic, adipogenic and chondrogenic in vitro differentiation 
For osteogenesis, quadruplicates of 3.5  104 MSCs were seeded 
per well of a 24-well plate in osteogenic medium consisting 
of high-glucose DMEM supplemented with 10% FCS, 0.1 lM 
dexamethasone, 0.17 mM ascorbic acid 2-phosphate, 10 mM 
b-glycero-phosphate (Sigma-Aldrich, Taufkirchen, Germany), 
100 U ml–1 penicillin and 100 lgml–1 streptomycin. At days 1, 7 
and 14 the alkaline phosphatase (ALP) enzyme activity of cell 
lysate was measured as described previously [18]. For adipogenic 
differentiation cells were seeded in duplicates at 3.5  104 cells 
per well of a 24-well plate in high-glucose DMEM supplemented 
with 10% FCS, 0.01 lM dexamethasone, 5 lgml–1 insulin (Sanofi- 
Aventis, Frankfurt, Germany), 100 U ml–1 penicillin and 
100 lgml–1 streptomycin [26]. Formation of lipid droplets was 
evaluated by Oil Red O staining (Chroma, Münster, Germany) after 
fixing the cells in 4% paraformaldehyde (PFA, Merck, Darmstadt, 
Germany). Chondrogenesis was performed with 5  105 cells in a 
3D pellet-culture in high-glucose DMEM supplemented with 
5 lgml–1 transferrin, 5 ng ml–1 sodium selenite, 0.1 lMdexameth-asone, 
0.17 mM ascorbic acid 2-phosphate, 1 mM sodium pyru-vate, 
0.35 mM proline, 1.25 mg ml–1 BSA (Sigma-Aldrich, 
Taufkirchen, Germany), 100 U ml–1 penicillin, 100 lg ml–1 strepto-mycin, 
5 lgml–1 insulin and 10 ng ml–1 transforming growth fac-tor- 
beta 3 (TGFb-3; Miltenyi Biotec GmbH, Bergisch Gladbach, 
Germany). Differentiation was evaluated on day 14 by Safranin O 
staining (Chroma, Münster, Germany) for proteoglycan deposition 
and collagen type II immunohistochemistry as described previ-ously 
[27].
4732 E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 
2.5. Preparation of fibrin-MSC composites for implantation 
MSCs were prepared separately for every animal, to limit the 
time period between preparing the fibrinogen cell suspension 
and implantation to 20 min. The fibrinogen solution (Baxter, 
Wien, Austria) was diluted 1:2 and the thrombin (Baxter, Wien, 
Austria) was diluted 1:50 in PBS containing protease inhibitor 
cocktail (cOmplete Mini: Roche, Mannheim, Germany). 1  106 
MSCs were resuspended in 40 ll of the fibrinogen solution, kept 
at room temperature until 40 ll of the thrombin solution was 
added. The solidifying gel was transferred into the bone defect. 
Fibrin clots for the control animals were prepared without cells. 
Five additional fibrin–MSC clots were frozen at 80 C and used 
for gene expression analysis of MSCs before implantation. Three 
additional fibrin(–MSC) clots were used for histological examina-tion 
of non-implanted samples. 
2.6. Implantation of fibrin–MSC composites in the bone defect 
Rats were anaesthetized by intraperitoneal injection of 
ketamine hydrochloride (Ketamin: 100 mg kg–1 body weight, 
Orion Pharma, Hamburg, Germany) and medetomidine hydrochlo-ride 
(Domitor: 0.2 mg kg–1 body weight, Orion Pharma, Hamburg, 
Germany). A longitudinal incision (30 mm) of skin and fascia of 
the right hind leg was performed. The vastus lateralis and biceps 
femoris muscles were carefully separated and the anterolateral 
side of the femoral shaft was exposed. The attached muscles were 
stripped from the bone and a polyether ether ketone (PEEK) plate 
(Rat Fix, AO, Davos, Switzerland) was adjusted to the femur. The 
PEEK plate was held in position and pilot holes were drilled 
through both cortices along the predrilled holes of the plate. The 
plate was fixed to the bone by three screws (Rat Fix, AO, Davos, 
Switzerland) at each side. A 2 mm bone defect was created with 
a sawing wire (Rat Fix, AO, Davos, Switzerland). Bleeding was pre-vented 
by pressing a gelatin sponge (Gelita-Spon: Gelita Medical, 
Amsterdam, the Netherlands) into the bone defect for 2 min. The 
polymerizing fibrin clot was quickly transferred into the bone 
gap via a pipette and allowed to solidify for 3 min. Muscles, deep 
fascia, subcutaneous fascia and skin were closed in a standard fash-ion. 
Animals were treated once with 0.02 ml gentamicin sulfate 
(Refobacin: 80 mg, Merck, Darmstadt, Germany) for antibiosis 
and buprenorphine hydrochloride (Temgesic: 0.01 mg kg–1 body 
weight, RB Pharmaceuticals, UK) was given subcutaneously for 
analgesia. The animals were allowed full activity in their cages 
and were medicated with buprenorphine hydrochloride (Temge-sic: 
0.01 mg kg–1 body weight) for the first two postoperative days. 
At days 3, 6, 14 or 28 after surgery animals were narcotized with 
CO2 and killed by exsanguination via heart puncture. Femora were 
dissected by opening the sutures and careful stripping off of the 
surrounding tissue. Callus was carefully isolated from bone defect 
and either directly frozen at 80 C for MSC detection and gene 
expression analysis or fixed in 4% PFA for histology. 
2.7. lCT 
New bone formation of day 14 and 28 samples was evaluated 
using a Sky-Scan 1076 in vivo X-ray microtomograph (Skyscan, 
Antwerpen, Belgium). Femora were scanned with surrounding 
muscles wrapped in plastic foil using the 0.025 mm titan filter, 
with the following settings: voxel size 17.7 lm, voltage 65 kV, cur-rent 
139 lA, exposure time 280 ms, frame averaging 3. Data were 
recorded every 0.4 rotation step through 180. Reconstruction was 
performed using NRecon software (version 1.6.3.2, Skyscan, 
Antwerp, Belgium). 3-D pictures were made with CTan/CTVol 
software (Skyscan, Antwerp, Belgium). 
2.8. DNA isolation and SRY-gene-specific PCR 
Genomic DNA from day 3 and 6 callus was extracted along with 
total RNA according to the manufacturer’s protocol using the peq- 
GOLD TriFast technique (peqlab, Erlangen, Germany). Day 14 or 28 
callus was digested in lysis buffer with 100 ng ml–1 proteinase K 
(Fermentas, St Leon-Rot, Germany) at 56 C overnight and DNA 
was extracted by ethanol precipitation. A SRY-gene-specific DNA 
fragment was amplified in a standard PCR using Taq DNA Polymer-ase 
(Invitrogen, Karlsruhe, Germany). For detection of implanted 
male MSCs in defined regions, day 6 callus was separated by laser 
microdissection, and DNA was isolated using the QIAmp DNA 
Micro Kit (Qiagen GmbH, Hilden, Germany) according to the man-ufacturer’s 
protocol. DNA analogously isolated from equivalent 
sections of the control samples was used as negative control. 
SRY-gene-specific DNA was amplified by nested PCR. PCR products 
were visualized on a 2% agarose gel and their specificity was con-firmed 
by DNA sequencing. 
2.9. Gene expression analysis 
Callus was homogenized in peqGOLD TriFast and total RNA 
was extracted according to the manufacturer’s protocol (peqlab, 
Erlangen, Germany). cDNA synthesis was performed with 1 lg 
total RNA using Omniscript reverse transcriptase (0.2 U ll–1), oli-go( 
dT) primer (1 lM, both Qiagen GmbH, Hilden, Germany) and 
a ribonuclease inhibitor (RNaseOUT: 40 U ll–1, Invitrogen, Kar-lsruhe, 
Germany) according to the manufacturer’s instructions. 
Expression levels of individual genes were analyzed by quantita-tive 
PCR (Stratagene Mx3000P; Agilent Technologies, Böblingen, 
Germany). cDNA were amplified using gene-specific primer sets 
(Table 1) obtained from Eurofins (Ebersberg, Germany) and real-time 
fluorimetric intensity of SYBR green I (Thermo Scientific, 
Rockford, USA) was monitored. The apparent threshold cycles 
(Ct) of the genes of interest were compared to the Ct of the refer-ence 
gene hypoxanthine-guanine phosphoribosyltransferase-1 
(HPRT-1) in the same cDNA sample. When indicated, the percent-age 
of the reference gene expression levels shown in the figures 
refer to relative levels as a percentage of HPRT-1. Melting curves 
and agarose gel electrophoresis of the PCR products were used 
for quality control. 
2.10. Histology 
Callus was isolated from the bone defect and subjected to his-tology 
without proximal and distal bone ends to allow preparation 
of the samples without decalcification. In brief, samples were fixed 
in 4% PFA, dehydrated in a graded alcohol series and embedded in 
paraffin. In vivo orientation of the callus was maintained through-out 
the embedding process and callus was cut along this axis. For 
histological investigations, sections (5 lm) from the callus center 
were used and stained with haematoxylin and eosin (HE: 
Chroma, Münster, Germany) or Movat’s Pentachrome (all colours: 
Chroma, Münster, Germany) in a standard procedure. 
For CD31 and CD68 immunohistochemistry, digestion with 
20 ng ml–1 proteinase K (Fermentas, St Leon-Rot, Germany) for 
20 min at 37 C was used for antigen retrieval. Sections for 
CD163 staining were pre-treated with 1 mg ml–1 pronase (Roche, 
Mannheim, Germany) for 30 min at 37 C. No antigen retrieval 
was required for CCR7 staining. Unspecific binding was blocked 
for 30 min with 5% BSA for CD31, CD68 and CD163, and with 5% 
human serum for CCR7, and then sections were incubated with 
the primary antibodies CD31 (1:200; Abbiotec, San Diego, CA, 
USA), CD68 (1:100; Abcam, Cambridge, UK), CD163 (1:100, AbD 
Serotec, Oxford, UK) and CCR7 (1:250; Epitomics, Burlingame, CA, 
USA) overnight at 4 C. After washing sections were incubated with
E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 4733 
the respective biotinylated secondary antibodies (CD31 and CCR7: 
anti-rabbit IgG; CD68 and CD163: anti-mouse IgG; 1:500; both 
Dianova, Hamburg, Germany), followed by ALP–streptavidin (Vec-tastain; 
Vector Laboratories, Burlingame, CA, USA) and Fast Red 
(Roche, Mannheim, Germany). The nuclei were counterstained 
with haematoxylin (Chroma) and slides were covered in Aquatex 
(Merck, Darmstadt, Germany). Negative controls were performed 
by omitting the respective primary antibody. Histomorphometric 
evaluation was performed on HE-stained sections of two zones 
of every callus respectively (n = 10 per group). Total callus area 
was marked black and the area infiltrated by host cells was labelled 
grey using Photoshop 7.0 (Adobe Systems Inc., USA). ImageJ 1.44p 
(Wayne Rasband, National Institutes of Health, USA) was used to 
assess the total area and the infiltrated area. Cell invasion is stated 
as the percentage of infiltrated area related to total area. 
2.11. Laser microdissection 
To evaluate persistence of implanted MSCs next to invading 
host cells we searched for possible SRY-signals in the infiltrated 
area of the callus vs. the non-infiltrated part of the MSC implant. 
Appropriate parts of the tissue section were separated by laser 
microdissection. In brief, sections (10 lm) of the MSC-containing 
callus were taken up on manually foil-coated slides. After HE 
staining, the area infiltrated by host cells was separated from the 
remaining implant using a PALM MicroBeam Laser Microdissection 
System (Carl Zeiss AG, Jena, Germany). To obtain enough material 
for nested PCR, tissue collected from the infiltrated area (two sec-tions 
per sample from five animals, n = 10) or of the remaining 
fibrin clot were pooled, respectively (n = 10) and subjected to 
DNA extraction. Analogously, tissue collected from the animals 
receiving cell-free fibrin (n = 10) were pooled and further used as 
female control. 
2.12. Statistical analysis 
For comparison of gene expression between the MSC groups 
and control groups of days 3 and 6 a Kruskal–Wallis test with 
post-hoc Mann–Whitney U-tests was conducted. Only if the Krus-kal– 
Wallis tests indicated significance were data analyzed post hoc 
and corrected with the Bonferroni test. The Mann–Whitney U-test 
was furthermore applied for histomorphometric evaluation of the 
MSC group and the control group at day 6 and for comparison of 
genomic DNA content between the MSC group and the control 
group at day 14. A two-tailed significance value of P  0.05 was 
considered statistically significant. Data analysis was performed 
with SPSS for Windows 16.0 (SPSS Inc., Chicago, IL, USA). 
3. Results 
3.1. Enhanced attraction of host cells into fibrin–MSC composites 
One pool of MSCs from long bones of three male rats was pro-duced 
and extensively characterized before application in all 
experiments. Cells were strongly positive for the rat surface mark-ers 
CD90 (97%) and CD106 (35%) while almost negative for CD45 
(1.8%) and CD68 (1.7%) as expected for MSC populations 
(Fig. 1A). During in vitro osteogenesis, cells showed a strong up-regulation 
of the osteogenic markers bone sialoprotein (BSP) and 
osteocalcin (OC) and an increasing ALP enzyme activity (Fig. 1B). 
During adipogenic induction cells up-regulated mRNA levels of 
the adipogenic marker peroxisome proliferator-activated recep-tor- 
gamma (PPAR-c and deposited lipid droplets as visualized by 
Oil Red O staining (Fig. 1C). Chondrogenic differentiation in high-density 
pellets resulted in deposition of a proteoglycan- and colla-gen 
type II-rich extracellular matrix (Fig. 1D). 
MSCs were seeded in fibrin hydrogel allowing a standardized 
implantation of a defined concentration of cells into the 2 mm 
plate-stabilized femur defect of female rats. The hydrogel was 
directly transferred after starting the polymerization and solidified 
within the defect, enabling a complete filling of the bone gap by 
the implant with intimate contact to the open bone endings 
(Fig. 2A). Six days after implantation cell-seeded and non-seeded 
fibrin implants were still present in the bone defect (Fig. 2B). The 
in vivo orientation was maintained for histology (Fig. 2C). As 
shown in Fig. 3, staining of parallel non-implanted fibrin–MSC 
composites showed a homogeneous distribution of the seeded cells 
within the fibrin hydrogel (Fig. 3H inset). Six days post-implanta-tion, 
overview staining of control samples revealed little infiltra-tion 
of host cells into the MSC-free implant but indicated some 
dissolution of the fibrin from the proximal side of the bone defect 
in two out of five samples (Fig. 3A). This appeared to create space 
for accumulation of some densely packed host cells in a callus 
aside from the fibrin (Fig. 3B), while the fibrin matrix itself 
remained almost empty (Fig. 3C, D). In contrast, large numbers of 
host cells infiltrated the implant in the MSC group, especially from 
the proximal bone ending (Fig. 3E). Interestingly, in three out of 
five samples, a dense migration front was formed consisting of 
packed cells which apparently moved through the defect from 
the proximal to the distal side (Fig. 3F, G). The majority of MSCs 
located in front of this border looked viable (Fig. 3H). Behind the 
migration front, the first signs of tissue maturation and extracellu-lar 
matrix deposition were observed (Fig. 3F) with evidence of 
some proteoglycan deposition in the three most advanced 
samples according to pentachrome staining (Fig. 3I). A comparable 
Table 1 
List of oligonucleotides used for qRT-PCR analysis and SRY-gene-specific PCR. 
Gene GenBank no. Forward primer Reverse primer 
BSP [NM_012587.2] ACGCTGGAAAGTTGGAGTTAG GACCTGCTCATTTTCATCCA 
CD45 [NM_001109890.1] GCATGCATCAATCCTAGTCC GGCCATGATGTCATAGAGGA 
HPRT-1 [NM_012583.2] GCCAGACTTTGTTGGATTTG CACTTTCGCTGATGACACAA 
IL-1b [NM_031512.2] GACAAGCAACGACAAAATCC ACCGCTTTTCCATCTTCTTC 
IL-2 [NM_053836.1] AGCGTGTGTTGGATTTGACT TCTCCTCAGAAATTCCACCA 
IL-6 [NM_012589.2] AGCCAGAGTCATTCAGAGCA AGTTGGATGGTCTTGGTCCT 
IL-10 [NM_012854.2] GACGCTGTCATCGATTTCTC TTCATGGCCTTGTAGACACC 
MIP-2 [NM_053647.1] TGAAGTTTGTCTCAACCCTGA GGTGCAGTTCGTTTCTTTTCT 
OC [NM_013414.1] AGGGCAGTAAGGTGGTGAAT CTAAACGGTGGTGCCATAGA 
PPAR-c [NM_013124.3] ATAAAGTCCTTCCCGCTGAC ATCTCTTGCACAGCTTCCAC 
SRY [X89730.1] CTTTCGGAGCAGTGACAGTT CACTGATATCCCAGCTGCTT 
SRY nested [X89730.1] CTTTCGGAGCAGTGACAGTT CATGCTGGGATTCTGTTGA 
TNF-a [NM_012675.3] TCTACTGAACTTCGGGGTGA CCACCAGTTGGTTGTCTTTG 
VEGFa [NM_031836.3] CAATGATGAAGCCCTGGA CTATGCTGCAGGAAGCTCAT
4734 E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 
Fig. 1. Characterization of rat MSCs. (A) Male rat MSCs pooled from three donors were expanded for two passages, labelled with the indicated antibodies and analyzed by 
flow cytometry. Representative histograms are shown with the stained population in black and the respective controls (no primary antibody) as grey lines. (B–D) MSCs were 
subjected to osteogenic (B), adipogenic (C) or chondrogenic conditions (D) for 2 weeks. (B) For osteogenesis, expression of bone sialoprotein (BSP) and osteocalcin (OC) was 
quantified by RT-qPCR and ALP-enzyme activity was measured (n = 2 experiments). Results are expressed as mean ± standard deviation. (C) Adipogenesis was confirmed by 
gene expression of peroxisome proliferator-activated receptor-gamma (PPAR-c) and Oil Red O staining of formed lipid droplets. (D) MSCs underwent chondrogenic 
differentiation according to deposition of proteoglycans (Safranin O staining, left side) and collagen type II (immunohistochemistry, right side). Staining is representative for 
three replicates. Scale bar: 100 lm. 
Fig. 2. Implantation of fibrin–MSC composites into long bone defects in rats and processing of callus for histology. A plate-stabilized 2 mm bone defect was created in the 
right femur of 42 female rats. Either 1  106 male MSCs embedded in a fibrin gel or empty fibrin gel (80 ll) were implanted into the defect. After 3, 6, 14 or 28 days femora 
were removed and early callus was lifted out from the defect for further analysis. (A) View directly after implantation of the fibrin gel with MSCs. (B) Early callus formation 
6 days after MSC implantation. (C) For histology, day 6 callus was separated by a scalpel from the bone endings and lifted out of the bone defect. In vivo orientation was 
recorded throughout the embedding process and during histological evaluation.
E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 4735 
Fig. 3. Histological and histomorphometric evaluation of day 6 callus. (A–H) Paraffin sections of formalin-fixed tissue were stained with HE by a standard procedure. (A, E) 
Overview pictures of the callus at day 6 with the proximal explant side in the upper left and distal explant side in the lower right corner. Scale bar: 500 lm. Enlarged areas as 
defined in (A) and (E) are shown in (B)–(D) and (F)–(H). (B) Host-derived callus; (C) contact zone between host tissue and implant; (D) empty fibrin clot. Inset: HE-stained 
empty fibrin clot before implantation. (F) Highly infiltrated area from (E); (G) the migration front; (H) remaining MSC-seeded fibrin clot. Inset: HE stained MSC-seeded fibrin 
clot before implantation. Representative pictures of five replicates are shown in (A)–(H). Scale bar: 100 lm. (I) Movat’s Pentachrome staining: the infiltrated area of the three 
most mature samples of the MSC group showed deposition of glycosaminoglycan (light blue staining) next to fibrous tissue (red staining). Scale bar: 200 lm. (J) For 
histomorphometric evaluation, callus was divided into infiltrated area (dark grey) and unaffected implant (light grey). (K) Histomorphometric evaluation of the area 
infiltrated by host cells in relation to the total callus area (%, n = 10 sections per group, two per each of the five animals). Boxes represent the 25th and 75th percentile, median 
is given as horizontal line and whiskers are maximal and minimal values. Significant difference (P  0.01) between with and without MSCs is designated by two asterisks. 
maturation tissue was not seen in the fibrin control group. 
Histomorphometric assessment of the host cell-infiltrated vs. non-infiltrated 
areas (Fig. 3J) revealed a significantly larger invaded 
area in the fibrin–MSC composites compared to control samples 
(P = 0.01; Fig. 3K). 
3.2. M1 macrophages and endothelial progenitor cells as main 
invaders 
Immunohistochemical staining revealed that the fibrin–MSC 
composites were mainly infiltrated by CD68- and CD31-positive 
cells indicative of macrophages and endothelial cells (Fig. 4). 
CD68-positive macrophages were present throughout the infil-trated 
area and were enriched in the migration front (Fig. 4B). 
Among ‘‘pioneer cells’’ migrating ahead of the more crowded zone 
and penetrating deep into the fibrin clot, many cells stained CD68 
positive (Fig. 4B, lower right corner). In several areas, elongated 
CD31-positive cells apparently migrated from the proximal to the 
distal part of the implant forming primitive structures consistent 
with the formation of immature microvessels (Fig. 4D, black 
arrows). 
Macrophages can occur in a predominantly pro-inflammatory 
subtype M1 characterized by CCR7 expression and a predomi-nantly 
anti-inflammatory subtype M2 characterized by CD163 
expression [28,29]. Staining for M1 and M2 macrophage markers 
at day 6 revealed that CCR7 signals were dominant in cell-dense 
areas. CCR7-positive cells also surrounded elongating structures, 
presumably areas of primitive vessel formation and tissue matura-tion 
(Fig. 4F, black arrows). In contrast, rather few CD163-positive 
cells were present in the host cell infiltrated area. Only in the sam-ple 
with the most advanced tissue regeneration had CD163- 
positive cells accumulated, especially in the cell-dense migration 
front (Fig. 4H). In the control group, the host tissue-derived callus 
contained CD31-, CD68- and/or CCR7-positive cells but hardly any 
cells were present in the fibrin hydrogel (Fig. 4A, C, E). CD163- 
positive (M2) cells, however, were not observed (Fig. 4G). 
3.3. Cytokine expression in the early callus 
In order to analyze the trophic and immune modulatory effects 
of MSCs in the early callus, implants were harvested 3 and 6 days 
after surgery, respectively, to assess mRNA levels by quantitative 
PCR for a panel of pro- and anti-inflammatory mediators (Fig. 5). 
At the time of implantation, MSCs expressed vascular endothelial 
growth factor (VEGF), interleukin-6 (IL-6) and macrophage inflam-matory 
protein-2 (MIP-2, Fig. 5A). In vivo, median VEGF expression 
per cell increased from day 0 to day 3, reaching significantly higher 
levels in the MSC group vs. controls at day 3 (3.7-fold; P = 0.009) 
suggesting an enriched pro-angiogenic environment (Fig. 5D). 
IL-6 and MIP-2 were also expressed in vivo (Fig. 5E, F). Negligible
4736 E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 
Fig. 4. Characterization of infiltrated host cells present in the callus 6 days after MSC implantation. (A, B) Macrophages were typed by immunohistochemistry for pan-marker 
CD68. (C, D) CD31 staining was performed to detect endothelial progenitor cells as indicators for neo-vascularization (black arrows: primitive vessel formation). (E, F) M1- 
typing of the infiltrated macrophages by CCR7 staining (black arrows: positive cells surrounding maturating tissue). (G, H) M2-typing of the infiltrated macrophages by 
CD163 staining. Positive cells are expressed with Fast Red and nuclei are counterstained with haematoxylin. Staining under omission of primary antibody served as negative 
controls (insets). Pictures are representative for five replicates. Scale bar: 100 lm. 
CD45 signals characterizing leukocytes were recorded in the 
fibrin–MSC composite before implantation (Fig. 5A). Day 3 and 
day 6 in vivo samples of both groups contained CD45 signals, con-firming 
the presence of leukocytes in the explants (Fig. 5B). As can 
be predicted from our histological findings, the relative signal for 
CD45 gene expression per cell (input RNA related to reference gene 
level) was significantly higher in the fibrin-only group compared to 
the MSC group (P = 0.016), which is in line with the notion that 
most host cells seen next to the implant in the control group were 
immune cells. In the MSC group, less CD45 expression per cell was 
observed since a large fraction of the mRNA was derived from the 
implanted (CD45-negative) MSCs. In line with the higher expres-sion 
levels of CD45, significantly higher mRNA levels for IL-1b 
tumor necrosis factor-alpha (TNF-a) and IL-10 (Fig. 5G–I) were 
obvious per cell in day 3 explants without MSCs vs. the group with 
MSCs, and a similar trend was seen for IL-2 (Fig. 5C) as well as for 
day 6 samples. 
In order to determine alterations in cytokine expression levels 
per immune cell in each sample, IL-1b, TNF-a and IL-10 expression 
levels were referred to the corresponding CD45 signal of the same 
sample. After adjustment per immune cell (Fig. 5J–L) no variation 
in the pro-inflammatory cytokines IL-1b (Fig. 5J) and TNF-a 
(Fig. 5K) or the anti-inflammatory cytokine IL-10 (Fig. 5L) was evi-dent 
between the controls and the MSC group at day 3 and day 6, 
indicating no major shift of the immune response by implanted 
MSCs. 
3.4. No long-term engraftment of fibrin-implanted MSCs 
At day 14, healing of the bone defect in the MSC group was 
more advanced at the proximal bone ending compared to the distal 
side (Fig. 6A). The amount of genomic DNA in the callus increased 
considerably until day 28 in line with an increasing cell content 
(Fig. 6B). The presence of significantly more DNA in the MSC group
E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 4737 
Fig. 5. Characterization of expression of specific immunomediators in day 3 and day 6 callus. RNA was isolated from (A) fibrin–MSC composites before implantation and (B– 
M) early callus with and without MSCs at day 3 and day 6. Relative mRNA levels were detected by RT-qPCR and expression was standardized to the signals of the reference 
gene HPRT-1. (A) Basal gene expression of rat CD45, VEGF, IL-6 and MIP-2 by the MSC-population (day 0) with n = 5 replicates. (B–I) Relative mRNA levels per cell of rat CD45, 
IL-2, VEGF, IL-6, MIP-2, IL-1b, TNF-a and IL-10 in the MSC-seeded fibrin clots before implantation (day 0) and 3 or 6 days after implantation (n = 5 per time-point and group). 
(J–L) Relative expression of IL-1b, TNF-a and IL-10 per CD45-positive immune cell. Boxes represent the 25th and 75th percentile, median is given as horizontal line and 
whiskers are maximal and minimal values. Outliers (1.5- to 3-fold interquartile range, IQR) are depicted as circles, and extreme values (3-fold IQR) as diamonds. Significant 
differences (P  0.05) between groups are designated by an asterisk. 
at day 14 is in line with an enhanced host cell attraction into MSC-containing 
fibrin implants and/or may indicate more cell prolifera-tion 
in the MSC-treated defects. This difference was no longer seen 
at day 28 (Fig. 6B). When DNA from day 3, 6, 14 or 28 callus was 
analyzed for the persistence of implanted male MSCs, SRY-gene 
specific signals were detected in all MSC-containing explants from 
postoperative days 3, 6 and 14, whereas no signals were detected 
in the controls (Fig. 6C, D). Remarkably, at day 28, all explants were 
SRY-negative (Fig. 6E). Furthermore, no signals were obtained from 
the distal and proximal 2 mm bone endings (day 28), indicating no 
migration of male MSCs into the bone. Even an attempt to detect 
signals by a nested PCR approach with greatly enhanced sensitivity 
yielded no residual signals, suggesting a loss of male cells from the 
regeneration tissue and newly forming bone. 
In order to decide whether the implanted MSCs had already 
been eliminated during host cell infiltration of the fibrin–MSC 
composite or whether they persisted beside invaders, histological 
sections from day 6 were processed by laser microdissection to 
assess non-infiltrated and infiltrated areas separately. Genomic 
DNA from both areas revealed a clear SRY-gene signal after nested
4738 E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 
Fig. 6. Bone regeneration at day 14 and day 28 after surgery and persistence of implanted MSCs at the defect site. (A) 3-D reconstruction of the bone defect 14 days after 
surgery based on lCT scanning. Sides of the defect gap were rotated apart to give an open view into the proximal (left) and distal (right) bone endings. Newly formed bone 
tissue is depicted by colour. (B) Total callus was digested for DNA extraction and absolute content of genomic DNA was determined spectrophotometrically (control group: 
day 14: n = 3 and day 28: n = 2; MSC-treated group: day 14 and day 28: n = 4). Boxes represent the 25th and 75th percentile, median is given as horizontal line and whiskers 
are maximal and minimal values. Significant difference (P  0.05) between with MSCs and fibrin alone at day 14 is designated by an asterisk. (C–E) Detection of implanted 
male MSCs by SRY-gene-specific PCR at indicated time points: () animals of control group and (+) animals receiving MSCs. Arrows indicate the SRY-gene-specific fragment 
with a length of 159 bp. (F) Evaluation of implanted male MSCs persisting besides invading host cells: histological sections of the MSC-callus were subjected to laser 
microdissection to separate areas infiltrated by host cells and non-infiltrated areas. Dissected samples of similar areas were pooled, DNA was extracted and SRY-gene-specific 
nested PCR was performed. P, positive control (male MSCs); N, control (water); 0, laser microdissection cDNA control; 1, remaining fibrin clot; 2, area infiltrated by host cells; 
3, female control callus. 
PCR (Fig. 6F), while control samples from sections of cell-free 
explants were negative. This indicated that the MSCs implanted 
in fibrin hydrogel were not immediately disappearing behind the 
invasion front after host cell infiltration. 
4. Discussion 
Recent investigations in tissue regeneration have focused on the 
activation of endogenous mechanisms at the defect site by trophic 
and immunomodulatory activity of implanted MSCs [12,13]. Our 
study for the first time investigates prevailing early cellular and 
molecular actions of fibrin–MSC composites implanted into a long 
bone defect to stimulate long bone healing. Our results highlight a 
strong early impact of implanted MSCs on host M1 macrophage 
and endothelial progenitor cell recruitment into the fibrin carrier 
which may be due to the presence of higher pro-angiogenic VEGF 
levels in MSC implants vs. controls. However, other chemokines, 
such as IL-6 and MIP-2 and other molecules of the MSC secretome 
[30,31], may contribute to this effect. Furthermore, activation of 
the MSCs by the fibrin hydrogel itself should be considered [32]. 
Since local expression of pro-inflammatory cytokines IL-1b and 
TNF-a and of anti-inflammatory IL-10 per immune cell was unal-tered 
in the MSC group vs. control callus at day 3 and day 6, MSCs 
here displayed no evident early immunomodulatory activity in 
fully immune competent animals. Accelerated early tissue matura-tion 
in the MSC group occurred despite a loss of implanted male 
MSCs before day 28 in our model. Overall, the fibrin carrier proved 
attractive for MSC implantation into bone defects permitting 
generation of a trophic and pro-angiogenic microenvironment by 
which MSCs stimulated implant integration and endogenous repair 
mechanisms without evident early immunomodulation or long-term 
osteogenic contribution to the repair tissue. 
4.1. MSCs as a trigger for cell invasion into fibrin hydrogel 
Cellular and molecular mechanisms of natural bone remodel-ling 
revealed a crucial role for resident tissue macrophages for full 
functional differentiation of osteoblasts and their maintenance 
in vivo [33]. In wound healing, one important early wave of 
cell migration towards a site of tissue injury is conducted by
E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 4739 
macrophages around 2–6 days after injury [34]. Macrophage-secreted 
cytokines affect endothelial cell proliferation, angiogene-sis 
and collagen synthesis [34], suggesting that they may act in 
concert with other resident cells to stimulate vascularization. Our 
model took advantage of an empty and a MSC-seeded fibrin 
hydrogel clot, allowing us to distinguish early arriving and possibly 
faster migrating cells penetrating deep into the implant, from later 
arriving and/or later migrating cells that are still closer to the 
implant edges on day 6. We consistently observed earlier host cell 
infiltration from the proximal defect side which is possibly caused 
by a relative shortage of blood-borne invading host cells at the dis-tal 
defect ending after disruption of blood flow by defect creation. 
Remarkably, host cell infiltration was only initiated in fibrin–MSC 
composites while host cells apparently showed little interest in 
invading a cell-free fibrin hydrogel in a bone defect. This demon-strated 
that signals from viable MSCs represented an important 
trigger for implant integration and remodelling, emphasizing the 
importance of—most likely—soluble factors as trophic mediators 
of MSC action. Most importantly, although the local number of 
osteoprogenitor cells in the bone cavity is high and may suffice 
to initiate bone healing, defect filling with empty fibrin hydrogel 
alone cannot be recommended due to its rather inert persistence 
without evident cell invasion. 
A large panel of trophic factors is secreted by MSCs [30,31] of 
which IL-6, VEGF and the neutrophil-attracting factor MIP-2 were 
here shown to be expressed by the fibrin-embedded MSCs before 
implantation. This first demonstration of significantly higher VEGF 
mRNA levels in vivo and differential expression patterns between 
fibrin–MSC composites and cell-free controls suggest VEGF as an 
important candidate molecule for endothelial cell attraction, angi-ogenesis, 
implant remodelling and bone regeneration in line with 
the literature [35–37]. Whether the higher VEGF mRNA levels 
resulted from an up-regulation of VEGF in MSCs or is contributed 
by attracted macrophages and endothelial cells remains to be 
determined. However, MSCs not only secrete growth factors and 
chemokines but also produce matrix-degrading proteases and 
extracellular matrix molecules which might act on angiogenesis 
and help in cell migration [38,39]. Overall our study, thus, demon-strates 
that fibrin supports MSCs in organizing a beneficial envi-ronment 
for bone regeneration by facilitating host macrophage 
invasion relevant for tissue remodelling and enhancing endothelial 
progenitor cell migration indispensible for new blood vessel 
formation. 
4.2. The cascade of invading cells 
Pioneering cells penetrating deep into the implant were mainly 
CD68-positive macrophages accompanied and followed by CD31- 
positive endothelial progenitor cells. To our knowledge this is the 
first study demonstrating a cell-dense migration front as a domi-nant 
structure of in vivo integration and remodelling of a fibrin– 
MSC composite. This cell-dense structure developing always from 
the proximal bone ending may thus be a special feature of long 
bone healing or be related to the fibrin hydrogel. At ectopic sites 
in immune-competent [40,41] and immune-deficient host animals 
[42,43] no cell-dense invasion front of host cells into tissue engi-neering 
constructs has been observed. This structure may so far, 
however, have escaped detection either because only late observa-tion 
time points were chosen or because no histology was 
performed at the early time points. 
In many stress situations monocytes enter the damaged area 
and differentiate into a spectrum of mononuclear phagocytes. 
These newly recruited cells usually exhibit pro-inflammatory 
action, and in the tissue inflammatory mediators such as IL-1, 
IL-12 and TNF-a must be balanced with the need to protect tissue 
integrity [44]. Our data for the first time provide evidence that 
pro-inflammatory M1 macrophages also prevailed early during 
long bone healing, while CD163-positive ‘‘anti-inflammatory’’ M2 
macrophages [45] remained rare at day 6. Macrophages remain 
responsive to further stimuli after their initial activation, are by 
no means restricted to only these two categories [46], and M1 mac-rophages 
can convert themselves into anti-inflammatory macro-phages 
with an M2 wound-healing phenotype [47]. Further 
studies at later time points are important to fully understand the 
cascade of invading cells and their fate during long bone healing. 
4.3. Immunomodulation by fibrin-based MSC implantation 
To analyze the effect of fibrin-implanted MSCs on the inflam-matory 
phase of bone healing, we looked for gene expression of 
CD45 as well as IL-1b, TNF-a and IL-10 which represent relevant 
immune mediators during early bone regeneration [48,49]. The 
natural callus adjacent to cell-free fibrin hydrogel in the control 
group contained a higher concentration of CD45-positive immune 
cells than MSC-seeded implants and thus contained higher mRNA 
levels of pro- and anti-inflammatory cytokines at day 3. The pres-ence 
of MSCs in the cell-seeded fibrin hydrogel did not only dilute 
the inflammatory cytokine-producing immune cells of the early 
callus, but can also be expected to actively contribute with a broad 
panel of trophic factors. These beneficial effects ultimately pro-duced 
a stimulatory microenvironment evident from histology, 
organizing remodelling of the fibrin hydrogel. Suppressive actions 
of MSCs towards immune cells, as reported in other settings 
in vitro and in vivo [50], were not apparent at this stage, since 
the average levels of IL-1b, TNF-a and IL-10 per CD45-positive 
immune cell were unaltered. 
Although Sprague–Dawley rats have frequently been used for 
studying outcome after cell and tissue transplantation between 
animals with no evidence for strain-intern rejection [51–53], we 
looked for local gene expression of the T-cell mediator IL-2 as an 
early rejection marker after allogeneic implantation of MSCs. IL-2 
mRNA levels were not increased in the MSC group at day 3 and 
day 6, indicating that no acute T-cell-mediated rejection of the 
implanted MSCs occurred within the fibrin hydrogel within the 
first 6 days, in line with the findings in a syngeneic ectopic mouse 
model [40]. Overall, this argues in favour of a microenvironment in 
MSC-seeded fibrin hydrogel without early silencing of host 
immune cells at days 3 and 6. 
4.4. Tissue maturation and the fate of fibrin-implanted MSCs 
According to detection of the implanted MSCs in our histologi-cal 
sections at day 6, fibrin hydrogel was stable and cell supportive 
enough to enable a local persistence of implanted MSCs for at least 
6 days in a bone defect without connection to the vasculature. Fur-thermore, 
according to detection of male DNA in laser-dissected 
tissue areas behind the migration front, arriving immune cells 
did not immediately degrade implanted MSCs during invasion. 
However, since all cells were lost within 4 weeks after implanta-tion, 
either their limited lifespan, a failure of re-adaptation to 
changing needs during tissue maturation or other weaknesses 
may have caused their ultimate replacement by host-derived 
osteoprogenitor cells localized in adjacent bone marrow. In ectopic 
models a mineral component such as beta-tricalcium phosphate 
(b-TCP) or hydroxyapatite-tricalcium phosphate (HA/TCP) is 
required for long-term MSC engraftment and MSC-mediated bone 
formation [18,54]. Due to our interest in the early immunomodu-latory 
actions of implanted MSCs, we decided to use a fibrin carrier 
without ceramic particles since some inflammatory activity has 
been reported for b-TCP [55,56] which would bias our results. In 
our setting it remains unclear whether or not fibrin-implanted 
MSCs have physically contributed to the maturing osteogenic
4740 E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 
tissue of the MSC group until day 14 before replacement by host 
bone. Although sex-mismatched animal models are well estab-lished 
for studying MSC-mediated bone formation [21,57] and ele-gantly 
permit the detection of persisting male cells in female 
recipients without a need for cell labelling [58], a long-term rejec-tion 
of male cells by the female immune system may have contrib-uted 
to our result. The transient role of fibrin-implanted MSCs seen 
here is, however, in line with data from genetically engineered 
ASCs, which effectively stimulated femoral bone healing in rabbits, 
despite some induction of a humoral- and cell-mediated immune 
response and limited cell persistence over 4 weeks [59]. 
5. Conclusions 
In summary, by using fibrin hydrogel—known for its biocom-patibility, 
biodegradability and cell support for implantation of 
MSCs into a long bone defect—we identified attraction of M1 mac-rophages 
and endothelial progenitor cells into the implant as a 
main early beneficial action of fibrin–MSC composites, promoting 
implant integration, angiogenesis and tissue maturation during 
bone regeneration. No evidence for an early immune suppression 
by fibrin-implanted MSCs was obtained, as expression levels of 
the pro-inflammatory cytokines IL-1b and TNF-a as well as the 
anti-inflammatory cytokine IL-10 by the invading immune cells 
were unaltered. Trophic and pro-angiogenic stimulation during 
bone regeneration occurred independently of a long-term engraft-ment 
of the implanted MSCs at the defect site. Early recruitment of 
host repair cells into fibrin hydrogel was most likely stimulated 
through the secretion of bioactive factors including VEGF whose 
elevated levels may have generated a pro-angiogenic environment 
beneficial for bone regeneration. With this advanced understand-ing 
of the mechanisms involved in fibrin hydrogel-based MSC-sup-ported 
bone regeneration, we suggest fibrin hydrogel can serve as 
an attractive carrier for MSC-based tissue engineering approaches 
in long bone repair, supporting early trophic and pro-angiogenic 
activities to improve clinical treatment of atrophic pseudarthrosis, 
bone injury or tumor resection. 
Disclosure 
The authors indicate no potential conflicts of interest. 
Acknowledgements 
We thank Svenja Schäfer and Viviana Grajales for their support 
with the surgery and with animal care. Furthermore we thank 
Nicole Buchta and Birgit Frey for their help with the experiments 
and Simone Gantz for her statistical support. This study (grant RI 
707/8-1) was funded by the priority program SPP1468 ‘‘Immuno-bone’’ 
of the German Research Foundation (DFG). 
Appendix A. Figures with essential color discrimination 
Certain figures in this article, particularly Figs. 1, 2 and 6 are 
difficult to interpret in black and white. The full color images can 
be found in the on-line version, at http://dx.doi.org/10.1016/ 
j.actbio.2014.07.017. 
References 
[1] Patterson TE, Kumagai K, Griffith L, Muschler GF. Cellular strategies for 
enhancement of fracture repair. J Bone Joint Surg Am 2008;90(Suppl. 1):111–9. 
[2] Wu X, Ren J, Li J. Fibrin glue as the cell-delivery vehicle for mesenchymal 
stromal cells in regenerative medicine. Cytotherapy 2012;14:555–62. 
[3] Bensaid W, Triffitt JT, Blanchat C, Oudina K, Sedel L, Petite H. A biodegradable 
fibrin scaffold for mesenchymal stem cell transplantation. Biomaterials 
2003;24:2497–502. 
[4] Dickhut A, Gottwald E, Steck E, Heisel C, Richter W. Chondrogenesis of 
mesenchymal stem cells in gel-like biomaterials in vitro and in vivo. Front 
Biosci 2008;13:4517–28. 
[5] Sierra DH. Fibrin sealant adhesive systems: a review of their chemistry, 
material properties and clinical applications. J Biomater Appl 1993;7:309–52. 
[6] Steck E, Fischer J, Lorenz H, Gotterbarm T, Jung M, Richter W. Mesenchymal 
stem cell differentiation in an experimental cartilage defect: restriction of 
hypertrophy to bone-close neocartilage. Stem Cells Dev 2009;18:969–78. 
[7] Jung M, Kaszap B, Redohl A, Steck E, Breusch S, Richter W, et al. Enhanced early 
tissue regeneration after matrix-assisted autologous mesenchymal stem cell 
transplantation in full thickness chondral defects in a minipig model. Cell 
Transplant 2009;18:923–32. 
[8] Friedenstein AJ, Petrakova KV, Kurolesova AI, Frolova GP. Heterotopic of bone 
marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. 
Transplantation 1968;6:230–47. 
[9] Sacchetti B et al. Self-renewing osteoprogenitors in bone marrow sinusoids can 
organize a hematopoietic microenvironment. Cell 2007;131:324–36. 
[10] Dominici M et al. Minimal criteria for defining multipotent mesenchymal 
stromal cells. The International Society for Cellular Therapy position 
statement. Cytotherapy 2006;8:315–7. 
[11] Pittenger MF et al. Multilineage potential of adult human mesenchymal stem 
cells. Science 1999;284:143–7. 
[12] Meirelles Lda S, Fontes AM, Covas DT, Caplan AI. Mechanisms involved in the 
therapeutic properties of mesenchymal stem cells. Cytokine Growth Factor 
Rev 2009;20:419–27. 
[13] Caplan AI, Correa D. The MSC: an injury drugstore. Cell Stem Cell 2011;9:11–5. 
[14] Kadiyala S, Jaiswal N, Bruder SP. Culture-expanded, bone marrow-derived 
mesenchymal stem cells can regenerate a critical-sized segmental bone defect. 
Tissue Eng 1997;3:173–85. 
[15] Bruder SP, Kraus KH, Goldberg VM, Kadiyala S. The effect of implants loaded 
with autologous mesenchymal stem cells on the healing of canine segmental 
bone defects. J Bone Joint Surg Am 1998;80:985–96. 
[16] Quarto R et al. Repair of large bone defects with the use of autologous bone 
marrow stromal cells. N Engl J Med 2001;344:385–6. 
[17] Marcacci M et al. Stem cells associated with macroporous bioceramics for long 
bone repair: 6- to 7-year outcome of a pilot clinical study. Tissue Eng 
2007;13:947–55. 
[18] Janicki P, Boeuf S, Steck E, Egermann M, Kasten P, Richter W. Prediction of 
in vivo bone forming potency of bone marrow-derived human mesenchymal 
stem cells. Eur Cell Mater 2011;21:488–507. 
[19] Janicki P, Kasten P, Kleinschmidt K, Luginbuehl R, Richter W. Chondrogenic 
pre-induction of human mesenchymal stem cells on beta-TCP: enhanced bone 
quality by endochondral heterotopic bone formation. Acta Biomater 
2010;6:3292–301. 
[20] Giannoni P, Scaglione S, Daga A, Ilengo C, Cilli M, Quarto R. Short-time survival 
and engraftment of bone marrow stromal cells in an ectopic model of bone 
regeneration. Tissue Eng Part A 2010;16:489–99. 
[21] Boukhechba F et al. Fate of bone marrow stromal cells in a syngenic model of 
bone formation. Tissue Eng Part A 2011;17:2267–78. 
[22] Zimmermann CE, Gierloff M, Hedderich J, Acil Y, Wiltfang J, Terheyden H. 
Survival of transplanted rat bone marrow-derived osteogenic stem cells 
in vivo. Tissue Eng Part A 2011;17:1147–56. 
[23] Prockop DJ. Repair of tissues by adult stem/progenitor cells (MSCs): 
controversies, myths, and changing paradigms. Mol Ther 2009;17:939–46. 
[24] Hughes PC, Tanner JM. The assessment of skeletal maturity in the growing rat. 
J Anat 1970;106:371–402. 
[25] Dobson KR, Reading L, Haberey M, Marine X, Scutt A. Centrifugal isolation of 
bone marrow from bone: an improved method for the recovery and 
quantitation of bone marrow osteoprogenitor cells from rat tibiae and 
femurae. Calcif Tissue Int 1999;65:411–3. 
[26] Meirelles Lda S, Nardi NB. Murine marrow-derived mesenchymal stem cell: 
isolation, in vitro expansion, and characterization. Br J Haematol 
2003;123:702–11. 
[27] Winter A et al. Cartilage-like gene expression in differentiated human stem 
cell spheroids: a comparison of bone marrow-derived and adipose tissue-derived 
stromal cells. Arthritis Rheum 2003;48:418–29. 
[28] Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine 
system in diverse forms of macrophage activation and polarization. Trends 
Immunol 2004;25:677–86. 
[29] Brown BN, Valentin JE, Stewart-Akers AM, McCabe GP, Badylak SF. 
Macrophage phenotype and remodeling outcomes in response to biologic 
scaffolds with and without a cellular component. Biomaterials 
2009;30:1482–91. 
[30] Wagner W et al. Molecular and secretory profiles of human mesenchymal 
stromal cells and their abilities to maintain primitive hematopoietic 
progenitors. Stem Cells 2007;25:2638–47. 
[31] Kinnaird T, Stabile E, Burnett MS, Lee CW, Barr S, Fuchs S, et al. Marrow-derived 
stromal cells express genes encoding a broad spectrum of arteriogenic 
cytokines and promote in vitro and in vivo arteriogenesis through paracrine 
mechanisms. Circ Res 2004;94:678–85. 
[32] Huang NF, Chu J, Lee RJ, Li S. Biophysical and chemical effects of fibrin on 
mesenchymal stromal cell gene expression. Acta Biomater 2010;6:3947–56. 
[33] Raggatt LJ, Partridge NC. Cellular and molecular mechanisms of bone 
remodeling. J Biol Chem 2010;285:25103–8. 
[34] Park JE, Barbul A. Understanding the role of immune regulation in wound 
healing. Am J Surg 2004;187:11S–6S.
E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 4741 
[35] Geiger F et al. VEGF producing bone marrow stromal cells (BMSC) enhance 
vascularization and resorption of a natural coral bone substitute. Bone 
2007;41:516–22. 
[36] Kumar S, Wan C, Ramaswamy G, Clemens TL, Ponnazhagan S. Mesenchymal 
stem cells expressing osteogenic and angiogenic factors synergistically 
enhance bone formation in a mouse model of segmental bone defect. Mol 
Ther 2010;18:1026–34. 
[37] Lin CY et al. Long-term tracking of segmental bone healing mediated by 
genetically engineered adipose-derived stem cells: focuses on bone 
remodeling and potential side effects. Tissue Eng Part A 2014;20:1392–402. 
[38] Choi YA et al. Secretome analysis of human BMSCs and identification of SMOC1 
as an important ECM protein in osteoblast differentiation. J Proteome Res 
2010;9:2946–56. 
[39] Polacek M, Bruun JA, Elvenes J, Figenschau Y, Martinez I. The secretory profiles 
of cultured human articular chondrocytes and mesenchymal stem cells: 
implications for autologous cell transplantation strategies. Cell Transplant 
2011;20:1381–93. 
[40] Tasso R et al. Recruitment of a host’s osteoprogenitor cells using exogenous 
mesenchymal stem cells seeded on porous ceramic. Tissue Eng Part A 
2009;15:2203–12. 
[41] Tasso R, Fais F, Reverberi D, Tortelli F, Cancedda R. The recruitment of two 
consecutive and different waves of host stem/progenitor cells during the 
development of tissue-engineered bone in a murine model. Biomaterials 
2010;31:2121–9. 
[42] Tortelli F, Tasso R, Loiacono F, Cancedda R. The development of tissue-engineered 
bone of different origin through endochondral and 
intramembranous ossification following the implantation of mesenchymal 
stem cells and osteoblasts in a murine model. Biomaterials 2010;31:242–9. 
[43] Ghanaati S et al. Scaffold vascularization in vivo driven by primary human 
osteoblasts in concert with host inflammatory cells. Biomaterials 
2011;32:8150–60. 
[44] Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage 
subsets. Nat Rev Immunol 2011;11:723–37. 
[45] Ambarus CA et al. Systematic validation of specific phenotypic markers for 
in vitro polarized human macrophages. J Immunol Methods 2012;375: 
196–206. 
[46] Novak ML, Koh TJ. Macrophage phenotypes during tissue repair. J Leukoc Biol 
2013;93:875–81. 
[47] Arnold L et al. Inflammatory monocytes recruited after skeletal muscle injury 
switch into antiinflammatory macrophages to support myogenesis. J Exp Med 
2007;204:1057–69. 
[48] Mountziaris PM, Mikos AG. Modulation of the inflammatory response for 
enhanced bone tissue regeneration. Tissue Eng Part B Rev 2008;14:179–86. 
[49] Kolar P et al. The early fracture hematoma and its potential role in fracture 
healing. Tissue Eng Part B Rev 2010;16:427–34. 
[50] Ghannam S, Bouffi C, Djouad F, Jorgensen C, Noel D. Immunosuppression by 
mesenchymal stem cells: mechanisms and clinical applications. Stem Cell Res 
Ther 2010;1:2. 
[51] Larsson J, Juliusson B, Holmdahl R, Ehinger B. MHC expression in syngeneic 
and allogeneic retinal cell transplants in the rat. Graefes Arch Clin Exp 
Ophthalmol 1999;237:82–5. 
[52] Troum S, Dalton Jr ML. Osteogenesis in a rat model: use of bone marrow cells 
and biodegradable gelatin matrix carrier. J South Orthop Assoc 2001;10: 
37–43. 
[53] Yang Y, Rossi FM, Putnins EE. Periodontal regeneration using engineered bone 
marrow mesenchymal stromal cells. Biomaterials 2010;31:8574–82. 
[54] Krebsbach PH, Kuznetsov SA, Satomura K, Emmons RV, Rowe DW, Robey PG. 
Bone formation in vivo: comparison of osteogenesis by transplanted mouse 
and human marrow stromal fibroblasts. Transplantation 1997;63:1059–69. 
[55] Lange T, Schilling AF, Peters F, Haag F, Morlock MM, Rueger JM, et al. 
Proinflammatory and osteoclastogenic effects of beta-tricalciumphosphate 
and hydroxyapatite particles on human mononuclear cells in vitro. 
Biomaterials 2009;30:5312–8. 
[56] Curran JM, Gallagher JA, Hunt JA. The inflammatory potential of biphasic 
calcium phosphate granules in osteoblast/macrophage co-culture. 
Biomaterials 2005;26:5313–20. 
[57] Nakamura A et al. Cell sheet transplantation of cultured mesenchymal stem 
cells enhances bone formation in a rat nonunion model. Bone 2010; 
46:418–24. 
[58] An J, Beauchemin N, Albanese J, Abney TO, Sullivan AK. Use of a rat cDNA probe 
specific for the Y chromosome to detect male-derived cells. J Androl 
1997;18:289–93. 
[59] Lin CY et al. Immune responses during healing of massive segmental femoral 
bone defects mediated by hybrid baculovirus-engineered ASCs. Biomaterials 
2012;33:7422–34.

Contenu connexe

Tendances

2007 Bailey Tissue Engineering
2007 Bailey Tissue Engineering2007 Bailey Tissue Engineering
2007 Bailey Tissue EngineeringMark Bailey
 
Orthopaedic tissue engineering
Orthopaedic tissue engineeringOrthopaedic tissue engineering
Orthopaedic tissue engineeringrajusvmc
 
Bone Marrow Stromal Stem cells (bMSCs) therapy for musculoskeletal problems ...
Bone Marrow Stromal Stem cells (bMSCs) therapy for musculoskeletal problems ...Bone Marrow Stromal Stem cells (bMSCs) therapy for musculoskeletal problems ...
Bone Marrow Stromal Stem cells (bMSCs) therapy for musculoskeletal problems ...Asosiasi Sel Punca Indonesia
 
bone and_cartilage_tissue_engineering by Sumit
 bone and_cartilage_tissue_engineering by Sumit bone and_cartilage_tissue_engineering by Sumit
bone and_cartilage_tissue_engineering by SumitDcrust
 
bone tissue engineering
bone tissue engineeringbone tissue engineering
bone tissue engineeringSomdutt Sharma
 
Study of cell therapy assisted regeneration of cartilage
Study of cell therapy assisted regeneration of cartilageStudy of cell therapy assisted regeneration of cartilage
Study of cell therapy assisted regeneration of cartilageSourabh Harankhedkar
 
Excellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterialsExcellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterialsComprehensiveBiologi
 
Cartilage Repair using Stem cell & Orthobiologics
Cartilage Repair using Stem cell & OrthobiologicsCartilage Repair using Stem cell & Orthobiologics
Cartilage Repair using Stem cell & OrthobiologicsVaibhav Bagaria
 
Peripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcordPeripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcordComprehensiveBiologi
 
Stemcells research paper on avn
Stemcells research paper on avnStemcells research paper on avn
Stemcells research paper on avnDr Pradeep Mahajan
 
Role of Mesenchymal Stem Cells in Orthopedic Treatment of Tendon and Ligament...
Role of Mesenchymal Stem Cells in Orthopedic Treatment of Tendon and Ligament...Role of Mesenchymal Stem Cells in Orthopedic Treatment of Tendon and Ligament...
Role of Mesenchymal Stem Cells in Orthopedic Treatment of Tendon and Ligament...Daniel Janisz
 
Rotator cuff repair using a stem cell approach
Rotator cuff repair using a stem cell approachRotator cuff repair using a stem cell approach
Rotator cuff repair using a stem cell approachZakary Bondy
 
Treatment of osteochondral lesions. past, present and future.
Treatment of osteochondral lesions. past, present and future.Treatment of osteochondral lesions. past, present and future.
Treatment of osteochondral lesions. past, present and future.Elhussein E. Mahmoud
 
Dr. A. Mobasheri Nc3 Rs And Bbsrc Symposium 1 2 April 2009 Final Version
Dr. A. Mobasheri   Nc3 Rs And Bbsrc Symposium 1 2 April 2009 Final VersionDr. A. Mobasheri   Nc3 Rs And Bbsrc Symposium 1 2 April 2009 Final Version
Dr. A. Mobasheri Nc3 Rs And Bbsrc Symposium 1 2 April 2009 Final VersionAli Mobasheri
 
Tissue engineering in endodontics
Tissue engineering in endodonticsTissue engineering in endodontics
Tissue engineering in endodonticsRachna Chaurasia
 
Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)ComprehensiveBiologi
 

Tendances (20)

2007 Bailey Tissue Engineering
2007 Bailey Tissue Engineering2007 Bailey Tissue Engineering
2007 Bailey Tissue Engineering
 
Orthopaedic tissue engineering
Orthopaedic tissue engineeringOrthopaedic tissue engineering
Orthopaedic tissue engineering
 
Bone Marrow Stromal Stem cells (bMSCs) therapy for musculoskeletal problems ...
Bone Marrow Stromal Stem cells (bMSCs) therapy for musculoskeletal problems ...Bone Marrow Stromal Stem cells (bMSCs) therapy for musculoskeletal problems ...
Bone Marrow Stromal Stem cells (bMSCs) therapy for musculoskeletal problems ...
 
bone and_cartilage_tissue_engineering by Sumit
 bone and_cartilage_tissue_engineering by Sumit bone and_cartilage_tissue_engineering by Sumit
bone and_cartilage_tissue_engineering by Sumit
 
Stemcells in Orthopaedic suergery.
Stemcells  in Orthopaedic suergery.Stemcells  in Orthopaedic suergery.
Stemcells in Orthopaedic suergery.
 
bone tissue engineering
bone tissue engineeringbone tissue engineering
bone tissue engineering
 
Study of cell therapy assisted regeneration of cartilage
Study of cell therapy assisted regeneration of cartilageStudy of cell therapy assisted regeneration of cartilage
Study of cell therapy assisted regeneration of cartilage
 
Excellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterialsExcellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterials
 
Cartilage Repair using Stem cell & Orthobiologics
Cartilage Repair using Stem cell & OrthobiologicsCartilage Repair using Stem cell & Orthobiologics
Cartilage Repair using Stem cell & Orthobiologics
 
Peripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcordPeripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcord
 
Stemcells research paper on avn
Stemcells research paper on avnStemcells research paper on avn
Stemcells research paper on avn
 
Role of Mesenchymal Stem Cells in Orthopedic Treatment of Tendon and Ligament...
Role of Mesenchymal Stem Cells in Orthopedic Treatment of Tendon and Ligament...Role of Mesenchymal Stem Cells in Orthopedic Treatment of Tendon and Ligament...
Role of Mesenchymal Stem Cells in Orthopedic Treatment of Tendon and Ligament...
 
Stem cell research
Stem cell researchStem cell research
Stem cell research
 
Rotator cuff repair using a stem cell approach
Rotator cuff repair using a stem cell approachRotator cuff repair using a stem cell approach
Rotator cuff repair using a stem cell approach
 
Treatment of osteochondral lesions. past, present and future.
Treatment of osteochondral lesions. past, present and future.Treatment of osteochondral lesions. past, present and future.
Treatment of osteochondral lesions. past, present and future.
 
Whartons jelly ms_cs_a4_m
Whartons jelly ms_cs_a4_mWhartons jelly ms_cs_a4_m
Whartons jelly ms_cs_a4_m
 
Dr. A. Mobasheri Nc3 Rs And Bbsrc Symposium 1 2 April 2009 Final Version
Dr. A. Mobasheri   Nc3 Rs And Bbsrc Symposium 1 2 April 2009 Final VersionDr. A. Mobasheri   Nc3 Rs And Bbsrc Symposium 1 2 April 2009 Final Version
Dr. A. Mobasheri Nc3 Rs And Bbsrc Symposium 1 2 April 2009 Final Version
 
Tissue assembly in microgravity
Tissue assembly in microgravityTissue assembly in microgravity
Tissue assembly in microgravity
 
Tissue engineering in endodontics
Tissue engineering in endodonticsTissue engineering in endodontics
Tissue engineering in endodontics
 
Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)
 

En vedette

NOVELA. Equilibrio en la sombra. ERNESTO RAMÍREZ
NOVELA. Equilibrio en la sombra. ERNESTO RAMÍREZNOVELA. Equilibrio en la sombra. ERNESTO RAMÍREZ
NOVELA. Equilibrio en la sombra. ERNESTO RAMÍREZErnesto Ramirez
 
Star Greetings - Working perspectives
Star Greetings - Working perspectives Star Greetings - Working perspectives
Star Greetings - Working perspectives Star Cooperation GmbH
 
Cristina y la sardina
Cristina y la sardinaCristina y la sardina
Cristina y la sardinaanprape
 
Tech MeetUp: Agile Methodologies in eCommerce
Tech MeetUp: Agile Methodologies in eCommerceTech MeetUp: Agile Methodologies in eCommerce
Tech MeetUp: Agile Methodologies in eCommerceSantex Group
 
gorilla-journal-48-english
gorilla-journal-48-englishgorilla-journal-48-english
gorilla-journal-48-englishJoseph Arinaitwe
 
Preguntas y respuestas sobre la ley de ilicitos cambiarios
Preguntas y respuestas sobre la ley de ilicitos cambiariosPreguntas y respuestas sobre la ley de ilicitos cambiarios
Preguntas y respuestas sobre la ley de ilicitos cambiariosOscar Barreto
 
Edu møde 2011 finale
Edu møde 2011 finaleEdu møde 2011 finale
Edu møde 2011 finaleHans Pedersen
 
ASE2014 Modelos parentales y demandas educativas - Daniel Gabaldón Estevan; M...
ASE2014 Modelos parentales y demandas educativas - Daniel Gabaldón Estevan; M...ASE2014 Modelos parentales y demandas educativas - Daniel Gabaldón Estevan; M...
ASE2014 Modelos parentales y demandas educativas - Daniel Gabaldón Estevan; M...Daniel Gabadón-Estevan
 
¿Qué esperar del futuro de las ciudades?
¿Qué esperar del futuro de las ciudades?¿Qué esperar del futuro de las ciudades?
¿Qué esperar del futuro de las ciudades?Manu Fernández
 
Bảng giá gia công các loại chậu rửa inox
Bảng giá gia công các loại chậu rửa inoxBảng giá gia công các loại chậu rửa inox
Bảng giá gia công các loại chậu rửa inoxGia công inox OSATAVN.COM
 
My circle of friends
My circle of friendsMy circle of friends
My circle of friendsfplatin63
 

En vedette (20)

Arashi magazine
Arashi magazineArashi magazine
Arashi magazine
 
NOVELA. Equilibrio en la sombra. ERNESTO RAMÍREZ
NOVELA. Equilibrio en la sombra. ERNESTO RAMÍREZNOVELA. Equilibrio en la sombra. ERNESTO RAMÍREZ
NOVELA. Equilibrio en la sombra. ERNESTO RAMÍREZ
 
Star Greetings - Working perspectives
Star Greetings - Working perspectives Star Greetings - Working perspectives
Star Greetings - Working perspectives
 
Secretos de la santa cena
Secretos de la santa cenaSecretos de la santa cena
Secretos de la santa cena
 
Lolek
LolekLolek
Lolek
 
El Desacuerdo #1
El Desacuerdo #1El Desacuerdo #1
El Desacuerdo #1
 
Cristina y la sardina
Cristina y la sardinaCristina y la sardina
Cristina y la sardina
 
Nbms intro
Nbms introNbms intro
Nbms intro
 
Sumita's Graphic Design Portfolio
Sumita's Graphic Design PortfolioSumita's Graphic Design Portfolio
Sumita's Graphic Design Portfolio
 
Tech MeetUp: Agile Methodologies in eCommerce
Tech MeetUp: Agile Methodologies in eCommerceTech MeetUp: Agile Methodologies in eCommerce
Tech MeetUp: Agile Methodologies in eCommerce
 
gorilla-journal-48-english
gorilla-journal-48-englishgorilla-journal-48-english
gorilla-journal-48-english
 
La ofen a
La ofen aLa ofen a
La ofen a
 
Certificación Galicia QCOACH - IESEC
Certificación Galicia QCOACH - IESECCertificación Galicia QCOACH - IESEC
Certificación Galicia QCOACH - IESEC
 
Preguntas y respuestas sobre la ley de ilicitos cambiarios
Preguntas y respuestas sobre la ley de ilicitos cambiariosPreguntas y respuestas sobre la ley de ilicitos cambiarios
Preguntas y respuestas sobre la ley de ilicitos cambiarios
 
2016 04 --curso_cliente_movil_v3
2016 04 --curso_cliente_movil_v32016 04 --curso_cliente_movil_v3
2016 04 --curso_cliente_movil_v3
 
Edu møde 2011 finale
Edu møde 2011 finaleEdu møde 2011 finale
Edu møde 2011 finale
 
ASE2014 Modelos parentales y demandas educativas - Daniel Gabaldón Estevan; M...
ASE2014 Modelos parentales y demandas educativas - Daniel Gabaldón Estevan; M...ASE2014 Modelos parentales y demandas educativas - Daniel Gabaldón Estevan; M...
ASE2014 Modelos parentales y demandas educativas - Daniel Gabaldón Estevan; M...
 
¿Qué esperar del futuro de las ciudades?
¿Qué esperar del futuro de las ciudades?¿Qué esperar del futuro de las ciudades?
¿Qué esperar del futuro de las ciudades?
 
Bảng giá gia công các loại chậu rửa inox
Bảng giá gia công các loại chậu rửa inoxBảng giá gia công các loại chậu rửa inox
Bảng giá gia công các loại chậu rửa inox
 
My circle of friends
My circle of friendsMy circle of friends
My circle of friends
 

Similaire à 1 s2.0-s1742706114003110-main(4)

Mesenchymal stem cells in Orthopaedics
Mesenchymal stem cells in OrthopaedicsMesenchymal stem cells in Orthopaedics
Mesenchymal stem cells in OrthopaedicsDr. Bushu Harna
 
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...Lipogems Equine & Lipogems Canine
 
Stem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder DysfunctionStem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder DysfunctionAnkita-rastogi
 
Stem cells for Bladder Dysfunction
Stem cells for Bladder DysfunctionStem cells for Bladder Dysfunction
Stem cells for Bladder DysfunctionAnkita-rastogi
 
Autologous Mesenchymal Stem Cells in Orthopaedics
Autologous Mesenchymal Stem Cells in OrthopaedicsAutologous Mesenchymal Stem Cells in Orthopaedics
Autologous Mesenchymal Stem Cells in OrthopaedicsVladimir Bobic
 
Mammalian MSC from Selected Species: Features and Applications Christiane Ude...
Mammalian MSC from Selected Species: Features and Applications Christiane Ude...Mammalian MSC from Selected Species: Features and Applications Christiane Ude...
Mammalian MSC from Selected Species: Features and Applications Christiane Ude...Lipogems Equine & Lipogems Canine
 
Journal of stem cells research
Journal of stem cells researchJournal of stem cells research
Journal of stem cells researchScidoc Publishers
 
Collagen II Microbeads Final
Collagen II Microbeads FinalCollagen II Microbeads Final
Collagen II Microbeads FinalDavid R. Mertz
 
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...ComprehensiveBiologi
 
Bone tissue engineering
Bone tissue engineeringBone tissue engineering
Bone tissue engineeringMehdi Chamani
 
Adult mesenchymal stem cells /dental courses
Adult mesenchymal stem cells  /dental coursesAdult mesenchymal stem cells  /dental courses
Adult mesenchymal stem cells /dental coursesIndian dental academy
 
De novo kidney regeneration with stem cells
De novo kidney regeneration with stem cellsDe novo kidney regeneration with stem cells
De novo kidney regeneration with stem cellsSheila Palacios
 
NSF Proposal Project Discription
NSF Proposal Project DiscriptionNSF Proposal Project Discription
NSF Proposal Project Discriptionguestc121aae
 

Similaire à 1 s2.0-s1742706114003110-main(4) (20)

Mesenchymal stem cells in Orthopaedics
Mesenchymal stem cells in OrthopaedicsMesenchymal stem cells in Orthopaedics
Mesenchymal stem cells in Orthopaedics
 
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
 
Stem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder DysfunctionStem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder Dysfunction
 
Stem cells for Bladder Dysfunction
Stem cells for Bladder DysfunctionStem cells for Bladder Dysfunction
Stem cells for Bladder Dysfunction
 
International Journal of Stem Cells & Research
International Journal of Stem Cells & ResearchInternational Journal of Stem Cells & Research
International Journal of Stem Cells & Research
 
Autologous Mesenchymal Stem Cells in Orthopaedics
Autologous Mesenchymal Stem Cells in OrthopaedicsAutologous Mesenchymal Stem Cells in Orthopaedics
Autologous Mesenchymal Stem Cells in Orthopaedics
 
Irmi unige
Irmi unigeIrmi unige
Irmi unige
 
Mammalian MSC from Selected Species: Features and Applications Christiane Ude...
Mammalian MSC from Selected Species: Features and Applications Christiane Ude...Mammalian MSC from Selected Species: Features and Applications Christiane Ude...
Mammalian MSC from Selected Species: Features and Applications Christiane Ude...
 
Regenerative endodontics
Regenerative endodonticsRegenerative endodontics
Regenerative endodontics
 
Transplantation journal
Transplantation journalTransplantation journal
Transplantation journal
 
Stem cells journal
Stem cells  journalStem cells  journal
Stem cells journal
 
Journal of stem cells research
Journal of stem cells researchJournal of stem cells research
Journal of stem cells research
 
Collagen II Microbeads Final
Collagen II Microbeads FinalCollagen II Microbeads Final
Collagen II Microbeads Final
 
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
 
Application of Nanoscaffolds in Mesenchymal
Application of Nanoscaffolds in MesenchymalApplication of Nanoscaffolds in Mesenchymal
Application of Nanoscaffolds in Mesenchymal
 
Poster SVF
Poster SVFPoster SVF
Poster SVF
 
Bone tissue engineering
Bone tissue engineeringBone tissue engineering
Bone tissue engineering
 
Adult mesenchymal stem cells /dental courses
Adult mesenchymal stem cells  /dental coursesAdult mesenchymal stem cells  /dental courses
Adult mesenchymal stem cells /dental courses
 
De novo kidney regeneration with stem cells
De novo kidney regeneration with stem cellsDe novo kidney regeneration with stem cells
De novo kidney regeneration with stem cells
 
NSF Proposal Project Discription
NSF Proposal Project DiscriptionNSF Proposal Project Discription
NSF Proposal Project Discription
 

Dernier

Kottayam Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Kottayam Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real MeetKottayam Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Kottayam Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real MeetCall Girls Chandigarh
 
Gorgeous Call Girls Mohali {7435815124} ❤️VVIP ANGEL Call Girls in Mohali Punjab
Gorgeous Call Girls Mohali {7435815124} ❤️VVIP ANGEL Call Girls in Mohali PunjabGorgeous Call Girls Mohali {7435815124} ❤️VVIP ANGEL Call Girls in Mohali Punjab
Gorgeous Call Girls Mohali {7435815124} ❤️VVIP ANGEL Call Girls in Mohali PunjabSheetaleventcompany
 
(Deeksha) 💓 9920725232 💓High Profile Call Girls Navi Mumbai You Can Get The S...
(Deeksha) 💓 9920725232 💓High Profile Call Girls Navi Mumbai You Can Get The S...(Deeksha) 💓 9920725232 💓High Profile Call Girls Navi Mumbai You Can Get The S...
(Deeksha) 💓 9920725232 💓High Profile Call Girls Navi Mumbai You Can Get The S...Ahmedabad Call Girls
 
Sexy Call Girl Villupuram Arshi 💚9058824046💚 Villupuram Escort Service
Sexy Call Girl Villupuram Arshi 💚9058824046💚 Villupuram Escort ServiceSexy Call Girl Villupuram Arshi 💚9058824046💚 Villupuram Escort Service
Sexy Call Girl Villupuram Arshi 💚9058824046💚 Villupuram Escort Servicejaanseema653
 
Independent Call Girls Service Chandigarh Sector 17 | 8868886958 | Call Girl ...
Independent Call Girls Service Chandigarh Sector 17 | 8868886958 | Call Girl ...Independent Call Girls Service Chandigarh Sector 17 | 8868886958 | Call Girl ...
Independent Call Girls Service Chandigarh Sector 17 | 8868886958 | Call Girl ...Sheetaleventcompany
 
Call Girls In Indore 📞9235973566📞Just Call Inaaya📲 Call Girls Service In Indo...
Call Girls In Indore 📞9235973566📞Just Call Inaaya📲 Call Girls Service In Indo...Call Girls In Indore 📞9235973566📞Just Call Inaaya📲 Call Girls Service In Indo...
Call Girls In Indore 📞9235973566📞Just Call Inaaya📲 Call Girls Service In Indo...Sheetaleventcompany
 
Gorgeous Call Girls In Pune {9xx000xx09} ❤️VVIP ANKITA Call Girl in Pune Maha...
Gorgeous Call Girls In Pune {9xx000xx09} ❤️VVIP ANKITA Call Girl in Pune Maha...Gorgeous Call Girls In Pune {9xx000xx09} ❤️VVIP ANKITA Call Girl in Pune Maha...
Gorgeous Call Girls In Pune {9xx000xx09} ❤️VVIP ANKITA Call Girl in Pune Maha...Sheetaleventcompany
 
Rishikesh Call Girls Service 6398383382 Real Russian Girls Looking Models
Rishikesh Call Girls Service 6398383382 Real Russian Girls Looking ModelsRishikesh Call Girls Service 6398383382 Real Russian Girls Looking Models
Rishikesh Call Girls Service 6398383382 Real Russian Girls Looking ModelsRupali Sharma
 
👉Bangalore Call Girl Service👉📞 7304373326 👉📞 Just📲 Call Rajveer Call Girls Se...
👉Bangalore Call Girl Service👉📞 7304373326 👉📞 Just📲 Call Rajveer Call Girls Se...👉Bangalore Call Girl Service👉📞 7304373326 👉📞 Just📲 Call Rajveer Call Girls Se...
👉Bangalore Call Girl Service👉📞 7304373326 👉📞 Just📲 Call Rajveer Call Girls Se...Sheetaleventcompany
 
Sexy Call Girl Tiruvannamalai Arshi 💚9058824046💚 Tiruvannamalai Escort Service
Sexy Call Girl Tiruvannamalai Arshi 💚9058824046💚 Tiruvannamalai Escort ServiceSexy Call Girl Tiruvannamalai Arshi 💚9058824046💚 Tiruvannamalai Escort Service
Sexy Call Girl Tiruvannamalai Arshi 💚9058824046💚 Tiruvannamalai Escort Servicejaanseema653
 
Ludhiana Call Girls Service Just Call 6367187148 Top Class Call Girl Service ...
Ludhiana Call Girls Service Just Call 6367187148 Top Class Call Girl Service ...Ludhiana Call Girls Service Just Call 6367187148 Top Class Call Girl Service ...
Ludhiana Call Girls Service Just Call 6367187148 Top Class Call Girl Service ...Escorts In Kolkata
 
Kochi call girls Mallu escort girls available 7877702510
Kochi call girls Mallu escort girls available 7877702510Kochi call girls Mallu escort girls available 7877702510
Kochi call girls Mallu escort girls available 7877702510Vipesco
 
Sexy Call Girl Dharmapuri Arshi 💚9058824046💚 Dharmapuri Escort Service
Sexy Call Girl Dharmapuri Arshi 💚9058824046💚 Dharmapuri Escort ServiceSexy Call Girl Dharmapuri Arshi 💚9058824046💚 Dharmapuri Escort Service
Sexy Call Girl Dharmapuri Arshi 💚9058824046💚 Dharmapuri Escort Servicejaanseema653
 
Kolkata Call Girls Miss Inaaya ❤️ at @30% discount Everyday Call girl
Kolkata Call Girls Miss Inaaya ❤️ at @30% discount Everyday Call girlKolkata Call Girls Miss Inaaya ❤️ at @30% discount Everyday Call girl
Kolkata Call Girls Miss Inaaya ❤️ at @30% discount Everyday Call girlonly4webmaster01
 
Best Lahore Escorts 😮‍💨03250114445 || VIP escorts in Lahore
Best Lahore Escorts 😮‍💨03250114445 || VIP escorts in LahoreBest Lahore Escorts 😮‍💨03250114445 || VIP escorts in Lahore
Best Lahore Escorts 😮‍💨03250114445 || VIP escorts in LahoreDeny Daniel
 
Call Girls Service Mohali {7435815124} ❤️VVIP PALAK Call Girl in Mohali Punjab
Call Girls Service Mohali {7435815124} ❤️VVIP PALAK Call Girl in Mohali PunjabCall Girls Service Mohali {7435815124} ❤️VVIP PALAK Call Girl in Mohali Punjab
Call Girls Service Mohali {7435815124} ❤️VVIP PALAK Call Girl in Mohali PunjabSheetaleventcompany
 
Top 20 Famous Indian Female Pornstars Name List 2024
Top 20 Famous Indian Female Pornstars Name List 2024Top 20 Famous Indian Female Pornstars Name List 2024
Top 20 Famous Indian Female Pornstars Name List 2024Sheetaleventcompany
 
Sexy Call Girl Nagercoil Arshi 💚9058824046💚 Nagercoil Escort Service
Sexy Call Girl Nagercoil Arshi 💚9058824046💚 Nagercoil Escort ServiceSexy Call Girl Nagercoil Arshi 💚9058824046💚 Nagercoil Escort Service
Sexy Call Girl Nagercoil Arshi 💚9058824046💚 Nagercoil Escort Servicejaanseema653
 
Call Girl in Bangalore 9632137771 {LowPrice} ❤️ (Navya) Bangalore Call Girls ...
Call Girl in Bangalore 9632137771 {LowPrice} ❤️ (Navya) Bangalore Call Girls ...Call Girl in Bangalore 9632137771 {LowPrice} ❤️ (Navya) Bangalore Call Girls ...
Call Girl in Bangalore 9632137771 {LowPrice} ❤️ (Navya) Bangalore Call Girls ...mahaiklolahd
 

Dernier (20)

Kottayam Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Kottayam Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real MeetKottayam Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
Kottayam Call Girls 👙 6297143586 👙 Genuine WhatsApp Number for Real Meet
 
Gorgeous Call Girls Mohali {7435815124} ❤️VVIP ANGEL Call Girls in Mohali Punjab
Gorgeous Call Girls Mohali {7435815124} ❤️VVIP ANGEL Call Girls in Mohali PunjabGorgeous Call Girls Mohali {7435815124} ❤️VVIP ANGEL Call Girls in Mohali Punjab
Gorgeous Call Girls Mohali {7435815124} ❤️VVIP ANGEL Call Girls in Mohali Punjab
 
(Deeksha) 💓 9920725232 💓High Profile Call Girls Navi Mumbai You Can Get The S...
(Deeksha) 💓 9920725232 💓High Profile Call Girls Navi Mumbai You Can Get The S...(Deeksha) 💓 9920725232 💓High Profile Call Girls Navi Mumbai You Can Get The S...
(Deeksha) 💓 9920725232 💓High Profile Call Girls Navi Mumbai You Can Get The S...
 
Sexy Call Girl Villupuram Arshi 💚9058824046💚 Villupuram Escort Service
Sexy Call Girl Villupuram Arshi 💚9058824046💚 Villupuram Escort ServiceSexy Call Girl Villupuram Arshi 💚9058824046💚 Villupuram Escort Service
Sexy Call Girl Villupuram Arshi 💚9058824046💚 Villupuram Escort Service
 
Independent Call Girls Service Chandigarh Sector 17 | 8868886958 | Call Girl ...
Independent Call Girls Service Chandigarh Sector 17 | 8868886958 | Call Girl ...Independent Call Girls Service Chandigarh Sector 17 | 8868886958 | Call Girl ...
Independent Call Girls Service Chandigarh Sector 17 | 8868886958 | Call Girl ...
 
Call Girls In Indore 📞9235973566📞Just Call Inaaya📲 Call Girls Service In Indo...
Call Girls In Indore 📞9235973566📞Just Call Inaaya📲 Call Girls Service In Indo...Call Girls In Indore 📞9235973566📞Just Call Inaaya📲 Call Girls Service In Indo...
Call Girls In Indore 📞9235973566📞Just Call Inaaya📲 Call Girls Service In Indo...
 
Gorgeous Call Girls In Pune {9xx000xx09} ❤️VVIP ANKITA Call Girl in Pune Maha...
Gorgeous Call Girls In Pune {9xx000xx09} ❤️VVIP ANKITA Call Girl in Pune Maha...Gorgeous Call Girls In Pune {9xx000xx09} ❤️VVIP ANKITA Call Girl in Pune Maha...
Gorgeous Call Girls In Pune {9xx000xx09} ❤️VVIP ANKITA Call Girl in Pune Maha...
 
Rishikesh Call Girls Service 6398383382 Real Russian Girls Looking Models
Rishikesh Call Girls Service 6398383382 Real Russian Girls Looking ModelsRishikesh Call Girls Service 6398383382 Real Russian Girls Looking Models
Rishikesh Call Girls Service 6398383382 Real Russian Girls Looking Models
 
👉Bangalore Call Girl Service👉📞 7304373326 👉📞 Just📲 Call Rajveer Call Girls Se...
👉Bangalore Call Girl Service👉📞 7304373326 👉📞 Just📲 Call Rajveer Call Girls Se...👉Bangalore Call Girl Service👉📞 7304373326 👉📞 Just📲 Call Rajveer Call Girls Se...
👉Bangalore Call Girl Service👉📞 7304373326 👉📞 Just📲 Call Rajveer Call Girls Se...
 
Sexy Call Girl Tiruvannamalai Arshi 💚9058824046💚 Tiruvannamalai Escort Service
Sexy Call Girl Tiruvannamalai Arshi 💚9058824046💚 Tiruvannamalai Escort ServiceSexy Call Girl Tiruvannamalai Arshi 💚9058824046💚 Tiruvannamalai Escort Service
Sexy Call Girl Tiruvannamalai Arshi 💚9058824046💚 Tiruvannamalai Escort Service
 
Ludhiana Call Girls Service Just Call 6367187148 Top Class Call Girl Service ...
Ludhiana Call Girls Service Just Call 6367187148 Top Class Call Girl Service ...Ludhiana Call Girls Service Just Call 6367187148 Top Class Call Girl Service ...
Ludhiana Call Girls Service Just Call 6367187148 Top Class Call Girl Service ...
 
Punjab Call Girls Contact Number +919053,900,678 Punjab Call Girls
Punjab Call Girls Contact Number +919053,900,678 Punjab Call GirlsPunjab Call Girls Contact Number +919053,900,678 Punjab Call Girls
Punjab Call Girls Contact Number +919053,900,678 Punjab Call Girls
 
Kochi call girls Mallu escort girls available 7877702510
Kochi call girls Mallu escort girls available 7877702510Kochi call girls Mallu escort girls available 7877702510
Kochi call girls Mallu escort girls available 7877702510
 
Sexy Call Girl Dharmapuri Arshi 💚9058824046💚 Dharmapuri Escort Service
Sexy Call Girl Dharmapuri Arshi 💚9058824046💚 Dharmapuri Escort ServiceSexy Call Girl Dharmapuri Arshi 💚9058824046💚 Dharmapuri Escort Service
Sexy Call Girl Dharmapuri Arshi 💚9058824046💚 Dharmapuri Escort Service
 
Kolkata Call Girls Miss Inaaya ❤️ at @30% discount Everyday Call girl
Kolkata Call Girls Miss Inaaya ❤️ at @30% discount Everyday Call girlKolkata Call Girls Miss Inaaya ❤️ at @30% discount Everyday Call girl
Kolkata Call Girls Miss Inaaya ❤️ at @30% discount Everyday Call girl
 
Best Lahore Escorts 😮‍💨03250114445 || VIP escorts in Lahore
Best Lahore Escorts 😮‍💨03250114445 || VIP escorts in LahoreBest Lahore Escorts 😮‍💨03250114445 || VIP escorts in Lahore
Best Lahore Escorts 😮‍💨03250114445 || VIP escorts in Lahore
 
Call Girls Service Mohali {7435815124} ❤️VVIP PALAK Call Girl in Mohali Punjab
Call Girls Service Mohali {7435815124} ❤️VVIP PALAK Call Girl in Mohali PunjabCall Girls Service Mohali {7435815124} ❤️VVIP PALAK Call Girl in Mohali Punjab
Call Girls Service Mohali {7435815124} ❤️VVIP PALAK Call Girl in Mohali Punjab
 
Top 20 Famous Indian Female Pornstars Name List 2024
Top 20 Famous Indian Female Pornstars Name List 2024Top 20 Famous Indian Female Pornstars Name List 2024
Top 20 Famous Indian Female Pornstars Name List 2024
 
Sexy Call Girl Nagercoil Arshi 💚9058824046💚 Nagercoil Escort Service
Sexy Call Girl Nagercoil Arshi 💚9058824046💚 Nagercoil Escort ServiceSexy Call Girl Nagercoil Arshi 💚9058824046💚 Nagercoil Escort Service
Sexy Call Girl Nagercoil Arshi 💚9058824046💚 Nagercoil Escort Service
 
Call Girl in Bangalore 9632137771 {LowPrice} ❤️ (Navya) Bangalore Call Girls ...
Call Girl in Bangalore 9632137771 {LowPrice} ❤️ (Navya) Bangalore Call Girls ...Call Girl in Bangalore 9632137771 {LowPrice} ❤️ (Navya) Bangalore Call Girls ...
Call Girl in Bangalore 9632137771 {LowPrice} ❤️ (Navya) Bangalore Call Girls ...
 

1 s2.0-s1742706114003110-main(4)

  • 1. Acta Biomaterialia 10 (2014) 4730–4741 Contents lists available at ScienceDirect Acta Biomaterialia journal homepage: www.elsevier.com/locate/actabiomat Mesenchymal stroma cells trigger early attraction of M1 macrophages and endothelial cells into fibrin hydrogels, stimulating long bone healing without long-term engraftment Elisabeth Seebach, Holger Freischmidt, Jeannine Holschbach 1, Jörg Fellenberg, Wiltrud Richter ⇑ Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany a r t i c l e i n f o Article history: Received 22 May 2014 Received in revised form 10 July 2014 Accepted 14 July 2014 Available online 22 July 2014 Keywords: Bone healing Mesenchymal stroma cells Fibrin hydrogel Host cell recruitment Macrophage a b s t r a c t Implantation of mesenchymal stroma cells (MSCs) is an attractive approach to stimulate closure of large bone defects but an optimal carrier has yet to be defined. MSCs may display trophic and/or immunomod-ulatory features or stimulate bone healing by their osteogenic activity. The aim of this study was to unra-vel whether fibrin hydrogel supports early actions of implanted MSCs, such as host cell recruitment, immunomodulation and tissue regeneration, in long bone defects. Female rats received cell-free fibrin or male MSCs embedded in a fibrin carrier into plate-stabilized femoral bone defects. Removed callus was analyzed for host cell invasion (day 6), local cytokine expression (days 3 and 6) and persistence of male MSCs (days 3, 6, 14 and 28). Fibrin–MSC composites triggered fast attraction of host cells into the hydrogel while cell-free fibrin implants were not invaded. A migration front dominated by M1 macrophages and endothelial progenitor cells formed while M2 macrophages remained sparse. Only MSC-seeded fibrin hydrogel stimulated early tissue maturation and primitive vessel formation at day 6 in line with significantly higher VEGF mRNA levels recorded at day 3. Local TNF-a, IL-1b and IL-10 expres-sion indicated a balanced immune cell activity independent of MSC implantation. Implanted MSCs persisted until day 14 but not day 28. Our results demonstrate that fibrin hydrogel is an attractive carrier for MSC implantation into long bone defects, supporting host cell attraction and pro-angiogenic activity. By this angiogenesis, implant integration and tissue maturation was stimulated in long bone healing independent of long-term engraftment of implanted MSCs. 2014 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved. 1. Introduction The closure of large bone defects secondary to trauma or tumor resection remains a challenge in orthopedic surgery. One promis-ing approach to support natural bone healing is the implantation of bone marrow-derived mesenchymal stroma cells (MSCs) in a suitable carrier [1]. So far, sponge-like scaffolds have mainly been used for cell implantation although these have disadvantages, such as limited cell seeding, unequal cell distribution and non-homoge-neous matrix deposition. Hydrogels are attractive vehicles for cell transplantation because of their easy adaptation to any shape of defect, homogeneous cell distribution, even extracellular matrix deposition and beneficial interaction with the host tissue [2–4]. High biocompatibility, biodegradation and the adhesiveness of fibrin glue [5] have rendered fibrin hydrogel a common carrier for autologous cell transplantation in cartilage repair [6,7]. Surpris-ingly, fibrin has so far attracted little attention as a carrier for MSC-based treatment of bone defects and little is known as to whether fibrin hydrogels support early actions of MSCs in stimulating long bone healing. MSCs can easily be isolated from bone marrow [8], are characterized by fast proliferation in culture, and express CD90, CD105 and vascular cell adhesion molecule 1 (VCAM1, CD106) on their surface in the absence of CD34 and CD45 [9,10]. After expansion culture, MSCs can be induced to differentiate into chondrogenic, adipogenic and osteogenic lineages in vitro [11]. Moreover, MSCs provide a supportive cellular microenvironment by secreting a variety of molecules including growth factors and cytokines which potentially mediate host cell recruitment, angio-genesis and immune modulation [12,13]. MSCs further possess a significant osteogenic potential in vivo and have successfully been used to induce repair of bone defects in animals [14,15] and in humans [16,17]. ⇑ Corresponding author. Tel.: +49 (0) 6221 5629254; fax: +49 (0) 6221 5629288. E-mail address: wiltrud.richter@med.uni-heidelberg.de (W. Richter). 1 Current address: Merck KGaA, Frankfurter Strasse 250, 64293 Darmstadt, Germany. http://dx.doi.org/10.1016/j.actbio.2014.07.017 1742-7061/ 2014 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
  • 2. E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 4731 According to the concept of stem cell-based bone regeneration, implanted MSCs contribute physically to the repair tissue by gen-erating new bone tissue, as has been reported in ectopic implanta-tion models in immune-deficient mice in which fibrin hydrogel formed part of the carrier [18,19]. The role and fate of MSCs implanted into a bone defect is, however, unclear and long-term persistence, especially in immune competent animals, has been discussed [20–22]. Fast attraction of a large number of diverse host cells to the defect site, acceleration of new vessel formation and modulation of the early inflammatory reaction may be important trophic roles of MSCs in stimulation of bone healing which should be supported by the carrier used for implantation. In an orthotopic setting, these mechanisms are potentially more important for improved bone regeneration than enhancing the number of local bone-forming cells by implantation of additional osteogenic pro-genitors. Whether fibrin hydrogel is an attractive carrier for MSC implantation in this respect remains to be determined. A highly potent MSC population has to be produced in a stan-dardized fashion from each donor to allow optimal healing results in the patient. In view of the changing paradigm of MSC function [23], the therapeutically most potent MSC subpopulations may, however, be characterized by pronounced trophic and/or immuno-modulatory features displayed in the early callus instead of a high osteogenic differentiation capacity. Thus, it is essential to better understand relevant actions of MSC composites in a bone defect in order to optimize MSC-based bone tissue engineering approaches and to design an optimal application strategy for clin-ical regeneration of large bone defects. The aim of this study was to assess whether fibrin hydrogel is an attractive carrier for MSC implantation supporting early bone regeneration with regard to host cell attraction, angiogenesis, immunomodulation and physical contribution of implanted MSCs to newly formed bone tissue. In order to follow up implanted MSCs without extra manipulation by a labelling reaction, our approach was to implant sex-mismatched MSCs in a fibrin hydrogel into a long bone defect in rats. Callus was removed from female recipi-ents at distinct time points after surgery and analyzed for host cell recruitment, the presence of inflammatory cytokines and the per-sistence of male MSCs to pinpoint the dominant actions of fibrin–MSC composites in early bone regeneration. 2. Materials and methods 2.1. Animals and study design Sprague–Dawley rats (n = 46; 17–23 weeks of age, Charles River, Sulzfeld, Germany) were included in the animal study. The hind limbs in the growing rat are skeletally mature at around 15 weeks of age [24]. The experimental protocol was approved by the local animal experimental ethics committee and all proce-dures were performed according to the European Laboratory Ani-mal Science Guidelines. Male rats were used for MSC donation and female rats served as MSC recipients. A total of 43 animals received a non-critical 2 mm bone defect in the right femur. Two experimental groups were defined. Rats of group 1 received an empty fibrin clot and served as controls (n = 20), while rats of group 2 were treated with 1 106 MSCs in a fibrin clot (n = 23). Animals were killed 3, 6, 14 or 28 days after surgery. Histological evaluation of callus was performed at day 6 (five animals per group). Gene expression analysis and detection of the sex-deter-mining region of Y-Gen (SRY)-specific DNA were assessed in the same sample at days 3 and 6 from the callus of five animals per condition. Visualization of new bone formation by micro-com-puted tomography (lCT) and SRY-DNA detection was done on the same animals with n = 3 animals for the cell-free and n = 4 animals for the MSC-treated group at day 14, and with n = 2 animals for the cell-free and n = 4 animals for the MSC-treated group at day 28. 2.2. MSC isolation and expansion MSCs were isolated from bone marrow of three male rats using a modified centrifugation method [25]. Animals were killed by asphyxiation with CO2 and MSCs were extracted from tibiae and femora by centrifugation at 750g for 2 min. All cells were pooled, washed with phosphate-buffered saline (PBS) and seeded into 0.1% gelatin-coated flasks in a high-glucose Dulbecco’s modified Eagle’s medium (DMEM) containing 12.5% fetal calf serum (FCS), 2mM L-glutamine, 1% non-essential amino acids, 0.1% 2-mercap-toethanol (Invitrogen, Karlsruhe, Germany), 100 U ml–1 penicillin, 100 lgml–1 streptomycin (Biochrom, Berlin, Germany) and 4 ngml–1 recombinant human fibroblast growth factor-2 (Active Bioscience, Hamburg, Germany) at a density of 1.25 105 cells cm–2. After 5 days under standard culturing conditions (37 C and 6% CO2) non-adherent cells were removed by washing. Medium was changed every 2 days and cells were split at 80% confluency and replated at a density of 4–6 103 cells cm–2. All cells were frozen in passage 1 in liquid nitrogen and recultivated at a density of 6 103 cells cm–2 3 days before implantation. 2.3. Flow cytometry At the time of implantation cells were harvested with trypsin and 1 105 cells were labelled with anti-CD45 (1:250), anti- CD68 (1:100), anti-CD90-FITC (1:25; Abcam, Cambridge, UK) and anti-CD106-PE (1:50; BD, Heidelberg, Germany) antibodies, respectively. For CD45 and CD68 staining cells were incubated with a secondary anti-mouse FITC-conjugated antibody (1:100; Jackson ImmunoResearch Laboratories, Suffolk, UK). Surface mar-ker positive cells were quantified in a FACSCalibur (BD, Heidelberg, Germany). 2.4. Osteogenic, adipogenic and chondrogenic in vitro differentiation For osteogenesis, quadruplicates of 3.5 104 MSCs were seeded per well of a 24-well plate in osteogenic medium consisting of high-glucose DMEM supplemented with 10% FCS, 0.1 lM dexamethasone, 0.17 mM ascorbic acid 2-phosphate, 10 mM b-glycero-phosphate (Sigma-Aldrich, Taufkirchen, Germany), 100 U ml–1 penicillin and 100 lgml–1 streptomycin. At days 1, 7 and 14 the alkaline phosphatase (ALP) enzyme activity of cell lysate was measured as described previously [18]. For adipogenic differentiation cells were seeded in duplicates at 3.5 104 cells per well of a 24-well plate in high-glucose DMEM supplemented with 10% FCS, 0.01 lM dexamethasone, 5 lgml–1 insulin (Sanofi- Aventis, Frankfurt, Germany), 100 U ml–1 penicillin and 100 lgml–1 streptomycin [26]. Formation of lipid droplets was evaluated by Oil Red O staining (Chroma, Münster, Germany) after fixing the cells in 4% paraformaldehyde (PFA, Merck, Darmstadt, Germany). Chondrogenesis was performed with 5 105 cells in a 3D pellet-culture in high-glucose DMEM supplemented with 5 lgml–1 transferrin, 5 ng ml–1 sodium selenite, 0.1 lMdexameth-asone, 0.17 mM ascorbic acid 2-phosphate, 1 mM sodium pyru-vate, 0.35 mM proline, 1.25 mg ml–1 BSA (Sigma-Aldrich, Taufkirchen, Germany), 100 U ml–1 penicillin, 100 lg ml–1 strepto-mycin, 5 lgml–1 insulin and 10 ng ml–1 transforming growth fac-tor- beta 3 (TGFb-3; Miltenyi Biotec GmbH, Bergisch Gladbach, Germany). Differentiation was evaluated on day 14 by Safranin O staining (Chroma, Münster, Germany) for proteoglycan deposition and collagen type II immunohistochemistry as described previ-ously [27].
  • 3. 4732 E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 2.5. Preparation of fibrin-MSC composites for implantation MSCs were prepared separately for every animal, to limit the time period between preparing the fibrinogen cell suspension and implantation to 20 min. The fibrinogen solution (Baxter, Wien, Austria) was diluted 1:2 and the thrombin (Baxter, Wien, Austria) was diluted 1:50 in PBS containing protease inhibitor cocktail (cOmplete Mini: Roche, Mannheim, Germany). 1 106 MSCs were resuspended in 40 ll of the fibrinogen solution, kept at room temperature until 40 ll of the thrombin solution was added. The solidifying gel was transferred into the bone defect. Fibrin clots for the control animals were prepared without cells. Five additional fibrin–MSC clots were frozen at 80 C and used for gene expression analysis of MSCs before implantation. Three additional fibrin(–MSC) clots were used for histological examina-tion of non-implanted samples. 2.6. Implantation of fibrin–MSC composites in the bone defect Rats were anaesthetized by intraperitoneal injection of ketamine hydrochloride (Ketamin: 100 mg kg–1 body weight, Orion Pharma, Hamburg, Germany) and medetomidine hydrochlo-ride (Domitor: 0.2 mg kg–1 body weight, Orion Pharma, Hamburg, Germany). A longitudinal incision (30 mm) of skin and fascia of the right hind leg was performed. The vastus lateralis and biceps femoris muscles were carefully separated and the anterolateral side of the femoral shaft was exposed. The attached muscles were stripped from the bone and a polyether ether ketone (PEEK) plate (Rat Fix, AO, Davos, Switzerland) was adjusted to the femur. The PEEK plate was held in position and pilot holes were drilled through both cortices along the predrilled holes of the plate. The plate was fixed to the bone by three screws (Rat Fix, AO, Davos, Switzerland) at each side. A 2 mm bone defect was created with a sawing wire (Rat Fix, AO, Davos, Switzerland). Bleeding was pre-vented by pressing a gelatin sponge (Gelita-Spon: Gelita Medical, Amsterdam, the Netherlands) into the bone defect for 2 min. The polymerizing fibrin clot was quickly transferred into the bone gap via a pipette and allowed to solidify for 3 min. Muscles, deep fascia, subcutaneous fascia and skin were closed in a standard fash-ion. Animals were treated once with 0.02 ml gentamicin sulfate (Refobacin: 80 mg, Merck, Darmstadt, Germany) for antibiosis and buprenorphine hydrochloride (Temgesic: 0.01 mg kg–1 body weight, RB Pharmaceuticals, UK) was given subcutaneously for analgesia. The animals were allowed full activity in their cages and were medicated with buprenorphine hydrochloride (Temge-sic: 0.01 mg kg–1 body weight) for the first two postoperative days. At days 3, 6, 14 or 28 after surgery animals were narcotized with CO2 and killed by exsanguination via heart puncture. Femora were dissected by opening the sutures and careful stripping off of the surrounding tissue. Callus was carefully isolated from bone defect and either directly frozen at 80 C for MSC detection and gene expression analysis or fixed in 4% PFA for histology. 2.7. lCT New bone formation of day 14 and 28 samples was evaluated using a Sky-Scan 1076 in vivo X-ray microtomograph (Skyscan, Antwerpen, Belgium). Femora were scanned with surrounding muscles wrapped in plastic foil using the 0.025 mm titan filter, with the following settings: voxel size 17.7 lm, voltage 65 kV, cur-rent 139 lA, exposure time 280 ms, frame averaging 3. Data were recorded every 0.4 rotation step through 180. Reconstruction was performed using NRecon software (version 1.6.3.2, Skyscan, Antwerp, Belgium). 3-D pictures were made with CTan/CTVol software (Skyscan, Antwerp, Belgium). 2.8. DNA isolation and SRY-gene-specific PCR Genomic DNA from day 3 and 6 callus was extracted along with total RNA according to the manufacturer’s protocol using the peq- GOLD TriFast technique (peqlab, Erlangen, Germany). Day 14 or 28 callus was digested in lysis buffer with 100 ng ml–1 proteinase K (Fermentas, St Leon-Rot, Germany) at 56 C overnight and DNA was extracted by ethanol precipitation. A SRY-gene-specific DNA fragment was amplified in a standard PCR using Taq DNA Polymer-ase (Invitrogen, Karlsruhe, Germany). For detection of implanted male MSCs in defined regions, day 6 callus was separated by laser microdissection, and DNA was isolated using the QIAmp DNA Micro Kit (Qiagen GmbH, Hilden, Germany) according to the man-ufacturer’s protocol. DNA analogously isolated from equivalent sections of the control samples was used as negative control. SRY-gene-specific DNA was amplified by nested PCR. PCR products were visualized on a 2% agarose gel and their specificity was con-firmed by DNA sequencing. 2.9. Gene expression analysis Callus was homogenized in peqGOLD TriFast and total RNA was extracted according to the manufacturer’s protocol (peqlab, Erlangen, Germany). cDNA synthesis was performed with 1 lg total RNA using Omniscript reverse transcriptase (0.2 U ll–1), oli-go( dT) primer (1 lM, both Qiagen GmbH, Hilden, Germany) and a ribonuclease inhibitor (RNaseOUT: 40 U ll–1, Invitrogen, Kar-lsruhe, Germany) according to the manufacturer’s instructions. Expression levels of individual genes were analyzed by quantita-tive PCR (Stratagene Mx3000P; Agilent Technologies, Böblingen, Germany). cDNA were amplified using gene-specific primer sets (Table 1) obtained from Eurofins (Ebersberg, Germany) and real-time fluorimetric intensity of SYBR green I (Thermo Scientific, Rockford, USA) was monitored. The apparent threshold cycles (Ct) of the genes of interest were compared to the Ct of the refer-ence gene hypoxanthine-guanine phosphoribosyltransferase-1 (HPRT-1) in the same cDNA sample. When indicated, the percent-age of the reference gene expression levels shown in the figures refer to relative levels as a percentage of HPRT-1. Melting curves and agarose gel electrophoresis of the PCR products were used for quality control. 2.10. Histology Callus was isolated from the bone defect and subjected to his-tology without proximal and distal bone ends to allow preparation of the samples without decalcification. In brief, samples were fixed in 4% PFA, dehydrated in a graded alcohol series and embedded in paraffin. In vivo orientation of the callus was maintained through-out the embedding process and callus was cut along this axis. For histological investigations, sections (5 lm) from the callus center were used and stained with haematoxylin and eosin (HE: Chroma, Münster, Germany) or Movat’s Pentachrome (all colours: Chroma, Münster, Germany) in a standard procedure. For CD31 and CD68 immunohistochemistry, digestion with 20 ng ml–1 proteinase K (Fermentas, St Leon-Rot, Germany) for 20 min at 37 C was used for antigen retrieval. Sections for CD163 staining were pre-treated with 1 mg ml–1 pronase (Roche, Mannheim, Germany) for 30 min at 37 C. No antigen retrieval was required for CCR7 staining. Unspecific binding was blocked for 30 min with 5% BSA for CD31, CD68 and CD163, and with 5% human serum for CCR7, and then sections were incubated with the primary antibodies CD31 (1:200; Abbiotec, San Diego, CA, USA), CD68 (1:100; Abcam, Cambridge, UK), CD163 (1:100, AbD Serotec, Oxford, UK) and CCR7 (1:250; Epitomics, Burlingame, CA, USA) overnight at 4 C. After washing sections were incubated with
  • 4. E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 4733 the respective biotinylated secondary antibodies (CD31 and CCR7: anti-rabbit IgG; CD68 and CD163: anti-mouse IgG; 1:500; both Dianova, Hamburg, Germany), followed by ALP–streptavidin (Vec-tastain; Vector Laboratories, Burlingame, CA, USA) and Fast Red (Roche, Mannheim, Germany). The nuclei were counterstained with haematoxylin (Chroma) and slides were covered in Aquatex (Merck, Darmstadt, Germany). Negative controls were performed by omitting the respective primary antibody. Histomorphometric evaluation was performed on HE-stained sections of two zones of every callus respectively (n = 10 per group). Total callus area was marked black and the area infiltrated by host cells was labelled grey using Photoshop 7.0 (Adobe Systems Inc., USA). ImageJ 1.44p (Wayne Rasband, National Institutes of Health, USA) was used to assess the total area and the infiltrated area. Cell invasion is stated as the percentage of infiltrated area related to total area. 2.11. Laser microdissection To evaluate persistence of implanted MSCs next to invading host cells we searched for possible SRY-signals in the infiltrated area of the callus vs. the non-infiltrated part of the MSC implant. Appropriate parts of the tissue section were separated by laser microdissection. In brief, sections (10 lm) of the MSC-containing callus were taken up on manually foil-coated slides. After HE staining, the area infiltrated by host cells was separated from the remaining implant using a PALM MicroBeam Laser Microdissection System (Carl Zeiss AG, Jena, Germany). To obtain enough material for nested PCR, tissue collected from the infiltrated area (two sec-tions per sample from five animals, n = 10) or of the remaining fibrin clot were pooled, respectively (n = 10) and subjected to DNA extraction. Analogously, tissue collected from the animals receiving cell-free fibrin (n = 10) were pooled and further used as female control. 2.12. Statistical analysis For comparison of gene expression between the MSC groups and control groups of days 3 and 6 a Kruskal–Wallis test with post-hoc Mann–Whitney U-tests was conducted. Only if the Krus-kal– Wallis tests indicated significance were data analyzed post hoc and corrected with the Bonferroni test. The Mann–Whitney U-test was furthermore applied for histomorphometric evaluation of the MSC group and the control group at day 6 and for comparison of genomic DNA content between the MSC group and the control group at day 14. A two-tailed significance value of P 0.05 was considered statistically significant. Data analysis was performed with SPSS for Windows 16.0 (SPSS Inc., Chicago, IL, USA). 3. Results 3.1. Enhanced attraction of host cells into fibrin–MSC composites One pool of MSCs from long bones of three male rats was pro-duced and extensively characterized before application in all experiments. Cells were strongly positive for the rat surface mark-ers CD90 (97%) and CD106 (35%) while almost negative for CD45 (1.8%) and CD68 (1.7%) as expected for MSC populations (Fig. 1A). During in vitro osteogenesis, cells showed a strong up-regulation of the osteogenic markers bone sialoprotein (BSP) and osteocalcin (OC) and an increasing ALP enzyme activity (Fig. 1B). During adipogenic induction cells up-regulated mRNA levels of the adipogenic marker peroxisome proliferator-activated recep-tor- gamma (PPAR-c and deposited lipid droplets as visualized by Oil Red O staining (Fig. 1C). Chondrogenic differentiation in high-density pellets resulted in deposition of a proteoglycan- and colla-gen type II-rich extracellular matrix (Fig. 1D). MSCs were seeded in fibrin hydrogel allowing a standardized implantation of a defined concentration of cells into the 2 mm plate-stabilized femur defect of female rats. The hydrogel was directly transferred after starting the polymerization and solidified within the defect, enabling a complete filling of the bone gap by the implant with intimate contact to the open bone endings (Fig. 2A). Six days after implantation cell-seeded and non-seeded fibrin implants were still present in the bone defect (Fig. 2B). The in vivo orientation was maintained for histology (Fig. 2C). As shown in Fig. 3, staining of parallel non-implanted fibrin–MSC composites showed a homogeneous distribution of the seeded cells within the fibrin hydrogel (Fig. 3H inset). Six days post-implanta-tion, overview staining of control samples revealed little infiltra-tion of host cells into the MSC-free implant but indicated some dissolution of the fibrin from the proximal side of the bone defect in two out of five samples (Fig. 3A). This appeared to create space for accumulation of some densely packed host cells in a callus aside from the fibrin (Fig. 3B), while the fibrin matrix itself remained almost empty (Fig. 3C, D). In contrast, large numbers of host cells infiltrated the implant in the MSC group, especially from the proximal bone ending (Fig. 3E). Interestingly, in three out of five samples, a dense migration front was formed consisting of packed cells which apparently moved through the defect from the proximal to the distal side (Fig. 3F, G). The majority of MSCs located in front of this border looked viable (Fig. 3H). Behind the migration front, the first signs of tissue maturation and extracellu-lar matrix deposition were observed (Fig. 3F) with evidence of some proteoglycan deposition in the three most advanced samples according to pentachrome staining (Fig. 3I). A comparable Table 1 List of oligonucleotides used for qRT-PCR analysis and SRY-gene-specific PCR. Gene GenBank no. Forward primer Reverse primer BSP [NM_012587.2] ACGCTGGAAAGTTGGAGTTAG GACCTGCTCATTTTCATCCA CD45 [NM_001109890.1] GCATGCATCAATCCTAGTCC GGCCATGATGTCATAGAGGA HPRT-1 [NM_012583.2] GCCAGACTTTGTTGGATTTG CACTTTCGCTGATGACACAA IL-1b [NM_031512.2] GACAAGCAACGACAAAATCC ACCGCTTTTCCATCTTCTTC IL-2 [NM_053836.1] AGCGTGTGTTGGATTTGACT TCTCCTCAGAAATTCCACCA IL-6 [NM_012589.2] AGCCAGAGTCATTCAGAGCA AGTTGGATGGTCTTGGTCCT IL-10 [NM_012854.2] GACGCTGTCATCGATTTCTC TTCATGGCCTTGTAGACACC MIP-2 [NM_053647.1] TGAAGTTTGTCTCAACCCTGA GGTGCAGTTCGTTTCTTTTCT OC [NM_013414.1] AGGGCAGTAAGGTGGTGAAT CTAAACGGTGGTGCCATAGA PPAR-c [NM_013124.3] ATAAAGTCCTTCCCGCTGAC ATCTCTTGCACAGCTTCCAC SRY [X89730.1] CTTTCGGAGCAGTGACAGTT CACTGATATCCCAGCTGCTT SRY nested [X89730.1] CTTTCGGAGCAGTGACAGTT CATGCTGGGATTCTGTTGA TNF-a [NM_012675.3] TCTACTGAACTTCGGGGTGA CCACCAGTTGGTTGTCTTTG VEGFa [NM_031836.3] CAATGATGAAGCCCTGGA CTATGCTGCAGGAAGCTCAT
  • 5. 4734 E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 Fig. 1. Characterization of rat MSCs. (A) Male rat MSCs pooled from three donors were expanded for two passages, labelled with the indicated antibodies and analyzed by flow cytometry. Representative histograms are shown with the stained population in black and the respective controls (no primary antibody) as grey lines. (B–D) MSCs were subjected to osteogenic (B), adipogenic (C) or chondrogenic conditions (D) for 2 weeks. (B) For osteogenesis, expression of bone sialoprotein (BSP) and osteocalcin (OC) was quantified by RT-qPCR and ALP-enzyme activity was measured (n = 2 experiments). Results are expressed as mean ± standard deviation. (C) Adipogenesis was confirmed by gene expression of peroxisome proliferator-activated receptor-gamma (PPAR-c) and Oil Red O staining of formed lipid droplets. (D) MSCs underwent chondrogenic differentiation according to deposition of proteoglycans (Safranin O staining, left side) and collagen type II (immunohistochemistry, right side). Staining is representative for three replicates. Scale bar: 100 lm. Fig. 2. Implantation of fibrin–MSC composites into long bone defects in rats and processing of callus for histology. A plate-stabilized 2 mm bone defect was created in the right femur of 42 female rats. Either 1 106 male MSCs embedded in a fibrin gel or empty fibrin gel (80 ll) were implanted into the defect. After 3, 6, 14 or 28 days femora were removed and early callus was lifted out from the defect for further analysis. (A) View directly after implantation of the fibrin gel with MSCs. (B) Early callus formation 6 days after MSC implantation. (C) For histology, day 6 callus was separated by a scalpel from the bone endings and lifted out of the bone defect. In vivo orientation was recorded throughout the embedding process and during histological evaluation.
  • 6. E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 4735 Fig. 3. Histological and histomorphometric evaluation of day 6 callus. (A–H) Paraffin sections of formalin-fixed tissue were stained with HE by a standard procedure. (A, E) Overview pictures of the callus at day 6 with the proximal explant side in the upper left and distal explant side in the lower right corner. Scale bar: 500 lm. Enlarged areas as defined in (A) and (E) are shown in (B)–(D) and (F)–(H). (B) Host-derived callus; (C) contact zone between host tissue and implant; (D) empty fibrin clot. Inset: HE-stained empty fibrin clot before implantation. (F) Highly infiltrated area from (E); (G) the migration front; (H) remaining MSC-seeded fibrin clot. Inset: HE stained MSC-seeded fibrin clot before implantation. Representative pictures of five replicates are shown in (A)–(H). Scale bar: 100 lm. (I) Movat’s Pentachrome staining: the infiltrated area of the three most mature samples of the MSC group showed deposition of glycosaminoglycan (light blue staining) next to fibrous tissue (red staining). Scale bar: 200 lm. (J) For histomorphometric evaluation, callus was divided into infiltrated area (dark grey) and unaffected implant (light grey). (K) Histomorphometric evaluation of the area infiltrated by host cells in relation to the total callus area (%, n = 10 sections per group, two per each of the five animals). Boxes represent the 25th and 75th percentile, median is given as horizontal line and whiskers are maximal and minimal values. Significant difference (P 0.01) between with and without MSCs is designated by two asterisks. maturation tissue was not seen in the fibrin control group. Histomorphometric assessment of the host cell-infiltrated vs. non-infiltrated areas (Fig. 3J) revealed a significantly larger invaded area in the fibrin–MSC composites compared to control samples (P = 0.01; Fig. 3K). 3.2. M1 macrophages and endothelial progenitor cells as main invaders Immunohistochemical staining revealed that the fibrin–MSC composites were mainly infiltrated by CD68- and CD31-positive cells indicative of macrophages and endothelial cells (Fig. 4). CD68-positive macrophages were present throughout the infil-trated area and were enriched in the migration front (Fig. 4B). Among ‘‘pioneer cells’’ migrating ahead of the more crowded zone and penetrating deep into the fibrin clot, many cells stained CD68 positive (Fig. 4B, lower right corner). In several areas, elongated CD31-positive cells apparently migrated from the proximal to the distal part of the implant forming primitive structures consistent with the formation of immature microvessels (Fig. 4D, black arrows). Macrophages can occur in a predominantly pro-inflammatory subtype M1 characterized by CCR7 expression and a predomi-nantly anti-inflammatory subtype M2 characterized by CD163 expression [28,29]. Staining for M1 and M2 macrophage markers at day 6 revealed that CCR7 signals were dominant in cell-dense areas. CCR7-positive cells also surrounded elongating structures, presumably areas of primitive vessel formation and tissue matura-tion (Fig. 4F, black arrows). In contrast, rather few CD163-positive cells were present in the host cell infiltrated area. Only in the sam-ple with the most advanced tissue regeneration had CD163- positive cells accumulated, especially in the cell-dense migration front (Fig. 4H). In the control group, the host tissue-derived callus contained CD31-, CD68- and/or CCR7-positive cells but hardly any cells were present in the fibrin hydrogel (Fig. 4A, C, E). CD163- positive (M2) cells, however, were not observed (Fig. 4G). 3.3. Cytokine expression in the early callus In order to analyze the trophic and immune modulatory effects of MSCs in the early callus, implants were harvested 3 and 6 days after surgery, respectively, to assess mRNA levels by quantitative PCR for a panel of pro- and anti-inflammatory mediators (Fig. 5). At the time of implantation, MSCs expressed vascular endothelial growth factor (VEGF), interleukin-6 (IL-6) and macrophage inflam-matory protein-2 (MIP-2, Fig. 5A). In vivo, median VEGF expression per cell increased from day 0 to day 3, reaching significantly higher levels in the MSC group vs. controls at day 3 (3.7-fold; P = 0.009) suggesting an enriched pro-angiogenic environment (Fig. 5D). IL-6 and MIP-2 were also expressed in vivo (Fig. 5E, F). Negligible
  • 7. 4736 E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 Fig. 4. Characterization of infiltrated host cells present in the callus 6 days after MSC implantation. (A, B) Macrophages were typed by immunohistochemistry for pan-marker CD68. (C, D) CD31 staining was performed to detect endothelial progenitor cells as indicators for neo-vascularization (black arrows: primitive vessel formation). (E, F) M1- typing of the infiltrated macrophages by CCR7 staining (black arrows: positive cells surrounding maturating tissue). (G, H) M2-typing of the infiltrated macrophages by CD163 staining. Positive cells are expressed with Fast Red and nuclei are counterstained with haematoxylin. Staining under omission of primary antibody served as negative controls (insets). Pictures are representative for five replicates. Scale bar: 100 lm. CD45 signals characterizing leukocytes were recorded in the fibrin–MSC composite before implantation (Fig. 5A). Day 3 and day 6 in vivo samples of both groups contained CD45 signals, con-firming the presence of leukocytes in the explants (Fig. 5B). As can be predicted from our histological findings, the relative signal for CD45 gene expression per cell (input RNA related to reference gene level) was significantly higher in the fibrin-only group compared to the MSC group (P = 0.016), which is in line with the notion that most host cells seen next to the implant in the control group were immune cells. In the MSC group, less CD45 expression per cell was observed since a large fraction of the mRNA was derived from the implanted (CD45-negative) MSCs. In line with the higher expres-sion levels of CD45, significantly higher mRNA levels for IL-1b tumor necrosis factor-alpha (TNF-a) and IL-10 (Fig. 5G–I) were obvious per cell in day 3 explants without MSCs vs. the group with MSCs, and a similar trend was seen for IL-2 (Fig. 5C) as well as for day 6 samples. In order to determine alterations in cytokine expression levels per immune cell in each sample, IL-1b, TNF-a and IL-10 expression levels were referred to the corresponding CD45 signal of the same sample. After adjustment per immune cell (Fig. 5J–L) no variation in the pro-inflammatory cytokines IL-1b (Fig. 5J) and TNF-a (Fig. 5K) or the anti-inflammatory cytokine IL-10 (Fig. 5L) was evi-dent between the controls and the MSC group at day 3 and day 6, indicating no major shift of the immune response by implanted MSCs. 3.4. No long-term engraftment of fibrin-implanted MSCs At day 14, healing of the bone defect in the MSC group was more advanced at the proximal bone ending compared to the distal side (Fig. 6A). The amount of genomic DNA in the callus increased considerably until day 28 in line with an increasing cell content (Fig. 6B). The presence of significantly more DNA in the MSC group
  • 8. E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 4737 Fig. 5. Characterization of expression of specific immunomediators in day 3 and day 6 callus. RNA was isolated from (A) fibrin–MSC composites before implantation and (B– M) early callus with and without MSCs at day 3 and day 6. Relative mRNA levels were detected by RT-qPCR and expression was standardized to the signals of the reference gene HPRT-1. (A) Basal gene expression of rat CD45, VEGF, IL-6 and MIP-2 by the MSC-population (day 0) with n = 5 replicates. (B–I) Relative mRNA levels per cell of rat CD45, IL-2, VEGF, IL-6, MIP-2, IL-1b, TNF-a and IL-10 in the MSC-seeded fibrin clots before implantation (day 0) and 3 or 6 days after implantation (n = 5 per time-point and group). (J–L) Relative expression of IL-1b, TNF-a and IL-10 per CD45-positive immune cell. Boxes represent the 25th and 75th percentile, median is given as horizontal line and whiskers are maximal and minimal values. Outliers (1.5- to 3-fold interquartile range, IQR) are depicted as circles, and extreme values (3-fold IQR) as diamonds. Significant differences (P 0.05) between groups are designated by an asterisk. at day 14 is in line with an enhanced host cell attraction into MSC-containing fibrin implants and/or may indicate more cell prolifera-tion in the MSC-treated defects. This difference was no longer seen at day 28 (Fig. 6B). When DNA from day 3, 6, 14 or 28 callus was analyzed for the persistence of implanted male MSCs, SRY-gene specific signals were detected in all MSC-containing explants from postoperative days 3, 6 and 14, whereas no signals were detected in the controls (Fig. 6C, D). Remarkably, at day 28, all explants were SRY-negative (Fig. 6E). Furthermore, no signals were obtained from the distal and proximal 2 mm bone endings (day 28), indicating no migration of male MSCs into the bone. Even an attempt to detect signals by a nested PCR approach with greatly enhanced sensitivity yielded no residual signals, suggesting a loss of male cells from the regeneration tissue and newly forming bone. In order to decide whether the implanted MSCs had already been eliminated during host cell infiltration of the fibrin–MSC composite or whether they persisted beside invaders, histological sections from day 6 were processed by laser microdissection to assess non-infiltrated and infiltrated areas separately. Genomic DNA from both areas revealed a clear SRY-gene signal after nested
  • 9. 4738 E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 Fig. 6. Bone regeneration at day 14 and day 28 after surgery and persistence of implanted MSCs at the defect site. (A) 3-D reconstruction of the bone defect 14 days after surgery based on lCT scanning. Sides of the defect gap were rotated apart to give an open view into the proximal (left) and distal (right) bone endings. Newly formed bone tissue is depicted by colour. (B) Total callus was digested for DNA extraction and absolute content of genomic DNA was determined spectrophotometrically (control group: day 14: n = 3 and day 28: n = 2; MSC-treated group: day 14 and day 28: n = 4). Boxes represent the 25th and 75th percentile, median is given as horizontal line and whiskers are maximal and minimal values. Significant difference (P 0.05) between with MSCs and fibrin alone at day 14 is designated by an asterisk. (C–E) Detection of implanted male MSCs by SRY-gene-specific PCR at indicated time points: () animals of control group and (+) animals receiving MSCs. Arrows indicate the SRY-gene-specific fragment with a length of 159 bp. (F) Evaluation of implanted male MSCs persisting besides invading host cells: histological sections of the MSC-callus were subjected to laser microdissection to separate areas infiltrated by host cells and non-infiltrated areas. Dissected samples of similar areas were pooled, DNA was extracted and SRY-gene-specific nested PCR was performed. P, positive control (male MSCs); N, control (water); 0, laser microdissection cDNA control; 1, remaining fibrin clot; 2, area infiltrated by host cells; 3, female control callus. PCR (Fig. 6F), while control samples from sections of cell-free explants were negative. This indicated that the MSCs implanted in fibrin hydrogel were not immediately disappearing behind the invasion front after host cell infiltration. 4. Discussion Recent investigations in tissue regeneration have focused on the activation of endogenous mechanisms at the defect site by trophic and immunomodulatory activity of implanted MSCs [12,13]. Our study for the first time investigates prevailing early cellular and molecular actions of fibrin–MSC composites implanted into a long bone defect to stimulate long bone healing. Our results highlight a strong early impact of implanted MSCs on host M1 macrophage and endothelial progenitor cell recruitment into the fibrin carrier which may be due to the presence of higher pro-angiogenic VEGF levels in MSC implants vs. controls. However, other chemokines, such as IL-6 and MIP-2 and other molecules of the MSC secretome [30,31], may contribute to this effect. Furthermore, activation of the MSCs by the fibrin hydrogel itself should be considered [32]. Since local expression of pro-inflammatory cytokines IL-1b and TNF-a and of anti-inflammatory IL-10 per immune cell was unal-tered in the MSC group vs. control callus at day 3 and day 6, MSCs here displayed no evident early immunomodulatory activity in fully immune competent animals. Accelerated early tissue matura-tion in the MSC group occurred despite a loss of implanted male MSCs before day 28 in our model. Overall, the fibrin carrier proved attractive for MSC implantation into bone defects permitting generation of a trophic and pro-angiogenic microenvironment by which MSCs stimulated implant integration and endogenous repair mechanisms without evident early immunomodulation or long-term osteogenic contribution to the repair tissue. 4.1. MSCs as a trigger for cell invasion into fibrin hydrogel Cellular and molecular mechanisms of natural bone remodel-ling revealed a crucial role for resident tissue macrophages for full functional differentiation of osteoblasts and their maintenance in vivo [33]. In wound healing, one important early wave of cell migration towards a site of tissue injury is conducted by
  • 10. E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 4739 macrophages around 2–6 days after injury [34]. Macrophage-secreted cytokines affect endothelial cell proliferation, angiogene-sis and collagen synthesis [34], suggesting that they may act in concert with other resident cells to stimulate vascularization. Our model took advantage of an empty and a MSC-seeded fibrin hydrogel clot, allowing us to distinguish early arriving and possibly faster migrating cells penetrating deep into the implant, from later arriving and/or later migrating cells that are still closer to the implant edges on day 6. We consistently observed earlier host cell infiltration from the proximal defect side which is possibly caused by a relative shortage of blood-borne invading host cells at the dis-tal defect ending after disruption of blood flow by defect creation. Remarkably, host cell infiltration was only initiated in fibrin–MSC composites while host cells apparently showed little interest in invading a cell-free fibrin hydrogel in a bone defect. This demon-strated that signals from viable MSCs represented an important trigger for implant integration and remodelling, emphasizing the importance of—most likely—soluble factors as trophic mediators of MSC action. Most importantly, although the local number of osteoprogenitor cells in the bone cavity is high and may suffice to initiate bone healing, defect filling with empty fibrin hydrogel alone cannot be recommended due to its rather inert persistence without evident cell invasion. A large panel of trophic factors is secreted by MSCs [30,31] of which IL-6, VEGF and the neutrophil-attracting factor MIP-2 were here shown to be expressed by the fibrin-embedded MSCs before implantation. This first demonstration of significantly higher VEGF mRNA levels in vivo and differential expression patterns between fibrin–MSC composites and cell-free controls suggest VEGF as an important candidate molecule for endothelial cell attraction, angi-ogenesis, implant remodelling and bone regeneration in line with the literature [35–37]. Whether the higher VEGF mRNA levels resulted from an up-regulation of VEGF in MSCs or is contributed by attracted macrophages and endothelial cells remains to be determined. However, MSCs not only secrete growth factors and chemokines but also produce matrix-degrading proteases and extracellular matrix molecules which might act on angiogenesis and help in cell migration [38,39]. Overall our study, thus, demon-strates that fibrin supports MSCs in organizing a beneficial envi-ronment for bone regeneration by facilitating host macrophage invasion relevant for tissue remodelling and enhancing endothelial progenitor cell migration indispensible for new blood vessel formation. 4.2. The cascade of invading cells Pioneering cells penetrating deep into the implant were mainly CD68-positive macrophages accompanied and followed by CD31- positive endothelial progenitor cells. To our knowledge this is the first study demonstrating a cell-dense migration front as a domi-nant structure of in vivo integration and remodelling of a fibrin– MSC composite. This cell-dense structure developing always from the proximal bone ending may thus be a special feature of long bone healing or be related to the fibrin hydrogel. At ectopic sites in immune-competent [40,41] and immune-deficient host animals [42,43] no cell-dense invasion front of host cells into tissue engi-neering constructs has been observed. This structure may so far, however, have escaped detection either because only late observa-tion time points were chosen or because no histology was performed at the early time points. In many stress situations monocytes enter the damaged area and differentiate into a spectrum of mononuclear phagocytes. These newly recruited cells usually exhibit pro-inflammatory action, and in the tissue inflammatory mediators such as IL-1, IL-12 and TNF-a must be balanced with the need to protect tissue integrity [44]. Our data for the first time provide evidence that pro-inflammatory M1 macrophages also prevailed early during long bone healing, while CD163-positive ‘‘anti-inflammatory’’ M2 macrophages [45] remained rare at day 6. Macrophages remain responsive to further stimuli after their initial activation, are by no means restricted to only these two categories [46], and M1 mac-rophages can convert themselves into anti-inflammatory macro-phages with an M2 wound-healing phenotype [47]. Further studies at later time points are important to fully understand the cascade of invading cells and their fate during long bone healing. 4.3. Immunomodulation by fibrin-based MSC implantation To analyze the effect of fibrin-implanted MSCs on the inflam-matory phase of bone healing, we looked for gene expression of CD45 as well as IL-1b, TNF-a and IL-10 which represent relevant immune mediators during early bone regeneration [48,49]. The natural callus adjacent to cell-free fibrin hydrogel in the control group contained a higher concentration of CD45-positive immune cells than MSC-seeded implants and thus contained higher mRNA levels of pro- and anti-inflammatory cytokines at day 3. The pres-ence of MSCs in the cell-seeded fibrin hydrogel did not only dilute the inflammatory cytokine-producing immune cells of the early callus, but can also be expected to actively contribute with a broad panel of trophic factors. These beneficial effects ultimately pro-duced a stimulatory microenvironment evident from histology, organizing remodelling of the fibrin hydrogel. Suppressive actions of MSCs towards immune cells, as reported in other settings in vitro and in vivo [50], were not apparent at this stage, since the average levels of IL-1b, TNF-a and IL-10 per CD45-positive immune cell were unaltered. Although Sprague–Dawley rats have frequently been used for studying outcome after cell and tissue transplantation between animals with no evidence for strain-intern rejection [51–53], we looked for local gene expression of the T-cell mediator IL-2 as an early rejection marker after allogeneic implantation of MSCs. IL-2 mRNA levels were not increased in the MSC group at day 3 and day 6, indicating that no acute T-cell-mediated rejection of the implanted MSCs occurred within the fibrin hydrogel within the first 6 days, in line with the findings in a syngeneic ectopic mouse model [40]. Overall, this argues in favour of a microenvironment in MSC-seeded fibrin hydrogel without early silencing of host immune cells at days 3 and 6. 4.4. Tissue maturation and the fate of fibrin-implanted MSCs According to detection of the implanted MSCs in our histologi-cal sections at day 6, fibrin hydrogel was stable and cell supportive enough to enable a local persistence of implanted MSCs for at least 6 days in a bone defect without connection to the vasculature. Fur-thermore, according to detection of male DNA in laser-dissected tissue areas behind the migration front, arriving immune cells did not immediately degrade implanted MSCs during invasion. However, since all cells were lost within 4 weeks after implanta-tion, either their limited lifespan, a failure of re-adaptation to changing needs during tissue maturation or other weaknesses may have caused their ultimate replacement by host-derived osteoprogenitor cells localized in adjacent bone marrow. In ectopic models a mineral component such as beta-tricalcium phosphate (b-TCP) or hydroxyapatite-tricalcium phosphate (HA/TCP) is required for long-term MSC engraftment and MSC-mediated bone formation [18,54]. Due to our interest in the early immunomodu-latory actions of implanted MSCs, we decided to use a fibrin carrier without ceramic particles since some inflammatory activity has been reported for b-TCP [55,56] which would bias our results. In our setting it remains unclear whether or not fibrin-implanted MSCs have physically contributed to the maturing osteogenic
  • 11. 4740 E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 tissue of the MSC group until day 14 before replacement by host bone. Although sex-mismatched animal models are well estab-lished for studying MSC-mediated bone formation [21,57] and ele-gantly permit the detection of persisting male cells in female recipients without a need for cell labelling [58], a long-term rejec-tion of male cells by the female immune system may have contrib-uted to our result. The transient role of fibrin-implanted MSCs seen here is, however, in line with data from genetically engineered ASCs, which effectively stimulated femoral bone healing in rabbits, despite some induction of a humoral- and cell-mediated immune response and limited cell persistence over 4 weeks [59]. 5. Conclusions In summary, by using fibrin hydrogel—known for its biocom-patibility, biodegradability and cell support for implantation of MSCs into a long bone defect—we identified attraction of M1 mac-rophages and endothelial progenitor cells into the implant as a main early beneficial action of fibrin–MSC composites, promoting implant integration, angiogenesis and tissue maturation during bone regeneration. No evidence for an early immune suppression by fibrin-implanted MSCs was obtained, as expression levels of the pro-inflammatory cytokines IL-1b and TNF-a as well as the anti-inflammatory cytokine IL-10 by the invading immune cells were unaltered. Trophic and pro-angiogenic stimulation during bone regeneration occurred independently of a long-term engraft-ment of the implanted MSCs at the defect site. Early recruitment of host repair cells into fibrin hydrogel was most likely stimulated through the secretion of bioactive factors including VEGF whose elevated levels may have generated a pro-angiogenic environment beneficial for bone regeneration. With this advanced understand-ing of the mechanisms involved in fibrin hydrogel-based MSC-sup-ported bone regeneration, we suggest fibrin hydrogel can serve as an attractive carrier for MSC-based tissue engineering approaches in long bone repair, supporting early trophic and pro-angiogenic activities to improve clinical treatment of atrophic pseudarthrosis, bone injury or tumor resection. Disclosure The authors indicate no potential conflicts of interest. Acknowledgements We thank Svenja Schäfer and Viviana Grajales for their support with the surgery and with animal care. Furthermore we thank Nicole Buchta and Birgit Frey for their help with the experiments and Simone Gantz for her statistical support. This study (grant RI 707/8-1) was funded by the priority program SPP1468 ‘‘Immuno-bone’’ of the German Research Foundation (DFG). Appendix A. Figures with essential color discrimination Certain figures in this article, particularly Figs. 1, 2 and 6 are difficult to interpret in black and white. The full color images can be found in the on-line version, at http://dx.doi.org/10.1016/ j.actbio.2014.07.017. References [1] Patterson TE, Kumagai K, Griffith L, Muschler GF. Cellular strategies for enhancement of fracture repair. J Bone Joint Surg Am 2008;90(Suppl. 1):111–9. [2] Wu X, Ren J, Li J. Fibrin glue as the cell-delivery vehicle for mesenchymal stromal cells in regenerative medicine. Cytotherapy 2012;14:555–62. [3] Bensaid W, Triffitt JT, Blanchat C, Oudina K, Sedel L, Petite H. A biodegradable fibrin scaffold for mesenchymal stem cell transplantation. Biomaterials 2003;24:2497–502. [4] Dickhut A, Gottwald E, Steck E, Heisel C, Richter W. Chondrogenesis of mesenchymal stem cells in gel-like biomaterials in vitro and in vivo. Front Biosci 2008;13:4517–28. [5] Sierra DH. Fibrin sealant adhesive systems: a review of their chemistry, material properties and clinical applications. J Biomater Appl 1993;7:309–52. [6] Steck E, Fischer J, Lorenz H, Gotterbarm T, Jung M, Richter W. Mesenchymal stem cell differentiation in an experimental cartilage defect: restriction of hypertrophy to bone-close neocartilage. Stem Cells Dev 2009;18:969–78. [7] Jung M, Kaszap B, Redohl A, Steck E, Breusch S, Richter W, et al. Enhanced early tissue regeneration after matrix-assisted autologous mesenchymal stem cell transplantation in full thickness chondral defects in a minipig model. Cell Transplant 2009;18:923–32. [8] Friedenstein AJ, Petrakova KV, Kurolesova AI, Frolova GP. Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation 1968;6:230–47. [9] Sacchetti B et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 2007;131:324–36. [10] Dominici M et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006;8:315–7. [11] Pittenger MF et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284:143–7. [12] Meirelles Lda S, Fontes AM, Covas DT, Caplan AI. Mechanisms involved in the therapeutic properties of mesenchymal stem cells. Cytokine Growth Factor Rev 2009;20:419–27. [13] Caplan AI, Correa D. The MSC: an injury drugstore. Cell Stem Cell 2011;9:11–5. [14] Kadiyala S, Jaiswal N, Bruder SP. Culture-expanded, bone marrow-derived mesenchymal stem cells can regenerate a critical-sized segmental bone defect. Tissue Eng 1997;3:173–85. [15] Bruder SP, Kraus KH, Goldberg VM, Kadiyala S. The effect of implants loaded with autologous mesenchymal stem cells on the healing of canine segmental bone defects. J Bone Joint Surg Am 1998;80:985–96. [16] Quarto R et al. Repair of large bone defects with the use of autologous bone marrow stromal cells. N Engl J Med 2001;344:385–6. [17] Marcacci M et al. Stem cells associated with macroporous bioceramics for long bone repair: 6- to 7-year outcome of a pilot clinical study. Tissue Eng 2007;13:947–55. [18] Janicki P, Boeuf S, Steck E, Egermann M, Kasten P, Richter W. Prediction of in vivo bone forming potency of bone marrow-derived human mesenchymal stem cells. Eur Cell Mater 2011;21:488–507. [19] Janicki P, Kasten P, Kleinschmidt K, Luginbuehl R, Richter W. Chondrogenic pre-induction of human mesenchymal stem cells on beta-TCP: enhanced bone quality by endochondral heterotopic bone formation. Acta Biomater 2010;6:3292–301. [20] Giannoni P, Scaglione S, Daga A, Ilengo C, Cilli M, Quarto R. Short-time survival and engraftment of bone marrow stromal cells in an ectopic model of bone regeneration. Tissue Eng Part A 2010;16:489–99. [21] Boukhechba F et al. Fate of bone marrow stromal cells in a syngenic model of bone formation. Tissue Eng Part A 2011;17:2267–78. [22] Zimmermann CE, Gierloff M, Hedderich J, Acil Y, Wiltfang J, Terheyden H. Survival of transplanted rat bone marrow-derived osteogenic stem cells in vivo. Tissue Eng Part A 2011;17:1147–56. [23] Prockop DJ. Repair of tissues by adult stem/progenitor cells (MSCs): controversies, myths, and changing paradigms. Mol Ther 2009;17:939–46. [24] Hughes PC, Tanner JM. The assessment of skeletal maturity in the growing rat. J Anat 1970;106:371–402. [25] Dobson KR, Reading L, Haberey M, Marine X, Scutt A. Centrifugal isolation of bone marrow from bone: an improved method for the recovery and quantitation of bone marrow osteoprogenitor cells from rat tibiae and femurae. Calcif Tissue Int 1999;65:411–3. [26] Meirelles Lda S, Nardi NB. Murine marrow-derived mesenchymal stem cell: isolation, in vitro expansion, and characterization. Br J Haematol 2003;123:702–11. [27] Winter A et al. Cartilage-like gene expression in differentiated human stem cell spheroids: a comparison of bone marrow-derived and adipose tissue-derived stromal cells. Arthritis Rheum 2003;48:418–29. [28] Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 2004;25:677–86. [29] Brown BN, Valentin JE, Stewart-Akers AM, McCabe GP, Badylak SF. Macrophage phenotype and remodeling outcomes in response to biologic scaffolds with and without a cellular component. Biomaterials 2009;30:1482–91. [30] Wagner W et al. Molecular and secretory profiles of human mesenchymal stromal cells and their abilities to maintain primitive hematopoietic progenitors. Stem Cells 2007;25:2638–47. [31] Kinnaird T, Stabile E, Burnett MS, Lee CW, Barr S, Fuchs S, et al. Marrow-derived stromal cells express genes encoding a broad spectrum of arteriogenic cytokines and promote in vitro and in vivo arteriogenesis through paracrine mechanisms. Circ Res 2004;94:678–85. [32] Huang NF, Chu J, Lee RJ, Li S. Biophysical and chemical effects of fibrin on mesenchymal stromal cell gene expression. Acta Biomater 2010;6:3947–56. [33] Raggatt LJ, Partridge NC. Cellular and molecular mechanisms of bone remodeling. J Biol Chem 2010;285:25103–8. [34] Park JE, Barbul A. Understanding the role of immune regulation in wound healing. Am J Surg 2004;187:11S–6S.
  • 12. E. Seebach et al. / Acta Biomaterialia 10 (2014) 4730–4741 4741 [35] Geiger F et al. VEGF producing bone marrow stromal cells (BMSC) enhance vascularization and resorption of a natural coral bone substitute. Bone 2007;41:516–22. [36] Kumar S, Wan C, Ramaswamy G, Clemens TL, Ponnazhagan S. Mesenchymal stem cells expressing osteogenic and angiogenic factors synergistically enhance bone formation in a mouse model of segmental bone defect. Mol Ther 2010;18:1026–34. [37] Lin CY et al. Long-term tracking of segmental bone healing mediated by genetically engineered adipose-derived stem cells: focuses on bone remodeling and potential side effects. Tissue Eng Part A 2014;20:1392–402. [38] Choi YA et al. Secretome analysis of human BMSCs and identification of SMOC1 as an important ECM protein in osteoblast differentiation. J Proteome Res 2010;9:2946–56. [39] Polacek M, Bruun JA, Elvenes J, Figenschau Y, Martinez I. The secretory profiles of cultured human articular chondrocytes and mesenchymal stem cells: implications for autologous cell transplantation strategies. Cell Transplant 2011;20:1381–93. [40] Tasso R et al. Recruitment of a host’s osteoprogenitor cells using exogenous mesenchymal stem cells seeded on porous ceramic. Tissue Eng Part A 2009;15:2203–12. [41] Tasso R, Fais F, Reverberi D, Tortelli F, Cancedda R. The recruitment of two consecutive and different waves of host stem/progenitor cells during the development of tissue-engineered bone in a murine model. Biomaterials 2010;31:2121–9. [42] Tortelli F, Tasso R, Loiacono F, Cancedda R. The development of tissue-engineered bone of different origin through endochondral and intramembranous ossification following the implantation of mesenchymal stem cells and osteoblasts in a murine model. Biomaterials 2010;31:242–9. [43] Ghanaati S et al. Scaffold vascularization in vivo driven by primary human osteoblasts in concert with host inflammatory cells. Biomaterials 2011;32:8150–60. [44] Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol 2011;11:723–37. [45] Ambarus CA et al. Systematic validation of specific phenotypic markers for in vitro polarized human macrophages. J Immunol Methods 2012;375: 196–206. [46] Novak ML, Koh TJ. Macrophage phenotypes during tissue repair. J Leukoc Biol 2013;93:875–81. [47] Arnold L et al. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. J Exp Med 2007;204:1057–69. [48] Mountziaris PM, Mikos AG. Modulation of the inflammatory response for enhanced bone tissue regeneration. Tissue Eng Part B Rev 2008;14:179–86. [49] Kolar P et al. The early fracture hematoma and its potential role in fracture healing. Tissue Eng Part B Rev 2010;16:427–34. [50] Ghannam S, Bouffi C, Djouad F, Jorgensen C, Noel D. Immunosuppression by mesenchymal stem cells: mechanisms and clinical applications. Stem Cell Res Ther 2010;1:2. [51] Larsson J, Juliusson B, Holmdahl R, Ehinger B. MHC expression in syngeneic and allogeneic retinal cell transplants in the rat. Graefes Arch Clin Exp Ophthalmol 1999;237:82–5. [52] Troum S, Dalton Jr ML. Osteogenesis in a rat model: use of bone marrow cells and biodegradable gelatin matrix carrier. J South Orthop Assoc 2001;10: 37–43. [53] Yang Y, Rossi FM, Putnins EE. Periodontal regeneration using engineered bone marrow mesenchymal stromal cells. Biomaterials 2010;31:8574–82. [54] Krebsbach PH, Kuznetsov SA, Satomura K, Emmons RV, Rowe DW, Robey PG. Bone formation in vivo: comparison of osteogenesis by transplanted mouse and human marrow stromal fibroblasts. Transplantation 1997;63:1059–69. [55] Lange T, Schilling AF, Peters F, Haag F, Morlock MM, Rueger JM, et al. Proinflammatory and osteoclastogenic effects of beta-tricalciumphosphate and hydroxyapatite particles on human mononuclear cells in vitro. Biomaterials 2009;30:5312–8. [56] Curran JM, Gallagher JA, Hunt JA. The inflammatory potential of biphasic calcium phosphate granules in osteoblast/macrophage co-culture. Biomaterials 2005;26:5313–20. [57] Nakamura A et al. Cell sheet transplantation of cultured mesenchymal stem cells enhances bone formation in a rat nonunion model. Bone 2010; 46:418–24. [58] An J, Beauchemin N, Albanese J, Abney TO, Sullivan AK. Use of a rat cDNA probe specific for the Y chromosome to detect male-derived cells. J Androl 1997;18:289–93. [59] Lin CY et al. Immune responses during healing of massive segmental femoral bone defects mediated by hybrid baculovirus-engineered ASCs. Biomaterials 2012;33:7422–34.