You are on page 1of 7

Journal of Oral and Maxillofacial Surgery, Medicine, and Pathology 29 (2017) 563–569

Contents lists available at ScienceDirect

Journal of Oral and Maxillofacial Surgery,


Medicine, and Pathology
journal homepage: www.elsevier.com/locate/jomsmp

Oral Medicine/Original research

Comparison study between plasma rich in growth factors and


platelet-rich plasma for osteoconduction in rat calvaria
Takashi Eda ∗ , Kosuke Takahashi, Shingo Kanao, Akinobu Aoki, Naomi Ogura, Ko Ito,
Hiroyasu Tsukahara, Masaaki Suemitsu, Kayo Kuyama, Toshirou Kondoh
Department of Maxillofacial Surgery, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakaecho-Nishi, Matsudo, Chiba 271-8587, Japan

a r t i c l e i n f o a b s t r a c t

Article history: Objective: Plasma rich in growth factors (PRGF) and platelet-rich plasma (PRP) can be rapidly obtained
Received 22 January 2017 from patient blood. They are a new and potentially useful adjunct in oral and maxillofacial bone repair or
Received in revised form 9 April 2017 regenerative surgery. The aim of this study was to compare the possibility of new bone formation using
Accepted 28 June 2017
PRGF and PRP.
Available online 13 July 2017
Methods: The osteogenic potential with transplantation of PRGF or PRP onto rat calvaria was evaluated
by histologic examination and microCT. PRGF or PRP was prepared by centrifugation of rat whole blood
Keywords:
(WB). First, the cells in the blood product were counted; there were no leukocytes in PRGF, and PRP
Plasma rich in growth factors
Platelet-rich plasma
included leukocytes. PRGF contained higher levels of TGF-␤1 and PDGF-BB than PRP. Furthermore, PRGF
Bone regeneration or PRP was transplanted onto calvarial bone of rats.
Results: MicroCT showed that PRGF promoted an increase in bone volume when compared to PRP. His-
tological observation demonstrated that the PRGF group showed newly formed bone in a wide range. In
addition, the PRP group showed numerous inflammatory cells compared to the PRGF group in HE-stained
specimens. This suggests that PRP might delay bone regeneration due to the inflammatory response.
Conclusions: PRGF has more availability for bone regeneration than PRP, and PRGF may be useful in bone
regeneration treatment.
© 2017 Asian AOMS, ASOMP, JSOP, JSOMS, JSOM, and JAMI. Published by Elsevier Ltd. All rights reserved.夽

1. Introduction to have several advantages, including the enhancement of bone


regeneration and rapid soft tissue healing [11]. This system is
Platelet concentration products, which are autologous con- advantageous as it requires only one step of centrifugation and
stituents of inductive factors obtained from blood, have high is leukocyte-free, thus avoiding higher levels of pro-inflammatory
concentrations of platelets containing various growth factors [1]. cytokines [3]. In addition, PRGF contains high levels of growth
The concentration of autologous platelets in plasma [platelet-rich factors such as transforming growth factor (TGF)-␤ and platelet-
plasma (PRP)] and the growth factors contained within platelets derived growth factor (PDGF), which are associated with bone
have been studied since 1998 [2]. The previous study reported that regeneration [12]. However, there has been little basic research into
the combined use of autogenous bone with PRP increased radio- the efficacy of PRGF in bone regeneration [9].
graphic and histomorphometric bone densities [2]. In addition, PRP The aim of this study was to compare between PRGF and PRP
has been used in bone augmentation for dental implants or frac- to the availability of bone formation in transplantation of using
ture healing of jaw bone [2–5]. However, it has been shown that histologic findings and microCT analysis.
PRP formulations have different biological activities, depending on
their preparation and administration [6–8].
The preparation of plasma rich in growth factors (PRGF) is one 2. Materials and methods
way to concentrate platelets [9,10], and in 1999, it was shown
2.1. Preparations of PRGF and PRP

夽 AsianAOMS: Asian Association of Oral and Maxillofacial Surgeons; ASOMP: Asian A total of 27 Sprague-Dawley male rats (age: 15 weeks) weigh-
Society of Oral and Maxillofacial Pathology; JSOP: Japanese Society of Oral Pathol- ing 405–415 g were purchased from Japan SLC (Shizuoka, Japan).
ogy; JSOMS: Japanese Society of Oral and Maxillofacial Surgeons; JSOM: Japanese
Society of Oral Medicine; JAMI: Japanese Academy of Maxillofacial Implants.
Rat whole blood (5 ml) was collected from the external jugular
∗ Corresponding author. vein by syringe aspiration via direct venipuncture with a 21-gauge
E-mail address: Eda.takashi.12.22@gmail.com (T. Eda). needle. Blood was immediately placed into 5-ml sterile extraction

http://dx.doi.org/10.1016/j.ajoms.2017.06.011
2212-5558/© 2017 Asian AOMS, ASOMP, JSOP, JSOMS, JSOM, and JAMI. Published by Elsevier Ltd. All rights reserved.夽
564 T. Eda et al. / Journal of Oral and Maxillofacial Surgery, Medicine, and Pathology 29 (2017) 563–569

Fig. 1. Blood plasma fractions were obtained from citrate-anticoagulated whole blood.
(A) Tubes were centrifuged at 460g for 8 min to give PRGF. The bottom fraction contains erythrocytes, and the top fraction contains platelets. Both fractions are separated
by the whitish buffy coat, which contains leukocytes. The plasma fraction (1 ml over the buffy coat) was collected to prepare Fraction 2 (F2). Fraction 1 (F1) was prepared
from the upper layer of F2. (B, C) PRP was prepared at a separation spin of 1000 × g for 4 min, followed by a concentration spin of 800 × g for 9 min. (B) The layer of plasma
containing the buffy coat was concentrated. (C) Plasma (500 ␮l) containing WBCs and RBCs were collected to prepare PRP. PPP was prepared from the upper layer of PRP in
plasma.

tubes containing 0.5 ml of 3.8% sodium citrate as an anticoagulant ter, 4 mm; height, 2 mm) in vivo. Rats were randomly divided into
(BTI Biotechnology Institute, Vitoria, Spain). three groups (n = 3 per group): the Control group (PTFE tube only),
PRGF was prepared from whole blood (WB) centrifuged in accor- the PRP group (PRP in the PTFE tube), and the PRGF group (PRGF F2
dance with Anitua’s protocol [9,10,13] (Fig. 1A). Briefly, tubes were in the PTFE tube).
centrifuged at 460g for 8 min. The bottom fraction containing ery- All animals were anesthetized by intraperitoneal injection of
®
throcytes and the top fraction containing platelets are separated 25 mg/kg pentobarbital sodium (Somnopentyl ; Kyoritsu Seiyaku,
by the whitish buffy coat, which contains leukocytes. The plasma Tokyo, Japan), and the surgical site was prepared aseptically. A
fraction (1 ml over the buffy coat) was collected as Fraction 2 (F2), previous study reported that PRGF F2 had a higher platelet con-
while Fraction 1 (F1) was the layer above F2. centration and growth factors compared with PRGF F1 [9,13]. From
PRP was prepared from WB using the double-spin method this kind of reason, we used F2 in transplantation. Platelets in PRP
described by Marx [5] (Fig. 1B and C). Briefly, WB was centrifuged or PRGF F2 were activated using 10% calcium chloride solution just
at 1000 × g for 4 min, and the plasma layer containing the buffy prior to use [9,14,15]. Transplantation protocol into rats was per-
coat was collected (Fig. 1B), followed by centrifugation at 800g for formed as in previous reports [16–18]. Briefly, a linear skin incision
9 min. The lower plasma layer (500 ␮l) containing white blood cells was made along the edge of the skull with a #15 surgical blade.
(WBCs) and red blood cells (RBCs) was collected to prepare PRP. The The periosteum of the calvaria was carefully removed with a ras-
upper layer was platelet-poor plasma (PPP) (Fig. 1C). All animals patorium after elevation of the skin-periosteum flap. PRP or PRGF
were maintained and used in accordance with the Nihon University F2 was inserted into PTFE tubes, which were then placed on the
Intramural Animal Use guidelines (Ethics Committee Registration calvarial surface. Overlying tissue was closed with sutures.
No: AP12MD018).
2.4. MicroCT analysis of transplants
2.2. Characteristics of autologous blood product
Quantitative image analysis of new bone formation was per-
Platelets, WBCs, and RBCs in PRGF and PRP were counted using formed using an in vivo microCT system (Rigaku-mCT, Tokyo,
Celltac ␣ (MEK-6358; NIHON KOHDEN, Tokyo, Japan). Samples of Japan). Rats were deeply anesthetized with intraperitoneal injec-
serum, PPP, PRP, PRGF F1, and PRGF F2 for evaluating growth factor tion of sodium pentobarbital (25 mg/kg) and individually placed on
concentrations were stored at −80 ◦ C before use. Levels of TGF-␤1, the object stage. Calvarial bone at the PTFE tube site was scanned
PDGF-BB, and insulin-like growth factor (IGF)-1 were measured using a microCT system with an X-ray source of 90 kV/150 ␮A at
using ELISA (enzyme-linked immunosorbent assay) kits (Quan- 2, 4, and 8 weeks after transplantation. Imaging of newly formed
tikine colorimetric ELISA kits; R&D Systems, Minneapolis, MN) in bone in the area containing the inner cavity of the PTFE tube was
accordance with the manufacturer’s instructions. then performed over a full 360◦ , with an exposure time of 2 min.
An isotropic resolution of 20 × 20 × 20 ␮m3 voxels was selected,
2.3. Transplantation procedure which displayed the microstructure of new bone formation in rats.
The original 3D images were displayed and analyzed using I-view
The transplantation procedure was designed to evaluate newly software (J. Morita, Kyoto, Japan). Analysis of new bone formation
formed bone in the presence of PRP or PRGF F2 with artificial dam was performed in the area containing the inner cavity of the PTFE
preparation using PTFE tubes (inner diameter, 3 mm; outer diame- tube. Linearity of the micro-CT scanner was established by scanning
T. Eda et al. / Journal of Oral and Maxillofacial Surgery, Medicine, and Pathology 29 (2017) 563–569 565

Fig. 2. Counting of inflammatory cells.


(A) The degree of inflammation is objectively assessed using ImageJ in terms of number of inflammatory cells, such as plasma cells and lymphocytes per unit area
(500 × 500 ␮m) in the tube center by H-E staining at 2 weeks after surgery. Columns represent mean ± standard deviation. ** P < 0.01. (B-D) Inflammatory cells are observed
by H-E staining at 2 weeks after surgery. The control group (B), the PRGF group (C) and the PRP group (D). Original magnification × 200.

a phantom containing several densities of a standard calibration Table 1


Cell count of platelet, white blood cell, and red blood cell in plasma preparations
material. The 3D image of newly formed bone was measured, and
(n = 3).
structural indices of the newly formed bone were calculated using
a morphometric program (TRI/3D-BON; Ratoc System Engineering, Sample Platelets ( × 104 /␮l) WBCs ( × 103 /␮l) RBCs ( × 104 /␮l)
Tokyo, Japan). 3D analysis directly measured bone mineral den- WB 97.7 ± 1.9 5.3 ± 0.1 908.0 ± 10.0
sity (BMD = BMC/BV; mg/cm3 ), bone mineral content (BMC; mg), F1 44.0 ± 2.6 0 4.6 ± 1.6
and bone volume (BV; cm3 ) of the new bone formation in the cav- F2 241.4 ± 19.2 0 84.6 ± 1.6
PPP 0.7 ± 0.4 0 0.8 ± 0.2
ity of the PTFE tube. In this experiment, a circle with a radius of
PRP 361.2 ± 34.0 2.8 ± 2.3 28.5 ± 8.6
1.5 mm defined to a height of 2.0 mm from the parietal bone to the
(WB; whole blood, F1; PRGF Fraction 1, F2; Fraction 2, PPP; platelet-poor plasma,
periosteum along PTFE tubes as the ROI of bone mineral quantity
PRP; platelet-rich plasma, WBCs; white blood cells, RBCs; red blood cells).
analysis.

2.5. Histologic analysis LB4005;Cosmo Bio, Tokyo, Japan) for 1 h at room temperature.
Secondary antibody staining was performed with reagents from
Rats were sacrificed 2, 4, and 8 weeks after transplantation. ChemMate ENVISION kit/HRP (DAB; Dako, Glostrup, Denmark), in
The calvarial bone area including the PTFE tube was extirpated accordance with the manufacturer’s protocol. Immune complexes
en bloc. Samples were fixed in 10% neutral-buffered formalin solu- were visualized using 3 3 -diaminobenzidine tetrachloride (DAB;
tion, decalcified in 10% EDTA (pH 7.0), and embedded in paraffin. Merck, Darmstadt, Germany). Immunostained sections were then
Semi-serial sections (4 ␮m) were obtained in the sagittal orienta- counterstained with Mayer’s hematoxylin. Normal bone was used
tion. Sections were stained with hematoxylin and eosin (H-E) and as a positive control in immunochemical staining, and negative
observed by light microscopy. Newly formed bone was defined as controls were prepared by omitting primary antibody [21].
bone-like hard tissue arranged regularly with osteoblasts between
the cortical bone and periosteum. Inflammatory changes were 2.7. Statistical analysis
observed in each group at 2 weeks after surgery in H-E-stained
specimens. At that time, infiltration of inflammatory cells was All results are expressed as the mean ± standard deviation (SD).
observed, and the density of inflammatory cells, such as plasma The ANOVA test was used to compare three groups. Tukey’s test
cells and lymphocytes, in the tissue was elevated. The degree of followed the ANOVA test. The significance of differences was evalu-
inflammation was objectively assessed using ImageJ in terms of ated by Tukey’s test under a normal distribution and equal variance.
the number of inflammatory cells per unit area [19,20]. The origi- Differences were considered significant at P < 0.05.
nal file type of the photographs was Tagged Image File Format (TIFF)
with a size of 4080 × 3072 pixels (872 × 656 ␮m). Cell counting was 3. Results
performed (500 ␮m2 ) in the tube center under high magnifica-
tion (200 × ) (Fig. 2B-D). Images were binarized with thresholds. 3.1. Characteristics of autologous blood products
Nucleus counting was selected depending on pixels (pixel size
250–500) and circularity (0.80–1.00). The number of cell nuclei Concentrations of platelets, WBCs, and RBCs in each sample are
was considered to be equivalent to the number of inflammatory shown in Table 1. Platelet concentration was 2.5-fold higher in PRGF
cells (Fig. 2A). F2 and 3.7-fold higher in PRP, as compared to WB. Platelets were not
concentrated in PPP and PRGF F1 when compared to WB. No WBCs
2.6. Immunohistochemical analysis were present in PRGF F1, PRGF F2, or PPP, while PRP included WBCs.
Furthermore, all blood products contained RBCs.
Sections of calvarial bone including the PTFE tube from The concentrations of TGF-␤1, PDGF-BB, and IGF-1 were then
all groups were stained immunohistochemically. Sections were evaluated in each blood product. PRGF F1 and PRGF F2 contained
deparaffinized, and endogenous peroxidase activity was quenched higher levels of TGF-␤1, PDGF-BB, and IGF-1 when compared with
by incubation in 3% H2 O2 in methanol for 15 min at room temper- serum, PPP, and PRP (Table 2).
ature. Sections were washed with PBS (phosphate-buffered saline)
and incubated with goat polyclonal antibody against rat Runx2 3.2. MicroCT analysis
(1:50; sc-12488; SANTA CRUZ, Tokyo, Japan), rabbit polyclonal
antibody against rat osterix (1:100, ab22552; Abcam, Tokyo, Japan), MicroCT was used to quantify the mineralization within the
and rabbit polyclonal antibody against rat osteocalcin (1:100, PTFE tubes at 2, 4, and 8 weeks, allowing assessment of the entire
566 T. Eda et al. / Journal of Oral and Maxillofacial Surgery, Medicine, and Pathology 29 (2017) 563–569

Fig. 3. Color images showing bone mineral density by TRI/3D-BON.


Color images showed bone mineral density at (A-C) 2, (D-F) 4 and (G-I) 8 weeks; the control group (A, D, G), the PRGF group (B, E, H) and the PRP group (C, F, I). (B) The inner
side of the tube shows a small amount of newly formed bone cultured with PRGF at 2 weeks. (D–I) The area of new bone formation has progressed into the tube in all groups
at 4 and 8 weeks. Scale bar; 3000 ␮m.

Table 2 than in the PRP group at 2, 4, and 8 weeks (Fig. 4 D-F). Similarly,
Levels of growth factors in plasma preparations (n = 3).
the value for BV was higher in the PRGF group than in the control
Sample TGF-␤1 (ng/ml) PDGF-BB (ng/ml) IGF-1 (ng/ml) group at 8 weeks (Fig. 4F), although the values for BV did not differ
Serum N.D. N.D. 1187.50 ± 94.60 in the PRGF group when compared to the control group at 2 and 4
F1 31.35 ± 0.57 1.04 ± 0.31 1387.50 ± 47.80 weeks (Fig. 4D and E).
F2 115.80 ± 2.13 5.01 ± 0.48 1237.50 ± 125.00
PPP 5.67 ± 0.07 N.D. 1000.00 ± 40.00
PRP 7.40 ± 0.35 N.D. 876.67 ± 25.17 3.3. Histological findings
(WB; whole blood, F1; PRGF Fraction 1, F2; Fraction 2, PPP; platelet-poor plasma,
PRP; platelet-rich plasma, TGF; transforming growth factor, PDGF; platelet-derived Newly formed bone was observed between the periosteum and
growth factor, IGF; insulin-like growth factor). cortical bone in the PRGF group at 2 weeks after transplantation
(Fig. 5B). However, little newly formed bone was observed in the
PRP group and the control group (Fig. 5A and C). As compared to the
volume of the new bone. Fig. 3 shows BMD color images of trans-
other groups, the PRGF group showed a wide range of newly formed
plants with microCT analysis. The inner PTFE tube contained a small
bone at 4 and 8 weeks after surgery (Fig. 5E and H). In addition,
amount of newly formed bone on the calvarial side at 2 weeks
osteoblasts were observed around the cortical and newly formed
(Fig. 3A-C). A small volume of newly formed bone was seen in
bone (data not shown). Next, the number of inflammatory cells was
the PRGF group when compared with the control and PRP groups
counted in H-E-stained sections at 2 weeks after transplantation
(Fig. 3A-C). The newly formed bone area increased inside the PTFE
(Fig. 2B-D). The PRP group showed numerous inflammatory cells,
tube after 4 and 8 weeks, as compared to after 2 weeks (Fig. 3D-
such as plasma cells, when compared to the other groups in the
I). In addition, the newly formed bone area increased inside the
tube center at 2 weeks (Fig. 2A).
PTFE tube in the PRGF group when compared to the control and
PRP groups (Fig. 3B, E and H). Furthermore, the PRGF group mostly
showed newly formed bone in the PTFE tube center area at 8 weeks 3.4. Immunohistological findings
(Fig. 3H).
BMD and BV were then analyzed using microCT (Fig. 4). BMD Immunohistochemical findings for Runx2, osterix, and osteocal-
did not differ among the three experimental groups at 2, 4, and 8 cin are shown in Fig. 6. Osteoblasts positive for Runx2 and osterix
weeks (Fig. 4A-C). The values for BV was higher in the PRGF group were seen in the margin of newly formed bone in all experimental
T. Eda et al. / Journal of Oral and Maxillofacial Surgery, Medicine, and Pathology 29 (2017) 563–569 567

Fig. 4. Quantitative measurement of BMD and BV.


BMD (A) and BV (D) at 2 weeks, BMD (B) and BV (E) at 4 weeks, and BMD (C) and BV (F) at 8 weeks. (BMD; bone mineral density, BV; bone volume. Columns represent
mean ± standard deviation. * P < 0.05, ** P < 0.01, *** P < 0.005).

Fig. 5. Histologic findings.


H-E staining showed new bone formation at 2 (A-C), 4 (D-F) and 8 weeks (G-I); the control group (A, D, G), the PRGF group (B, E, H) and the PRP group (C, F, I). (A-C) New bone
formation is observed on the calvarial bone side at 2 weeks. (B) New bone formation is observed in the PRGF group at 2 weeks. (A, C) Little newly formed bone is observed in
the PRP group and the control group. (D–I) A wide range of newly formed bone formation is observed at 4 and 8 weeks. Original magnification × 40. Scale bar; 500 ␮m. NB;
new bone, CB; calvarial bone, T; tube.

groups, and positive osteocytes were seen around newly formed Runx2 (Fig. 6A-C) and osterix (Fig. 6D-F). Osteocalcin (Fig. 6G-I)
bone in the PRGF group at 2 weeks (Fig. 6A-F). Numerous swollen staining was also seen in osteoblasts arranged in the margin and in
spindle-shaped cells in the connective tissue were positive for osteocytes around newly formed bone in the PRGF group at 2 weeks
568 T. Eda et al. / Journal of Oral and Maxillofacial Surgery, Medicine, and Pathology 29 (2017) 563–569

Fig. 6. Representative immunohistochemistry.


Runx2 (A–C), osterix (D–F), and osteocalcin (G–I) at 2 weeks. the control group (A, D, G), the PRGF group (B, E, H), and the PRP group (C, F, I). Original magnification × 400.
Scale bar; 100 ␮m. NB; new bone, CB; calvarial bone. Arrows indicate positive reactions.

(Fig. 6H). However, osteocalcin staining was negative in the control phenomenon when bone resorption and formation are occurring
and PRP groups at 2 weeks (Fig. 6G and I). In addition, immunohis- simultaneously [28,29]. This suggests that the control and PRP
tochemical staining for Runx2, osterix, and osteocalcin was positive groups at 2 weeks showed no osteocalcin because bone calcification
in all groups at 4 and 8 weeks (data not shown). had not started. In other hands, PRGF groups at 2 weeks accelerate
osteogenic differentiation and bone calcification.
This study showed that PRGF F1 and F2 contained higher lev-
4. Discussion
els of TGF-␤1, PDGF-BB, and IGF-1 than PRP (Table 2). It is known
that TGF-␤1 plays an important role in bone metabolism and in
In this study, bone formation was compared between PRGF and
the early phase of bone regeneration and remodeling [30]. TGF-␤
PRP transplanted onto rat calvaria. The grafting of PRGF accelerated
stimulated osteoblast differentiation from mesenchymal stem cells
new bone formation more than PRP. New bone formation at the
from bone marrow [31,32]. In vivo studies have shown that a lack of
grafting sites in the PRGF group was observed earlier and in greater
TGF-ß1 leads to a weaker bone structure and less mechanical sta-
amounts than in the PRP and control groups on microCT analysis
bility [33]. PDGF was confirmed to have a positive effect on bone
and histological observation (Figs. 3 and 5).
healing, and there were generally higher concentrations of PDGF
Immunohistological observations showed that Runx2, osterix,
observed during bone repair [34,35]. IGF-I is one of the most abun-
and osteocalcin were expressed in osteocytes around the newly
dant growth factors in the human skeleton [36] and is thought to
formed bone (Fig. 6). Runx2 and osterix, which are transcription fac-
be important for both systemic and local functions in skeletal and
tors that are critical for bone formation [22–24]. In addition, osterix
bone metabolism [37]. A higher concentration of these growth fac-
acts downstream of Runx2 during the differentiation of pluripotent
tors in PRGF appears to be one of the advantageous factors for new
mesenchymal cells into the osteoblastic lineage [24–26]. Runx2
bone formation.
and osterix were detected in all experimental groups. However,
PRP had a higher platelet concentration than PRGF and included
Osteocalcin was detected in the PRGF groups earlier than in the
WBCs in this study. A previous study reported that PRP had a
PRP groups. Osteocalcin was positive in the PRGF group, whereas
high platelet concentration when compared with PRGF in humans
the control and PRP groups were negative, at 2 weeks after trans-
[2,9,10]. In addition, the PRP group showed numerous inflamma-
plantation. Osteocalcin is known to be involved in osteogenic
tory cells, such as plasma cells and WBCs, when compared with
differentiation and is secreted by osteoblasts after bone calcifica-
the other groups at 2 weeks. Some researchers have suggested that
tion starts [27]. Osteocalcin is also associated with the turnover
T. Eda et al. / Journal of Oral and Maxillofacial Surgery, Medicine, and Pathology 29 (2017) 563–569 569

platelets play a role in the recruitment of inflammatory cells such as collateral ligament of the knee. Knee Surg Sports Traumatol Arthrosc
neutrophils and monocytes [4,38], and platelets themselves act as 2013;21:1763–9.
[14] Dunbar JC, Reinholt L, Henry RL, Mammen E. Platelet aggregation and
innate inflammatory cells with acute host defense function [4,38]. disaggregation in the streptozotocin induced diabetic rat: the effect of
Furthermore, a significant increase in IL-1␤ was observed from PRP sympathetic inhibition. Diabetes Res Clin Pract 1990;9:265–72.
when the concentration of WBCs was large, thus suggesting that [15] Nagata M, Messora M, Okamoto R, Campos N, Pola N, Esper L, et al. Influence
of the proportion of particulate autogenous bone graft/platelet-rich plasma
IL-1␤ synthesis from PRP is related to WBCs [39,40]. In comparison on bone healing in critical-size defects: an immunohistochemical analysis in
with PRGF and PRP, PRP was considered to induce inflammatory rat calvalia. Bone 2009;45:339–45.
reactions based on the relationship with WBCs, platelet number, [16] Iwai S, Kuyama K, Kuboyama N, Takiguchi S, Ogura N, Yamamoto H, et al.
Osteogenic potential of human dental follicle cells on rat calvaria. J Hard
and cytokines released, and it is suggested that PRP is undesirable
Tissue Biol 2013;22:95–104.
for treatment of bone regeneration therapy for these reason. [17] Takiguchi S, Kuboyama N, Kuyama K, Yamamoto H, Kondoh T. Experimental
study of bone formation ability with the periosteum on rat calvalia. J Hard
Tissue Biol 2009;18:149–60.
5. Conclusions
[18] Eda T, Takahashi K, Iwai S, Ogura N, Ito K, Tsukahara H, et al. Effects of plasma
rich in growth factors on bone formation in rat calvaria. J Hard Tissue Biol
In conclusion, microCT and histological observation demon- 2015;24:61–8.
[19] Suemitsu M, Utunomiya T, Yamamoto H. Histopathological image analysis
strated that PRGF enhanced new bone formation from early time
using the imageJ-Digitization of a component ratio-. Clin Med Pathol
points compared to PRP. In addition, PRP delays new bone for- 2012;30:95–8.
mation due to the inflammatory response. Thus, it appears that [20] Zhao WJ, Duan F, Li ZT, Yang HJ, Huang Q, Wu KL. Evaluation of regional
PRGF can enhance bone regeneration and may be available in bone bulbar redness using an image-based objective method. Int J Ophthalmol
2014;18:71–6.
regeneration treatment. [21] Kwapiszewska G, Chwalek K, Marsh LM, Wygrecka M, Whilhelm J, Best J, et al.
BDNF/TrkB signaling augments smooth muscle cell proliferation in
Role of the funding source pulmonary hypertension. Am J Pathol 2012;181:2018–29.
[22] Shirakabe K, Terasawa K, Miyama K, Shibuya H, Nishida E. Regulation of the
activity of the transcription factor Runx2 by two homeobox proteins, Msx2
No funding was received for this study. and Dlx5. Genes Cells 2001;6:851–6.
[23] Ryoo HM, Hoffmann HM, Beumer T, Frenkel B, Towler DA, Stein GS, et al.
Stage-specific expression of Dlx-5 during osteoblast differentiation:
Acknowledgment involvement in regulation of osteocalcin gene expression. Mol Endocrinol
1997;11:1681–94.
The authors have no conflicts of interest to declare. [24] Nakashima K, Zhou X, Kunkel G, Zhang Z, Deng JM, Behringer RR, et al. The
novel zinc finger-containing transcription factor osterix is required for
osteoblast differentiation and bone formation. Cell 2002;108:17–29.
References [25] Komori T. Regulation of skeletal development by the Runx family of
transcription factors. J Cell Biochem 2005;95:445–53.
[1] Park SY, Kim KH, Gwak EH, Rhee SH, Lee JC, Shin SY, et al. Ex vivo bone [26] Takahashi K, Ogura N, Aonuma H, Ito K, Ishigami D, Kamino Y, et al. Bone
morphogenetic protein 2 gene delivery using periodontal ligament stem cells morphogenetic protein 6 stimulates mineralization in human dental follicle
for enhanced re-osseointegration in the regenerative treatment of cells without dexamethasone. Arch Oral Biol 2013;58:690–8.
peri-implantitis. J Biomed Mater Res A 2014;103:38–47. [27] Tanaka S, Matsuzaka K, Sato D, Inoue T. Characteristics of newly formed bone
[2] Marx RE, Carlson ER, Eichstaedt RM, Schimmele SR, Strauss JE, Georgeff KR. during guided bone regeneration: analysis of cbfa-1, osteocalcin, and VEGF
Platelet-rich plasma: growth factor enhancement for bone grafts. Oral Surg expression. J Oral Implantol 2007;33:321–6.
Oral Med Oral Pathol Oral Radiol Endod 1998;85:638–46. [28] Hawthorne AC, Xavier SP, Okamoto R, Salvador SL, Antunes AA, Salata LA.
[3] Dohan Ehrenfest DM, Bielecki T, Del Corso M, Inchingolo F, Sammartino G. Immunohistochemical, tomographic, and histological study on onlay bone
Shedding light in the controversial terminology for platelet-rich products: graft remodeling. Part III: allografts. Clin Oral Implants Res 2013;24:1164–72.
platelet-rich plasma (PRP), platelet-rich fibrin (PRF), platelet-leukocyte gel [29] Ducy P, Desbois C, Boyce B, Pinero G, Story B, Dunstan C, et al. Increased bone
(PLG), preparation rich in growth factors (PRGF), classification and formation in osteocalsin-deficient mice. Nature 1996;382:448–52.
commercialism. J Biomed Mater Res A 2010;95:1280–2. [30] Hering S, Isken E, Knabbe C, Janott J, Jost C, Pommer A, et al. TGFbeta1 and
[4] Malhotra A, Pelletier MH, Yu Y, Walsh WR. Can platelet-rich plasma (PRP) TGFbeta2 mRNA and protein expression in human bone samples. Exp Clin
improve bone healing: a comparison between the theory and experimental Endocrinol Diabetes 2001;109:217–26.
outcomes. Arch Orthop Trauma Surg 2013;133:153–65. [31] Vitsky A, Warie J, Pawliuk R, Bond A, Matthews D, Lacasse E, et al.
[5] Mark RE, Garg AK. The science of platelet-rich plasma. In: Lisa CB, editor. Homeostatic role of transforming growth factor- beta in the oral cavity and
Dental and craniofacial applications of platelet-rich plasma. Chicago: esophagus of mice and its expression by mast cells in these tissues. Am J
Quintessence Publishing; 2005. p. 33–9. Pathol 2009;174:2137–49.
[6] Aghaloo TL, Moy PK, Freymiller EG. Investigation of platelet-rich plasma in [32] Letterio JJ, Roberts AB. Regulation of immune respnses by TGF- beta. Annu
rabbit cranial defects: a pilot study. J Oral Maxillofac Surg 2002;60:1176–81. Rev Immunol 1998;16:137–61.
[7] Choi BH, Im CJ, Huh JY, Suh JJ, Lee SH. Effect of platelet-rich plasma on bone [33] Bensamoun SF, Hawse JR, Subramaniam M, I.Iharreborde B, Bassillais A,
regeneration in autogenous bone graft. Int J Oral Maxillofac Surg Benhamou CL, et al. TGFbeta inducible early gene-1 knockout mice display
2004;33:56–9. defects in bone strength and microarchitecture. Bone 2006;39:1244–51.
[8] Gerard D, Carlson ER, Gotcher JE, Jacobs M. Effects of platelet-rich plasma on [34] Andrew JG, Hoyland JA, Freemont AJ, Marsh DR. Platelet-derived growth
the healing of autologous bone grafted mandibular defects in dogs. J Oral factor expression in normally healing human fractures. Bone 1995;16:455–60.
Maxillofac Surg 2006;64:443–51. [35] Weiss S, Zimmermann G, Pufe T, Varoga D, Henle P. The systemic angiogenic
[9] Anitua E, Tejero R, Zalduendo MM, Orive G. Plasma rich in growth factors response during bone healing. Arch Orthop Trauma Surg 2009;129:989–97.
promotes bone tissue regeneration by stimulating proliferation, migration, [36] Xian L, Wu X, Pang L, Lou M, Rosen CJ, Qiu T, et al. Matrix IGF-1 maintains
and autocrine secretion in primary human osteoblasts. J Periodontol bone mass by activation of mTOR in mesenchymal stem cell. Nat Med
2013;84:1180–90. 2012;18, 1095–01.
[10] Anitua E, Troya M, Orive G. An autologous platelet-rich plasma stimulates [37] Weiss S, Henle P, Bidlingmaier M, Moghaddam A, Kasten P, Zimmermann G.
periodontal ligament regeneration. J Periodontol 2013;84:1556–66. Systemic response of the GH/IGF-I axis in timely versus delayed fracture
[11] Anitua E. Plasma rich in growth factors: preliminary results of use in the healing. Growth Horm IGF Res 2008;18:205–12.
preparation of future sites for implants. Int J Oral Maxillofac Implants [38] Flad HD, Brandt E. Platelet-derived chemokines: pathophysiology and
1999;14:529–35. therapeutic aspects. Cell Mol Life Sci 2010;67:2363–86.
[12] Anitua E, Begoňa L, Orive G. Controlled ridge expansion using a two-stage [39] Fréchette JP, Martineau I, Gagnon G. Platelet-rich plasmas: growth factor
split-crest technique with ultrasonic bone surgery. Implant Dent content and roles in wound healing. J Dent Res 2005;84:434–9.
2012;21:163–70. [40] Hartwig D, Härtel C, Henning H, Müller-Steinhardt M, Schlenke P, Klüter H.
[13] Yoshioka T, Kanamori A, Washio T, Aoto K, Uemura K, Sakane M, et al. The Evidence for de novo synthesis of cytokines and chemokines in platelet
effects of plasma rich in growth factors (PRGF-Endoret) on healing of medial concentrates. Vox Sang 2002;82:182–90.

You might also like