Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Leukocyte Inclusion within a Platelet Rich Plasma-Derived Fibrin Scaffold Stimulates a More Pro-Inflammatory Environment and Alters Fibrin Properties

  • Eduardo Anitua,

    Affiliations Foundation Eduardo Anitua, Vitoria, Spain, Laboratory of Regenerative Medicine, Biotechnology Institute (BTI), Vitoria, Spain

  • Mar Zalduendo,

    Affiliation Laboratory of Regenerative Medicine, Biotechnology Institute (BTI), Vitoria, Spain

  • María Troya,

    Affiliation Laboratory of Regenerative Medicine, Biotechnology Institute (BTI), Vitoria, Spain

  • Sabino Padilla,

    Affiliation Laboratory of Regenerative Medicine, Biotechnology Institute (BTI), Vitoria, Spain

  • Gorka Orive

    gorka.orive@ehu.es

    Affiliation Foundation Eduardo Anitua, Vitoria, Spain

Abstract

One of the main differences among platelet-rich plasma (PRP) products is the inclusion of leukocytes that may affect the biological efficacy of these autologous preparations. The purpose of this study was to evaluate whether the addition of leukocytes modified the morphological, biomechanical and biological properties of PRP under normal and inflammatory conditions. The release of pro-inflammatory cytokines from plasma rich in growth factors (PRGF) and leukocyte-platelet rich plasma (L-PRP) scaffolds was determined by enzyme-linked immunosorbent assay (ELISA) and was significantly increased under an inflammatory condition when leukocytes were included in the PRP. Fibroblasts and osteoblasts treated with L-PRP, under an inflammatory situation, underwent a greater activation of NFĸB pathway, proliferated significantly less and secreted a higher concentration of pro-inflammatory cytokines. These cellular events were assessed through Western blot and fluorimetric and ELISA methods, respectively. Therefore, the inclusion of leukocytes induced significantly higher pro-inflammatory conditions.

Introduction

Over the last few years, translational medicine has emerged as a new trend in medical practice, which aims to promote the rapid clinical translation of a wide range of therapeutic options and to receive the feedback of basic research results [1]. One of the hottest topics in translation medicine deals with those therapies focused on enhancing and promoting tissue repair and regeneration [2]. Repair, restoration and healing of lost tissue are unique among clinical treatments due to the large number of patients suffering from tissue damage and injuries. Progress in this field is pertinent as it may help to reduce the burden on the world’s health systems and address the need of tissue replacement [3].

The use of patient-derived formulations and therapeutics is gaining the attention of the scientific community. Human plasma and especially human platelets contain a wide range of proteins and growth factors that have been demonstrated to promote tissue repair and regeneration in many different injured tissues [414]. In addition, the coagulation of plasma leads to the formation of a three-dimensional fibrin matrix that can provide transient space for the key tissue-forming cells at the same time that acts as a controlled protein delivery system [2, 13]. These and other advances have promoted the research and use of platelet rich plasma products as a biomolecules delivering system [15] for wound healing, tissue regeneration and tissue engineering [16].

There are however, several concerns related to the use of platelet rich plasma products that need to be addressed. First, how these preparations exert their effects in inflammatory conditions. To our knowledge, most of the studies evaluating the biological potential of platelet rich plasma products do not mimic tissue inflammatory conditions in which most of these therapies are used. Second, how the particular protein and cell composition of these products can affect their final therapeutic outcomes. In fact, while some platelet rich plasma products contain only platelet-enriched plasma, others concentrate leukocytes, red cells and other pro-inflammatory cells. As a consequence, the composition and biological properties of these products might be potentially different. It has been demonstrated that leukocyte concentration is negatively correlated with matrix gene expression and positively correlated with catabolic gene expression in tendon and ligament tissues [17]. Moreover, white blood cells (WBC) contain numerous pro-inflammatory interleukins and extracellular matrix degrading enzymes [18]. Third, only few studies have determined the role and properties of the fibrin scaffold formed with each type of platelet rich plasma product.

In the present study, we have carried out a complete evaluation of how including leukocytes (L-PRP) or not in plasma rich in growth factors (PRGF), a well-characterized platelet rich plasma therapy, may alter both protein and cytokine release and fibrin scaffold properties. Leukocyte containing and leukocyte free PRGFs were prepared and evaluated in non-inflammatory and inflammatory conditions that mimic a well-known chronic inflammatory disease such as periodontitis. The latter is caused by Porphyromonas gingivalis [19] and affects periodontal tissue due to the release of proteolytic enzymes [20, 21].

Materials and Methods

The study was performed following the principles of the Declaration of Helsinki, as revised in 2008 and after approval from the Foundation Eduardo Anitua Institutional Review Board.

Preparation of PRP (PRGF) products

Blood from a total of 6 patients was collected into 9-mL tubes with 3.8% (wt/v) sodium citrate, after written informed consent was provided (approved by our Institutional Review Board). The following products were generated immediately from the collected blood: plasma rich in growth factors (PRGF) and Leukocyte-PRP (L-PRP). PRGF was prepared according to manufacturer’s instructions (Endoret Dentistry, BTI Biotechnology Institute, S.L., Miñano, Álava, Spain). Briefly, samples were centrifuged at 580 g for 8 min at room temperature. For each tube, the 2 ml of plasma just above the buffy coat was collected in each donor. Leukocyte-PRP was prepared by the addition of the buffy coat into the platelet richest fraction. Platelets and leukocytes counts were performed on samples of blood, PRGF and L-PRP with a hematology analyzer (Micros 60, Horiba ABX, Montpelier, France).

Structure of fibrin net from both PRP products

The different fibrin scaffolds were prepared by activating different volumes of PRGF and L-PRP with calcium chloride (Endoret Dentistry) at 37°C for 1 hour (50 μL of calcium chloride for each mL of PRGF/L-PRP). After clot formation, the morphological analysis of PRGF and L-PRP fibrin scaffolds was performed with scanning electron microscopy (SEM). Briefly, the clots were rinsed with phosphate-buffered saline and fixed with 2% glutaraldehyde in 0.1 M cacodylate buffer for 4 h and washed in cacodylate-sucrose buffer. Then, the samples were post-fixed with osmium tetroxide for 1 h, washed again and dehydrated with serial concentrations of ethanol. Samples were critical point-dried (Tousimis Autosamdri 814), sputter-coated with 5 nm gold (Edwards E306A) and subsequently examined in a scanning electron microscope (Hitachi S-4800) [22].

Mechanical properties of PRGF and L-PRP fibrin scaffolds

The mechanical properties of PRGF and L-PRP fibrin scaffolds were evaluated. Two mL of each formulation and 100 μL of calcium chloride were added to the devices. Once the clot was formed, the devices were held on the mechanical testing station (858 Mini Bionix II, MTS, USA) with a loading cell of 10N, and de-moulded, leaving the fibrins ready to perform the assays. The mechanical testing station stretched the fibrin scaffolds at a constant speed of 2mm/s. Maximum elongation at failure was determined.

Culture of PRGF and L-PRP on inflammatory and non-inflammatory conditions

Two mL of PRGF or L-PRP of each of the six donors were added to each well of a 6-well cell culture plate and incubated with 100 μL of calcium chloride for 20–30 min at 37°C until clot formation. Thereafter, half of the scaffolds of both PRGF and L-PRP were incubated with 2 mL of Dulbecco’s modified Eagle’s medium (DMEM)/F12 (Gibco-Invitrogen, Grand Island, NY, USA) in the absence (normal condition) or presence (inflammatory condition) of 10 μg/ml of LPS from Porphyromonas gingivalis (P.g.-derived LPS) (InvivoGen, San Diego, CA, USA) at 37°C with 5% CO2 for 72h. The remaining scaffolds were incubated with 2 mL of osteoblast basal medium (ObM) (Sciencell Research Laboratories, Carlsbad, CA, USA), also under both normal and inflammatory conditions. After that, the conditioned media by the scaffolds was collected and centrifuged at 460 g for 10 min at room temperature. The supernatant was stored at -80°C for assay of protein levels.

Quantification of cytokines and growth factors by ELISA

The levels of interleukin 1 beta (IL-1β), interleukin 8 (IL-8) (Invitrogen), tumor necrosis factor-α (TNF-α, interleukin 6 (IL-6), vascular endothelial growth factor (VEGF), platelet-derived growth factor AB (PDGF-AB) and angiopoietin-1 (R&D Systems, Minneapolis, MN, USA) on the conditioned media of both PRGF and L-PRP clots under both normal and inflammatory conditions, were determined by enzyme-linked immunosorbent assay (ELISA), according to the manufacturer’s protocol.

MMP-1 activity assay

In order to determine the MMP-1 activity on the conditioned media of both PRGF and L-PRP clots under both study conditions, a MMP-1 colorimetric drug discovery kit (Enzo Life Sciences, Lausen, Switzerland) was used with some modifications to the manufacturer’s protocol. A standard curve for MMP-1 was performed. The enzyme activity was determined 1 minute after the reaction was started.

Cell treatment with PRGF and L-PRP

Primary human gingival fibroblasts and primary human alveolar osteoblasts, obtained from patients during routine surgical procedures and dental implant surgery, respectively, were isolated as previously described [23, 24] and were used to assess the effect of both PRGF and L-PRP releasates under inflammatory conditions. Written informed consent from those patients and approval by the Foundation Eduardo Anitua institutional review board were obtained.

Fibroblasts were routinely cultured in DMEM/F12 supplemented with 2 mM glutamine and 50 μg/mL gentamicin (Sigma-Aldrich, St. Louis, MO). Osteoblasts were cultured in ObM supplemented with 50 μg/mL gentamicin. Both cultured media were supplemented with 15% fetal bovine serum FBS (Biochrom AG, Leonorenstr, Berlin, Germany).

PRGF and L-PRP scaffolds from one donor were stimulated with 10 μg/mL of P.g.-derived LPS for 72 h, as previously described (Fig 1). The molecule releasates from the scaffolds incubated with LPS will be identified as PRGF+LPS and L-PRP+LPS.

thumbnail
Fig 1. Illustrative representation of the cell treatment experimental procedure with PRGF and L-PRP releasates under inflammatory conditions.

https://doi.org/10.1371/journal.pone.0121713.g001

The cell incubation experiments were performed as follows: Cells were pretreated with 10 μg/ml of P.g.-derived LPS in the appropriate culture medium for 72 hours. The pretreated cells were then cultured with the corresponding culture medium and 10 μg/ml of P.g.-derived LPS supplemented with either 70% PRGF+LPS or 70% of L-PRP+LPS. The treatment time was 24 h for Western blot analysis, 72 h for proliferation assay, and 24h and 72h for cytokine secretion assay.

Western blot analysis

After treatment, both fibroblasts and osteoblasts were washed with PBS and lysed with mammalian protein extraction reagent supplemented with protease and phosphatase inhibitors (Pierce Biotechnology, Bonn, Germany). Lysates were clarified by centrifugation at 14000 g for 10 min, and the supernatants were then collected. Cell lysates were concentrated using centrifugal filters (Amicon ultra-0.5 (3k), Chemicon-Millipore, Billerica, MA, USA) and the protein concentration was determined with the BCA assay (Pierce Biotechnology). TGX stain-free gels were used for protein electrophoresis (Bio-Rad Laboratories, Munich, Germany). The membranes were blocked using 5% non-fat dry milk in tris (hydroxymethyl) aminomethane-buffered saline containing 0.1% polysorbate surfactant (TBST) for 1 hour at room temperature. Then the membranes were incubated overnight at 4°C with the corresponding primary antibodies, nuclear factor-ĸB p65 (NF-ĸB p65), phosphorylated nuclear factor-ĸB p65 (pNF-ĸB p65) and inhibitor-ĸBα (IĸBα) (Cell signaling Technology, USA). Membranes were then washed several times with TBST and incubated with horseradish peroxidase-conjugated goat anti-rabbit and anti-mouse secondary antibodies (Cell signaling Technology) accordingly, for 1 hour. The blots were washed again and developed by chemiluminescence with substrate Immun-Star HRP substrate, (Bio-Rad Laboratories) using an image analyzer (Chemidoc image analyzer, Bio-Rad Laboratories). The Stain-Free technology was used as loading control method [25].

Cell Proliferation assay

Cell proliferation was evaluated by Cyquant cell proliferation assay (Molecular Probes-Invitrogen, Grand Island, NY, USA). Briefly, the treatments were removed and the wells were washed carefully with phosphate-buffered saline (PBS). Then, the microplates were frozen at -80°C until assayed, to allow cell lysis. After thawing the plates at room temperature, wells were incubated with RNase A (1.35 Ku/ml) diluted in cell lysis buffer for 1 hour at room temperature. Then, 2x GR dye/cell-lysis buffer was added to each sample well, mixed gently and incubated for 5 minutes at room temperature, protected from light. Sample fluorescence was measured with a fluorescence microplate reader (Twinkle LB 970, Berthold Technologies, Bad Wildbad, Germany). A DNA standard curve was included in each assay.

Cell-stimulated cytokine expression

ELISA was performed to analyze the concentration of pro-inflammatory cytokines secreted by human gingival fibroblasts and alveolar osteoblasts after treatment with the conditioned media from both PRGF and L-PRP. Briefly, the conditioned media was collected and levels of IL-1β, TNF-α IL-6 and IL-8 were measured by ELISA kits (R&D Systems). The concentrations of these proteins were determined according to the manufacturer’s instructions.

Statistical Analysis

The results were presented as mean ± standard deviations. Single-factor analysis of variance (ANOVA) and Kruskal-Wallis test for independent samples was used, as appropriate, to test the differences among groups. P < 0.05 was considered to indicate statistical significance.

Results

Composition and structure of fibrin scaffolds from PRGF and L-PRP

Cell content of PRGF and L-PRP preparations was measured (Fig 2A). No statistically significant differences were found in platelet counting. However, leukocyte concentration in L-PRP was 3.1 ± 0.3-fold higher than in blood while almost no white cell was detected in PRGF (0.2 ± 0.1 x103/μl) (Fig 2B).

thumbnail
Fig 2. Composition and structure of fibrin net from both PRP products.

(A) Concentration of platelets in PRGF and L-PRP preparations. (B) Leukocyte content in PRGF and L-PRP preparations. (C) Macroscopic appearance and ultraestructural composition of PRGF and L-PRP scaffolds.

https://doi.org/10.1371/journal.pone.0121713.g002

The fibrin scaffolds obtained with each type of platelet rich plasma showed different appearance and consistency when handled. PRGF fibrin scaffold was yellowish and easier to handle than the reddish one prepared from L-PRP (Fig 2C). In fact, the latter was easily broken after manipulation. Ultra structural analysis of both fibrin scaffolds showed the alteration of the fibrillar cross-linking due to the presence of cellular elements including leukocytes and erythrocytes in the L-PRP (Fig 2C).

Characterization of the PRGF and L-PRP-scaffold releasates

The different fibrin scaffolds were cultured at 37°C either in non-inflammatory (only culture medium: normal conditions) or inflammatory (LPS-containing) conditions. After 72 hours, several growth factors (GFs) involved in different phases of the regeneration process and the most relevant interleukins and pro-inflammatory molecules were measured. Under normal conditions, the IL-1β concentration in the PRGF releasate was 0.0 ± 0.4 pg/ml while it was higher in the L-PRP releasate (13 ± 8.5 pg/mL). However, under inflammatory conditions, these difference increased drastically, being the IL-1β concentration in the PRGF releasate of 111 ± 87 pg/mL and 22390 ± 16745 pg/mL in the L-PRP supernatant (Fig 3A). The presence of TNF-α and IL-6 cytokines in the PRGF and L-PRP releasates was almost non-detectable in normal conditions. However, once again, those levels were significantly higher in the case of L-PRP releasate under inflammatory conditions. TNF-α and IL-6 levels in PRGF were 66 ± 74 pg/mL and 896 ± 1131 pg/mL respectively, while in the case of L-PRP they were 1481 ± 835 pg/mL and 222100 ± 72878 pg/mL respectively (Fig 3B and 3C). Furthermore, a statistically significant higher IL-8 delivery from L-PRP scaffolds was measured in both analyzed conditions with respect to the PRGF scaffold releasates (129 ± 139 pg/mL vs 3535 ± 1791 pg/mL and 5415 ± 5207 pg/ml vs 2065000 ± 1067263 pg/ml, for PRGF and L-PRP under normal and inflammatory conditions, respectively) (Fig 3D).

thumbnail
Fig 3. Characterization of the PRP scaffold releasates.

Concentration of different cytokines, (A) IL1-beta, (B) TNF-alpha, (C) IL-6 and (D) IL-8 in the PRGF and L-PRP releasate under both normal and inflammatory conditions. *Statistically significant differences respect to the other treatments (p < 0.05). $Statistically significant differences respect to the PRGF treatment under normal conditions (p < 0.05).

https://doi.org/10.1371/journal.pone.0121713.g003

Release of PDGF and two additional proteins with important roles in angiogenesis, Angiopoietin-1 and VEGF, were analyzed in the different releasates. In the case of PDGF-AB and angiopoietin-1, no statistically significant differences were found between PRGF and L-PRP releasates either in normal or inflammatory condition (Fig 4A and 4C). Nevertheless, VEGF delivery from L-PRP scaffold was significantly higher than the PRGF one, but only in the absence of inflammation (88 ± 82 pg/ml vs 502 ± 432 pg/ml). Interestingly, under inflammatory conditions, no VEGF was detectable in the L-PRP releasate whereas VEGF levels were maintained in the case of PRGF releasates (Fig 4B).

thumbnail
Fig 4. Characterization of the PRP scaffold releasate.

Determination of different proteins concentration. (A) Angiopoietin-1 (B) VEGF and (C) PDGF-AB in the PRGF and L-PRP releasate under both normal and inflammatory conditions. *Statistically significant differences respect to the other treatments (p < 0.05).

https://doi.org/10.1371/journal.pone.0121713.g004

Analysis of the integrity and the mechanical properties of the fibrin scaffolds

PRGF and L-PRP-derived fibrin scaffolds from each of the six donors cultured with culture medium were maintained in the absence (normal condition) or presence (inflammatory condition) of LPS for 72h. PRGF fibrin scaffold preserved its stability under both normal and inflammatory conditions, while in the case of L-PRP, the fibrin scaffold were altered due to the presence of LPS (Fig 5A). In fact, half of the L-PRP fibrin scaffolds which were incubated with LPS appeared highly deteriorated after 72 hour of treatment, as it is shown in Fig 5A.

thumbnail
Fig 5. Analysis of the integrity and the mechanical properties of PRP scaffolds.

(A) Comparison of PRP scaffolds appearance after 72 hours incubations under normal and inflammatory conditions. In half of the L-PRP matrix a great degradation was observed. (B) MMP-1 activity in PRGF and L-PRP releasates was determined under both normal and inflammatory conditions. (One U = 100 pmol/min at 37°C, 100 μM thiopeptolide). (C) Mechanical testing station where the maximum elongation at failure of the PRP scaffolds was determined. (D) Maximum elongation of PRP scaffolds. *Statistically significant differences between L-PRP and PRGF under normal conditions and between L-PRP and PRGF under inflammatory conditions (p < 0.05).# Statistically significant differences respect to the PRGF treatment under normal conditions (p < 0.05).

https://doi.org/10.1371/journal.pone.0121713.g005

MMP-1 activity was measured in the different releasates under normal and inflammatory conditions. A statistically significant higher enzyme activity was detected in the L-PRP scaffold conditioned medium under both study conditions (586 ± 128 mU/μl respect to 1168 ± 401 mU/μl for PRGF- and L-PRP-releasates under normal conditions and 745 ± 96 mU/μl respect to 1315 ± 257 mU/μl for PRGF- and L-PRP-releasates under inflammatory conditions). The detected enzyme activity in the L-PRP-releasates under inflammatory conditions was also statistically superior than the one detected in the PRGF-releasates under normal conditions (Fig 5B).

In addition, the mechanical properties of the different fibrin scaffolds were determined. For this purpose, the maximum elongation at failure was determined after stretching fibrins at a constant speed of 2mm/s. The maximum elongation achieved with the PRGF scaffolds was higher than with L-PRP ones (6.1 ± 1.7 cm vs 4.4 ± 1.3 cm) (Fig 5C and 5D).

Evaluation of cell inflammatory NFĸB pathway

Primary human gingival fibroblasts and alveolar osteoblasts were treated with conditioned media by PRGF and L-PRP scaffolds under inflammatory conditions for 24 hours. After extracting and concentrating the protein content of the treated cells, IĸB-α, p-NFĸB and NFĸB synthesis were quantified by western blotting technique. In both of the two phenotypes included in the study, a decrease in IĸB-α expression was found when cells were incubated with the L-PRP releasate compared with the ones incubated with PRGF releasates (5.1 ± 0.0 vs 2.9 ± 0.4 and 15.6 ± 2.1 vs 13.7 ± 2.3 for PRGF and L-PRP in gingival and bone cells, respectively) (Fig 6A and 6B). On the contrary, a statistically significant increase in the p-NFĸB/NFĸB ratio was observed when cells were treated with L-PRP+LPS releasate when compared with PRGF+LPS treatment (2.1 ± 0.7 vs 10.0 ± 2.0 and 1.0 ± 0.1 vs 1.5 ± 0.3 for PRGF and L-PRP and in gingival and osteoblast cells respectively) (Fig 6C and 6D).

thumbnail
Fig 6. Analysis of the inflammatory mediators’ expression by western blot.

Cells were treated with PRGF+LPS and L-PRP+LPS releasates for 24 hours under inflammatory conditions and IĸB-α and p-NFĸB/NFĸB ratio were determined. (A) IĸB-α expression in gingival fibroblasts. (B) IĸB-α expression in alveolar osteoblasts. (C) p-NFĸB/NFĸB determination in gingival fibroblasts. (D) p-NFĸB/NFĸB determination in alveolar osteoblasts. *Statistically significant differences respect to the PRGF+LPS treatment (p < 0.05).

https://doi.org/10.1371/journal.pone.0121713.g006

Cell proliferation assay

Fibroblasts and osteoblasts were incubated with PRGF+LPS and L-PRP+LPS releasates for 72 hours and in LPS presence. Some morphological changes were observed after the treatment with L-PRP+LPS scaffold releasate, especially in the case of gingival fibroblasts. As it shown in Fig 7A, cells laid out in clusters and became brighter and with a thinner cell body. In addition, a statistically significant decrease in DNA concentration was detected in gingival fibroblast and osteoblast cultures incubated with L-PRP+LPS conditioned medium under inflammatory conditions compared to the ones detected after PRGF+LPS releasate treatment (84 ± 20 ng/ml vs 39 ± 11 ng/ml and 72 ± 10 ng/ml vs 60 ± 4 ng/ml for PRGF and L-PRP in gingival and bone cell cultures, respectively) (Fig 7B and 7C).

thumbnail
Fig 7. Effect of the releasates of PRGF+LPS and L-PRP+LPS on the proliferation of fibroblasts and osteoblast cells.

(A) Morphological alterations in cultures due to 72 hour treatment with PRP releasates under inflammatory conditions. Scale bar: 300 μm. (B) Gingival fibroblast proliferation after treatment. (C) DNA quantification of alveolar osteoblast proliferation after treatment. *Statistically significant differences respect to the L-PRP+LPS treatment (p < 0.05).

https://doi.org/10.1371/journal.pone.0121713.g007

Cell-derived cytokine expression

After treating gingival and bone cells for 24 and 72 hours with PRGF+LPS and L-PRP+LPS, the conditioned media was collected and the levels of IL-1β, TNF-α, IL-6 and IL-8 were measured. The concentration of all the pro-inflammatory cytokines was significantly enhanced after treating the cells with L-PRP+LPS releasate (Fig 8).

thumbnail
Fig 8. Pro-inflammatory cytokine synthesis due to cell treatment with PRP releasates.

Gingival and bone cells were incubated with PRGF+LPS and L-PRP+LPS releasates under inflammatory conditions for 24 and 72 hours. IL-1β, TNF-α, IL-6 and IL-8 synthesis was determined by ELISA techniques. (A) Treatment of gingival fibroblast during 24 hours. (B) Gingival fibroblast response to 72 hour treatment. (C) Treatment of alveolar osteoblasts during 24 hours. (D) Bone cells response to 72 hour treatment. *Statistically significant differences respect to the PRGF+LPS treatment (p < 0.05).

https://doi.org/10.1371/journal.pone.0121713.g008

As it is shown in Fig 8A (24 h treatment), gingival fibroblasts stimulated with PRGF released 6 ± 2 pg/mL of IL-1β while they released up to 41492 ± 3629 pg/mL when stimulated with L-PRP. Gingival cell expression of TNF-α was also significantly increased with L-PRP (500 ± 36 pg/mL) compared to PRGF (0 ± 3 pg/mL). A similar trend was observed for IL-6 and IL-8 expression. Cell culture with L-PRP significantly increased the amount of the proteins (244833 ± 87279 pg/mL for IL-6 and 1583333 ± 111430 pg/mL for IL-8) compared to those obtained with PRGF (80992 ± 23878 pg/mL and 110492 ± 7569 pg/mL for IL-6 and IL-8, respectively). A similar trend was observed after treating the cells for 72 h with PRGF or L-PRP (Fig 8B).

In a second set of experiments, the effects of the conditioned media on primary osteoblasts cells were evaluated (Fig 8C and 8D). Culturing the cells with the L-PRP media provoked the dramatic increase of the expression of the four cytokines. Interestingly, the use of a leukocyte containing PRP enhanced osteoblasts-derived release of IL-1β more than 700 times (Fig 8C). This striking increase in the synthesis of pro-inflammatory cytokines due to the leukocyte inclusion in the PRP-scaffold was also observed for the remaining three proteins. Osteoblast cell expressions of TNF-α (1152 ± 53 pg/mL), IL-6 (154333 ± 52495 pg/mL) and IL-8 (1259667 ± 124582 pg/mL) were significantly higher than those observed after PRGF treatment TNF-α (-3 ± 9 pg/mL), IL-6 (40050 ± 10300 pg/mL) and IL-8 (41056 ± 14340 pg/mL). A similar trend was observed after treating the cells for 72 h with PRGF or L-PRP (Fig 8D).

Discussion

Periodontal disease is the most prevalent chronic inflammatory condition in humans. It is characterized by the destruction of the tooth-supporting structures, namely, soft tissue, alveolar bone, periodontal ligament and cementum [2628]. Porphyromonas gingivalis is a gram-negative anaerobic bacterium that is considered to be the major etiologic agent of periodontitis. Lipopolysaccharide, a major component of the outer membrane of the gram-negative bacteria, is known to be the major virulent factor in periodontitis, as it is a potent stimulator of inflammatory mediators [26, 29, 30].

In the last few decades, there is growing interest in developing new cost-effective therapeutic alternatives for tissue repair and regeneration. Assuming the scarcity of human organs and tissues, there is interest in promoting new approaches that help to restore tissue homeostasis after inflammation and injury. The field of platelet rich plasma, that is, the use of patient’s own proteins, growth factors and biomaterials is gaining much interest not only due to its autologous origin, but also because it is a not expensive approach with a clear translation pathway to the clinics.

In the present paper, we have addressed one of the most important concerns about PRPs; how the composition of leukocytes may impact on the final biological and mechanical properties, which are key for final therapeutic outcomes of the approach. In addition, to our knowledge this is one of the few studies that investigates the effect of PRP composition under normal and inflammatory conditions.

According to our results, the different composition of PRPs containing leukocytes affects negatively the mechanical properties of their fibrin scaffolds and stimulates a more pro-inflammatory environment that is directly related with an increased cell-inflammatory condition and a reduced cell proliferation response. These effects may delay or impede a correct tissue repair or regeneration process.

The inclusion of leukocytes within platelet rich plasma products has always been a matter of debate. The deleterious effects of leukocytes have been widely described [3134]. In contrast to platelets, white blood cells are mainly considered to contain and produce inflammatory cytokines.[31, 32, 35, 36] The results obtained in the present study confirm that leukocyte free PRGF is more predictable as its properties (growth factor release and fibrin scaffold integrity) are almost preserved from non-inflammatory to inflammatory conditions [27, 37].

In our experiments, only the IL-8 released was found to be statistically significant different under normal conditions between PRGF and L-PRP scaffolds, being much higher in the case of L-PRP (Fig 3). However, under inflammatory conditions, the levels of IL-1β, IL-6, IL-8 and TNF-α released by L-PRP fibrin scaffolds were statistically much higher with respect to PRGF (Fig 3). Moreover, in the case of PRGF, no differences were found in protein release between normal or inflammatory conditions, except for IL-8.

Although cytokines play an important role in infection and inflammation, excessive cytokine expression may lead to tissue destruction [38, 39]. Inflammatory cytokines such as IL-6 and IL-8 are believed to be the main pathological mediators in periodontal diseases. IL-6 may activate osteoclasts, promoting bone loss and causing bone resorption. At the same time, this cytokine stimulates matrix metalloproteinases, therefore increasing matrix degradation. IL-8 in turn, may induce neutrophil chemotaxis and activation [4042]. Meanwhile, TNF-α and IL-1β have also a significant role in bone loss, playing a central role in inflammatory reaction [28, 43]. In fact, IL-1β is an essential pro-inflammatory cytokine that induces the infiltration of inflammatory cells [44].

Apart from the increased enzyme activity of MMP-1 in the L-PRP-releasates under both normal and inflammatory conditions, in the latter, half of the L-PRP fibrin scaffolds were degraded (Fig 5A and 5B). Matrix metalloproteinases (MMPs), a family of proteolytic enzymes, are responsible for degrading the main components of the extracellular matrix. MMP-1 is a collagenase that is involved in extracellular matrix breakdown during periodontitis. The production of this enzyme is stimulated by several growth factors and cytokines such as IL-1β, TNF-α and IL-6 [42, 45].

Several important differences were also observed after determining the release of growth factors from PRGF and L-PRP scaffolds. In normal conditions, the concentration of VEGF released by L-PRP scaffolds was higher than that released by PRGF scaffolds. However, under inflammatory conditions no VEGF was detected in the L-PRP fibrin-conditioned medium. The latter may be a limitation since VEGF is necessary in wound healing causing inflammation as it promotes the early events in angiogenesis [46, 47]. This finding could be explained by a possible VEGF uptake by its soluble receptor (sVEGFR) synthesized by leukocytes [48, 49]. Moreover, similar results were obtained in a previous publication where a VEGF concentration decrease was found in the conditioned medium of L-PRP scaffolds from the third day of assay [50]. Fibrin is the biological transient scaffold that it is firstly formed at injury’s sites and it is commonly used in tissue engineering. The mechanical properties and biodegradation of fibrin scaffolds are essential for their role as a provision matrix in tissue regeneration approaches and as a protein delivery system [2, 5153]. Viscoelastic properties largely depend on fibrin clot structure. Here, we have reported that the cell composition of L-PRP alters the clot structure and decreases the maximum elongation of the scaffold. Higher elongations indicate a greater elasticity which involves, more malleable, easier to handle and stronger fibrins to fulfill the required function as a biomaterial that can be used as a specific shape scaffold in tissue engineering [51]. PRGF scaffold has already been used in several applications, such as the treatment of ulcers, wound closure, tissue engineering or even combined with others materials [37].

Interestingly, LPS stimulates inflammatory cells, such as neutrophils, macrophages but also resident cells such as fibroblasts and osteoblasts. Therefore, cell responses to the treatment with both types of PRP under inflammatory conditions were also evaluated. Toll-like receptor 4 (TLR4) is the main receptor in the cellular response to LPS. Ligation of these receptors initiates a cascade of events that leads to the activation of transcription factors, including nuclear factor-ĸB (NF-ĸB), that eventually induce the production of pro-inflammatory cytokines [19, 30, 54]. Our findings revealed that the inhibitor-ĸBα (IĸBα expression was increased when gingival fibroblasts and alveolar osteoblasts were cultured with PRGF+LPS under inflammatory conditions (Fig 6). IĸBα is considered to have an anti-inflammatory role as it maintains the NF-ĸB in the cytosol in an inactivated state [55, 56]. These results are consistent with those observed for the p-NFĸB/NFĸB ratio. L-PRP+LPS induced a significantly higher phosphorylation of NF-ĸB p65 in both cell types (Fig 6). NF-ĸB p65 phosphorylation at Ser536 regulates nuclear localization, protein-protein interactions, activation of gene expression and transcriptional activity [56, 57]. Moreover, IL-1β and TNF-α are cytokines that activate the NFĸB pathway [55, 56]. Therefore, the pro-inflammatory environment generated by L-PRP scaffolds may be behind of this increase in the activation of NFĸB after cell treatment with L-PRP+LPS. This pro-inflammatory milieu and the greater activation of NFĸB after treating the cells with L-PRP+LPS had also a negative effect on cell proliferation. Both gingival fibroblasts and alveolar osteoblasts proliferated significantly less when treated with L-PRP+LPS than when treated with PRGF+LPS (Fig 7).

Finally, treatment with L-PRP+LPS also stimulated pro-inflammatory cytokine synthesis in both cell types compared with treatment with PRGF+LPS (Fig 8). This cell response was observed both at 24h and 72h. The excess of those pro-inflammatory proteins can lead to the destruction of the tissue, as previously described. This increase is related to the enhanced activation of NFĸB pathway after the treatment with L-PRP+LPS. In fact, NFĸB activation results in the production of those cytokines, which in turn, can amplify and increase the inflammatory response, thus keeping the NFĸB pathway active, establishing a positive feedback response. In the wake of this activation, the excessive pro-inflammatory cytokine release after L-PRP+LPS incubation may exacerbate the inflammatory response by stimulating the recruitment of new inflammatory cells to the injury site. This microenvironment may be conducive to a non-resolving inflammation that leads to a tissue fibrotic condition [58, 59].

Conclusions

We have shown that under inflammatory conditions, the different cell composition of L-PRP increases the delivery of pro-inflammatory cytokines, as well as stimulates the resident cells to produce a greater amount of the afore mentioned proteins. The NFĸB pathway is also stimulated in oral cells treated with L-PRP+LPS scaffold releasate under inflammatory conditions. The mechanical properties of the L-PRP scaffolds are poorer and the degradation of these fibrin meshes is promoted under inflammatory conditions. In summary, the composition of L-PRP negatively affects the mechanical properties of the fibrin scaffolds and stimulates a more pro-inflammatory environment that is directly related with an increased cell-inflammatory condition and a reduced cell proliferation response, which ultimately may be detrimental for tissue regeneration.

Author Contributions

Conceived and designed the experiments: EA GO. Performed the experiments: MZ MT. Analyzed the data: EA SP GO. Contributed reagents/materials/analysis tools: MZ MT. Wrote the paper: MZ MT SP.

References

  1. 1. Humes HD (2005) Translational medicine and the National Institutes of Health road map: steep grades and tortuous curves. J Lab Clin Med 146: 51–54. pmid:16099234
  2. 2. Anitua E, Sanchez M, Orive G (2010) Potential of endogenous regenerative technology for in situ regenerative medicine. Adv Drug Deliv Rev 62: 741–752. pmid:20102730
  3. 3. Ginty PJ, Rayment EA, Hourd P, Williams DJ (2011) Regenerative medicine, resource and regulation: lessons learned from the remedi project. Regen Med 6: 241–253. pmid:21391857
  4. 4. Anitua E, Alkhraisat MH, Orive G (2013) Novel technique for the treatment of the severely atrophied posterior mandible. Int J Oral Maxillofac Implants 28: 1338–1346. pmid:24066326
  5. 5. Crovetti G, Martinelli G, Issi M, Barone M, Guizzardi M, Campanati B, et al. (2004) Platelet gel for healing cutaneous chronic wounds. Transfus Apher Sci 30: 145–151. pmid:15062754
  6. 6. Albanese A, Licata ME, Polizzi B, Campisi G (2013) Platelet-rich plasma (PRP) in dental and oral surgery: from the wound healing to bone regeneration. Immun Ageing 10: 23. pmid:23763951
  7. 7. Del Corso M, Vervelle A, Simonpieri A, Jimbo R, Inchingolo F, Sammartino G, et al. (2012) Current knowledge and perspectives for the use of platelet-rich plasma (PRP) and platelet-rich fibrin (PRF) in oral and maxillofacial surgery part 1: Periodontal and dentoalveolar surgery. Curr Pharm Biotechnol 13: 1207–1230. pmid:21740371
  8. 8. Simonpieri A, Del Corso M, Vervelle A, Jimbo R, Inchingolo F, Sammartino G, et al. (2012) Current knowledge and perspectives for the use of platelet-rich plasma (PRP) and platelet-rich fibrin (PRF) in oral and maxillofacial surgery part 2: Bone graft, implant and reconstructive surgery. Curr Pharm Biotechnol 13: 1231–1256. pmid:21740370
  9. 9. Sanchez M, Anitua E, Azofra J, Andia I, Padilla S, Mujika I (2007) Comparison of surgically repaired Achilles tendon tears using platelet-rich fibrin matrices. Am J Sports Med 35: 245–251. pmid:17099241
  10. 10. Wroblewski AP, Mejia HA, Wright VJ (2010) Application of Platelet-Rich Plasma to Enhance Tissue Repair. Operative Techniques in Orthopaedics 20: 98–105.
  11. 11. Muto T, Kokubu T, Mifune Y, Sakata R, Nagura I, Nishimoto H, et al. (2013) Platelet-rich plasma protects rotator cuff-derived cells from the deleterious effects of triamcinolone acetonide. J Orthop Res 31: 976–982. pmid:23280560
  12. 12. Redler LH, Thompson SA, Hsu SH, Ahmad CS, Levine WN (2011) Platelet-rich plasma therapy: a systematic literature review and evidence for clinical use. Phys Sportsmed 39: 42–51. pmid:21378486
  13. 13. Anitua E, Sanchez M, Orive G, Andia I (2007) The potential impact of the preparation rich in growth factors (PRGF) in different medical fields. Biomaterials 28: 4551–4560. pmid:17659771
  14. 14. Hamid MS, Yusof A, Mohamed Ali MR (2014) Platelet-rich plasma (PRP) for acute muscle injury: a systematic review. PLoS One 9: e90538. pmid:24587389
  15. 15. Silva AK, Richard C, Bessodes M, Scherman D, Merten OW (2009) Growth factor delivery approaches in hydrogels. Biomacromolecules 10: 9–18. pmid:19032110
  16. 16. Kølle S-FT, Fischer-Nielsen A, Mathiasen AB, Elberg JJ, Oliveri RS, Glovinski PV, et al. (2013) Enrichment of autologous fat grafts with ex-vivo expanded adipose tissue-derived stem cells for graft survival: a randomised placebo-controlled trial. The Lancet 382: 1113–1120. pmid:24075051
  17. 17. McCarrel T, Fortier L (2009) Temporal growth factor release from platelet-rich plasma, trehalose lyophilized platelets, and bone marrow aspirate and their effect on tendon and ligament gene expression. J Orthop Res 27: 1033–1042. pmid:19170097
  18. 18. Sundman EA, Cole BJ, Fortier LA (2011) Growth factor and catabolic cytokine concentrations are influenced by the cellular composition of platelet-rich plasma. Am J Sports Med 39: 2135–2140. pmid:21846925
  19. 19. Hayashi C, Gudino CV, Gibson FC III, Genco CA (2010) Review: Pathogen-induced inflammation at sites distant from oral infection: bacterial persistence and induction of cell-specific innate immune inflammatory pathways. Mol Oral Microbiol 25: 305–316. pmid:20883220
  20. 20. Pihlstrom BL, Michalowicz BS, Johnson NW (2005) Periodontal diseases. Lancet 366: 1809–1820. pmid:16298220
  21. 21. Feng Z, Weinberg A (2006) Role of bacteria in health and disease of periodontal tissues. Periodontol 2000 40: 50–76. pmid:16398685
  22. 22. Anitua E, Prado R, Azkargorta M, Rodriguez-Suarez E, Iloro I, Casado-Vela J, et al. (2013) High-throughput proteomic characterization of plasma rich in growth factors (PRGF-Endoret)-derived fibrin clot interactome. J Tissue Eng Regen Med.
  23. 23. Anitua E, Troya M, Orive G (2012) Plasma rich in growth factors promote gingival tissue regeneration by stimulating fibroblast proliferation and migration and by blocking transforming growth factor-beta1-induced myodifferentiation. J Periodontol 83: 1028–1037. pmid:22145805
  24. 24. Anitua E, Tejero R, Zalduendo MM, Orive G (2013) Plasma rich in growth factors promotes bone tissue regeneration by stimulating proliferation, migration, and autocrine secretion in primary human osteoblasts. J Periodontol 84: 1180–1190. pmid:23088531
  25. 25. Colella AD, Chegenii N, Tea MN, Gibbins IL, Williams KA, Chataway TK (2012) Comparison of Stain-Free gels with traditional immunoblot loading control methodology. Anal Biochem 430: 108–110. pmid:22929699
  26. 26. Okahashi N, Inaba H, Nakagawa I, Yamamura T, Kuboniwa M, Nakayama K, et al. (2004) Porphyromonas gingivalis induces receptor activator of NF-kappaB ligand expression in osteoblasts through the activator protein 1 pathway. Infect Immun 72: 1706–1714. pmid:14977979
  27. 27. Chen FM, Zhang J, Zhang M, An Y, Chen F, Wu ZF (2010) A review on endogenous regenerative technology in periodontal regenerative medicine. Biomaterials 31: 7892–7927. pmid:20684986
  28. 28. Garlet GP (2010) Destructive and protective roles of cytokines in periodontitis: a re-appraisal from host defense and tissue destruction viewpoints. J Dent Res 89: 1349–1363. pmid:20739705
  29. 29. Seo T, Cha S, Kim TI, Lee JS, Woo KM (2012) Porphyromonas gingivalis-derived lipopolysaccharide-mediated activation of MAPK signaling regulates inflammatory response and differentiation in human periodontal ligament fibroblasts. J Microbiol 50: 311–319. pmid:22538661
  30. 30. Li JP, Li FY, Xu A, Cheng B, Tsao SW, Fung ML, et al. (2012) Lipopolysaccharide and hypoxia-induced HIF-1 activation in human gingival fibroblasts. J Periodontol 83: 816–824. pmid:22087807
  31. 31. McCarrel TM, Minas T, Fortier LA (2012) Optimization of leukocyte concentration in platelet-rich plasma for the treatment of tendinopathy. J Bone Joint Surg Am 94: e143(141–148). pmid:23032594
  32. 32. Scott A, Khan KM, Roberts CR, Cook JL, Duronio V (2004) What do we mean by the term "inflammation"? A contemporary basic science update for sports medicine. Br J Sports Med 38: 372–380. pmid:15155453
  33. 33. Portela GS, Cerci DX, Pedrotti G, Araujo MR, Deliberador TM, Zielak JC, et al. (2013) L-PRP diminishes bone matrix formation around autogenous bone grafts associated with changes in osteocalcin and PPAR-gamma immunoexpression. Int J Oral Maxillofac Surg.
  34. 34. Giovanini AF, Deliberador TM, Tannuri Nemeth JE, Crivellaro VR, Portela GS, de Oliveira Filho MA, et al. (2013) Leukocyte-platelet-rich plasma (L-PRP) impairs the osteoconductive capacity of the autograft associated to changes in the immunolocalization of TGF-beta1 and its co-expression with Wnt10b and CD34 cells. J Craniomaxillofac Surg 41: e180–186. pmid:23485483
  35. 35. Wasterlain AS, Braun HJ, Dragoo JL (2012) Contents and Formulations of Platelet-Rich Plasma. Operative Techniques in Orthopaedics 22: 33–42.
  36. 36. Pillitteri D, Bassus S, Boller K, Mahnel R, Scholz T, Westrup D, et al. (2007) Thrombin-induced interleukin 1beta synthesis in platelet suspensions: impact of contaminating leukocytes. Platelets 18: 119–127. pmid:17365860
  37. 37. Anitua E, Prado R, Sánchez M, Orive G (2012) Platelet-Rich Plasma: Preparation and Formulation. Operative Techniques in Orthopaedics 22: 25–32.
  38. 38. Imatani T, Kato T, Okuda K (2001) Production of inflammatory cytokines by human gingival fibroblasts stimulated by cell-surface preparations of Porphyromonas gingivalis. Oral Microbiol Immunol 16: 65–72. pmid:11240858
  39. 39. Belibasakis GN, Meier A, Guggenheim B, Bostanci N (2011) Oral biofilm challenge regulates the RANKL-OPG system in periodontal ligament and dental pulp cells. Microb Pathog 50: 6–11. pmid:21075196
  40. 40. Scheres N, de Vries TJ, Brunner J, Crielaard W, Laine ML, Everts V (2011) Diverse effects of Porphyromonas gingivalis on human osteoclast formation. Microb Pathog 51: 149–155. pmid:21539907
  41. 41. Tang L, Zhou XD, Wang Q, Zhang L, Wang Y, Li XY, et al. (2011) Expression of TRAF6 and pro-inflammatory cytokines through activation of TLR2, TLR4, NOD1, and NOD2 in human periodontal ligament fibroblasts. Arch Oral Biol 56: 1064–1072. pmid:21457942
  42. 42. Sundararaj KP, Samuvel DJ, Li Y, Sanders JJ, Lopes-Virella MF, Huang Y (2009) Interleukin-6 released from fibroblasts is essential for up-regulation of matrix metalloproteinase-1 expression by U937 macrophages in coculture: cross-talking between fibroblasts and U937 macrophages exposed to high glucose. J Biol Chem 284: 13714–13724. pmid:19307187
  43. 43. Graves D (2008) Cytokines that promote periodontal tissue destruction. J Periodontol 79: 1585–1591. pmid:18673014
  44. 44. Contassot E, Beer HD, French LE (2012) Interleukin-1, inflammasomes, autoinflammation and the skin. Swiss Med Wkly 142: w13590. pmid:22653747
  45. 45. Domeij H, Yucel-Lindberg T, Modeer T (2006) Cell interactions between human gingival fibroblasts and monocytes stimulate the production of matrix metalloproteinase-1 in gingival fibroblasts. J Periodontal Res 41: 108–117. pmid:16499713
  46. 46. Werner S, Grose R (2003) Regulation of wound healing by growth factors and cytokines. Physiol Rev 83: 835–870. pmid:12843410
  47. 47. Barrientos S, Stojadinovic O, Golinko MS, Brem H, Tomic-Canic M (2008) Growth factors and cytokines in wound healing. Wound Repair Regen 16: 585–601. pmid:19128254
  48. 48. Kishuku M, Nishioka Y, Abe S, Kishi J, Ogino H, Aono Y, et al. (2009) Expression of soluble vascular endothelial growth factor receptor-1 in human monocyte-derived mature dendritic cells contributes to their antiangiogenic property. J Immunol 183: 8176–8185. pmid:20007583
  49. 49. Wu FT, Stefanini MO, Mac Gabhann F, Kontos CD, Annex BH, Popel AS (2010) A systems biology perspective on sVEGFR1: its biological function, pathogenic role and therapeutic use. J Cell Mol Med 14: 528–552. pmid:19840194
  50. 50. Anitua E, Zalduendo MM, Prado R, Alkhraisat MH, Orive G (2014) Morphogen and proinflammatory cytokine release kinetics from PRGF-Endoret fibrin scaffolds: Evaluation of the effect of leukocyte inclusion. J Biomed Mater Res A.
  51. 51. Weisel JW (2004) The mechanical properties of fibrin for basic scientists and clinicians. Biophys Chem 112: 267–276. pmid:15572258
  52. 52. Rowe SL, Lee S, Stegemann JP (2007) Influence of thrombin concentration on the mechanical and morphological properties of cell-seeded fibrin hydrogels. Acta Biomater 3: 59–67. pmid:17085089
  53. 53. Anitua E, Sanchez M, Nurden AT, Zalduendo M, de la Fuente M, Orive G, et al. (2006) Autologous fibrin matrices: a potential source of biological mediators that modulate tendon cell activities. J Biomed Mater Res A 77: 285–293. pmid:16400654
  54. 54. Wang PL, Azuma Y, Shinohara M, Ohura K (2000) Toll-like receptor 4-mediated signal pathway induced by Porphyromonas gingivalis lipopolysaccharide in human gingival fibroblasts. Biochem Biophys Res Commun 273: 1161–1167. pmid:10891389
  55. 55. Yamamoto Y, Gaynor RB (2004) IkappaB kinases: key regulators of the NF-kappaB pathway. Trends Biochem Sci 29: 72–79. pmid:15102433
  56. 56. Viatour P, Merville MP, Bours V, Chariot A (2005) Phosphorylation of NF-kappaB and IkappaB proteins: implications in cancer and inflammation. Trends Biochem Sci 30: 43–52. pmid:15653325
  57. 57. Ghosh S, Karin M (2002) Missing pieces in the NF-kappaB puzzle. Cell 109 Suppl: S81–96. pmid:11983155
  58. 58. Nathan C (2002) Points of control in inflammation. Nature 420: 846–852. pmid:12490957
  59. 59. Nathan C, Ding A (2010) Nonresolving inflammation. Cell 140: 871–882. pmid:20303877