Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters

Publication year range
1.
Cell Tissue Bank ; 25(2): 559-570, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38363442

ABSTRACT

Scaffold design is one of the three most essential parts of tissue engineering. Platelet-rich plasma (PRP) and platelet-rich fibrin (PRF) have been used in clinics and regenerative medicine for years. However, the temporal release of their growth factors limits their efficacy in tissue engineering. In the present study, we planned to synthesize nanofibrous scaffolds with the incorporation of PRP and PRF by electrospinning method to evaluate the effect of the release of PRP and PRF growth factors on osteogenic gene expression, calcification, proliferation, and cell adhesion of human bone marrow mesenchymal stem cell (h-BMSC) as they are part of scaffold structures. Therefore, we combined PRP/PRF, derived from the centrifugation of whole blood, with gelatin and Polycaprolactone (PCL) and produced nanofibrous electrospun PCL/Gel/PRP and PCL/Gel/PRF scaffolds. Three groups of scaffolds were fabricated, and h-BMSCs were seeded on them: (1) PCL/Gel; (2) PCL/Gel/PRP; (3) PCL/Gel/PRF. MTS assay was performed to assess cell proliferation and adhesion, and alizarin red staining confirmed the formation of bone minerals during the experiment. The result indicated that PCL/Gel did not have any better outcomes than the PRP and PRF group in any study variants after the first day of the experiment. PCL/gelatin/PRF was more successful regarding cell proliferation and adhesion. Although PCL/gelatin/PRP showed more promising results on the last day of the experiment in mineralization and osteogenic gene expression, except RUNX2, in which the difference with PCL/gelatin/PRF group was not significant.


Subject(s)
Cell Adhesion , Cell Proliferation , Gelatin , Mesenchymal Stem Cells , Osteogenesis , Platelet-Rich Fibrin , Platelet-Rich Plasma , Polyesters , Tissue Scaffolds , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Gelatin/chemistry , Tissue Scaffolds/chemistry , Polyesters/chemistry , Platelet-Rich Plasma/metabolism , Platelet-Rich Plasma/chemistry , Cell Proliferation/drug effects , Cell Adhesion/drug effects , Platelet-Rich Fibrin/chemistry , Platelet-Rich Fibrin/metabolism , Cells, Cultured , Tissue Engineering/methods , Nanofibers/chemistry
2.
Clin Oral Investig ; 27(5): 2311-2319, 2023 May.
Article in English | MEDLINE | ID: mdl-37079155

ABSTRACT

OBJECTIVES: To assess the outcome of leukocyte-platelet-rich fibrin (L-PRF) on the rate of maxillary canine retraction and its correlation with the levels of Receptor activator of nuclear factor kappa-B ligand (RANKL), osteoprotegerin (OPG), and RANKL:OPG in the gingival crevicular fluid (GCF) during comprehensive orthodontic treatment. SUBJECTS AND METHODS: Eighteen females who required all 1st premolars extraction for the correction of their class I bimaxillary protrusion malocclusions were included. The L-PRF plugs were placed in the experimental side 1st premolar extraction sockets. Canine retraction was performed by sliding mechanics. Canine retraction was assessed from the maxillary study models prepared just before the extraction (T0) and then at 1 week (T1), 2 weeks (T2), 4 weeks (T3), and 8 weeks (T4) after the 1st premolar extraction and placement of L-PRF plugs. The concentrations of RANKL and OPG in the GCF were evaluated at T0, T1, T2, T3, and T4. RESULTS: In experimental sides, the amount of canine retraction was statistically more during the T0-T1, T1-T2, and T2-T3 periods. The mean concentration of RANKL at T1, T2, and T3 was significantly more in the experimental sides. The mean concentration of OPG was significantly less in the experimental sides at T2, T3, and T4. The RANKL:OPG was significantly more in the experimental sides at T1, T2, T3, and T4. No significant correlation was found between amount of canine retraction and concentration of RANKL and OPG and RANKL to OPG ratio in GCF. CONCLUSIONS: The L-PRF accelerated the rate of maxillary canine retraction by 0.28 mm over an 8-week period. The L-PRF favored the local osteoclastogenesis by enhancing the RANKL and suppressing the OPG concentrations. There was no significant correlation between the rate of maxillary canine retraction and expression of RANKL, OPG, and RANKL:OPG in GCF. TRIAL REGISTRATION: The Clinical Trials Registry of India (Reg. No. CTRI/2020/10/028390, Date-13.10.2020).


Subject(s)
Bone Density Conservation Agents , Platelet-Rich Fibrin , Female , Animals , Gingival Crevicular Fluid/chemistry , Tooth Movement Techniques , Platelet-Rich Fibrin/chemistry , Osteoprotegerin/metabolism , Biomarkers/metabolism
3.
Mol Biol Rep ; 48(5): 4573-4580, 2021 May.
Article in English | MEDLINE | ID: mdl-34146200

ABSTRACT

Leukocyte and platelet rich fibrin (L-PRF) is one of the platelet concentrates used to support regeneration and healing process. Many studies showed possible immunological and antibacterial properties of L-PRF. We perform an in vitro study to analyze the effect of L-PRF on platelet activation, platelet-leukocytes interactions and antimicrobial activity, important components in the healing process. Molecular biomarkers related with platelet activation and platelet-leukocyte interactions were analyzed by means of flow cytometry when L-PRF exudate was added to whole blood platelets. L-PRF membrane was used to evaluate antimicrobial activity using Enterococcus faecalis (ATCC 29212), Pseudomonas aeruginosa (ATCC 27853) and Candida albicans (ATCC 90028). Our experimental design allows to evaluate platelet activation and analyze molecular biomarkers of other immune cells and platelet-leukocyte interactions. From the results obtained we can conclude that L-PRF can be a valuable tool in healing process, efficient in activating platelets of whole blood and inhibiting microbial growth. In our opinion, the use of L-PRF exudate, in addition to L-PRF membrane, presents some advantages that have to be considered in clinical trials. Additional research on the characterization and quantification of cells and its products present in the L-PRF exudate, as well as on the temporal factor released. Also, further studies using strains isolated from clinical cases are needed.


Subject(s)
Anti-Infective Agents/pharmacology , Blood Platelets/metabolism , Platelet Activation/genetics , Platelet-Rich Fibrin/chemistry , Wound Healing/genetics , Anti-Infective Agents/blood , Anti-Infective Agents/chemistry , Candida albicans/drug effects , Enterococcus faecalis/drug effects , Humans , Leukocytes/chemistry , Platelet-Rich Plasma/metabolism , Pseudomonas aeruginosa/drug effects
4.
Molecules ; 26(23)2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34885714

ABSTRACT

BACKGROUND: Platelet-rich fibrin (PRF) has gained popularity in craniofacial surgery, as it provides an excellent reservoir of autologous growth factors (GFs) that are essential for bone regeneration. However, the low elastic modulus, short-term clinical application, poor storage potential and limitations in emergency therapy use restrict its more widespread clinical application. This study fabricates lyophilised PRF (Ly-PRF), evaluates its physical and biological properties, and explores its application for craniofacial tissue engineering purposes. MATERIAL AND METHODS: A lyophilisation method was applied, and the outcome was evaluated and compared with traditionally prepared PRF. We investigated how lyophilisation affected PRF's physical characteristics and biological properties by determining: (1) the physical and morphological architecture of Ly-PRF using SEM, and (2) the kinetic release of PDGF-AB using ELISA. RESULTS: Ly-PRF exhibited a dense and homogeneous interconnected 3D fibrin network. Moreover, clusters of morphologically consistent cells of platelets and leukocytes were apparent within Ly-PRF, along with evidence of PDGF-AB release in accordance with previously reports. CONCLUSIONS: The protocol established in this study for Ly-PRF preparation demonstrated versatility, and provides a biomaterial with growth factor release for potential use as a craniofacial bioscaffold.


Subject(s)
Intercellular Signaling Peptides and Proteins/chemistry , Platelet-Derived Growth Factor/biosynthesis , Platelet-Rich Fibrin/chemistry , Tissue Engineering , Adult , Blood Platelets/chemistry , Blood Platelets/metabolism , Bone Regeneration/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Freeze Drying , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/therapeutic use , Leukocytes/chemistry , Male , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Platelet-Rich Fibrin/metabolism , Tissue Donors , Young Adult
5.
Clin Oral Investig ; 24(3): 1151-1160, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31292776

ABSTRACT

OBJECTIVES: The aim of this study was to describe the histological and clinical outcome of "dentin block" (a mixture of autologous particulate dentin, leukocyte- and platelet-rich fibrin (L-PRF), and liquid fibrinogen) in alveolar ridge preservation. MATERIAL AND METHODS: Ten extraction sockets were grafted with "dentin block," a mixture of particulate autologous dentin with chopped leukocyte-platelet-rich fibrin (L-PRF) membranes at a 1:1 ratio, and liquid fibrinogen as a binder. Two grafted sites were followed at 4 and 5 months, and 6 sites at 6 months. Biopsies were taken from the core of the grafted site for histologic and histo-morphometric analysis. RESULTS: All patients completed the study without any adverse event. The vertical and horizontal dimensions of the alveolar ridge were preserved or even increased after 4, 5, or 6 months and remained stable after 6 months of the implant placement. The histological examination revealed a median relative percentage of bone, dentin, and connective tissue of 57.0, 0.9, and 39.3%, respectively. A comparison of samples at different time points (4, 5, and 6 months) showed a progressive increase in the proportion of bone with a decrease in the proportion of dentin. The bone was compact with normal osteocytes and moderate osteoblastic activity. In 4 out of 10 samples, no dentin was observed; in the other samples, it represented 1-5% (with geometric fragments). CONCLUSIONS: Dentin block showed to be a suitable bone substitute in an alveolar ridges preservation model. CLINICAL RELEVANCE: The promising results of dentin block as a bone substitute in alveolar ridge preservation could have an important clinical impact considering this biomaterial brings together the regenerative potential of three autologous products with excellent biological and clinical behavior, low risk of adverse effects, and feasible acquisition.


Subject(s)
Alveolar Ridge Augmentation , Bone Substitutes/therapeutic use , Dentin/chemistry , Fibrinogen/chemistry , Platelet-Rich Fibrin/chemistry , Adult , Female , Humans , Middle Aged , Pilot Projects , Tooth Extraction , Tooth Socket
6.
Int J Mol Sci ; 21(2)2020 Jan 16.
Article in English | MEDLINE | ID: mdl-31963217

ABSTRACT

The main aim of this study is to develop a one-stage method to combine platelet-rich fibrin (PRF) and autologous cartilage autografts for porcine articular cartilage repair. The porcine chondrocytes were treated with different concentrations of PRF-conditioned media and were evaluated for their cell viability and extracellular glycosaminoglycan (GAG) synthesis during six day cultivation. The chemotactic effects of PRF on chondrocytes on undigested cartilage autografts were revealed in explant cultures. For the in vivo part, porcine chondral defects were created at the medial femoral condyles of which were (1) left untreated, (2) implanted with PRF combined with hand-diced cartilage grafts, or (3) implanted with PRF combined with device-diced cartilage grafts. After six months, gross grades, histological, and immunohistochemical analyses were compared. The results showed that PRF promotes the viability and GAG expression of the cultured chondrocytes. Additionally, the PRF-conditioned media induce significant cellular migration and outgrowth of chondrocytes from undigested cartilage grafts. In the in vivo study, gross grading and histological scores showed significantly better outcomes in the treatment groups as compared with controls. Moreover, both treatment groups showed significantly more type II collagen staining and minimal type I collagen staining as compared with controls, indicating more hyaline-like cartilage and less fibrous tissue. In conclusion, PRF enhances the viability, differentiation, and migration of chondrocytes, thus, showing an appealing capacity for cartilage repair. The data altogether provide evidences to confirm the feasibility of a one-stage, culture-free method of combining PRF and cartilage autografts for repairing articular cartilage defects. From translational standpoints, these advantages benefit clinical applications by simplifying and potentiating the efficacy of cartilage autograft transplants.


Subject(s)
Cartilage, Articular/cytology , Cell Movement/physiology , Cell Survival/physiology , Chondrocytes/cytology , Platelet-Rich Fibrin/chemistry , Animals , Cartilage, Articular/metabolism , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Glycosaminoglycans/metabolism , Knee Joint/cytology , Swine , Swine, Miniature , Transplantation, Autologous
7.
Int J Mol Sci ; 21(20)2020 Oct 15.
Article in English | MEDLINE | ID: mdl-33076376

ABSTRACT

Solid platelet-rich fibrin (PRF) is produced with centrifugation tubes designed to accelerate clotting. Thus, activated platelets may accumulate within the fibrin-rich extracellular matrix even before centrifugation is initiated. It can thus be assumed that platelets and their growth factors such as transforming growth factor-ß (TGF-ß) are trapped within PRF independent of their relative centrifugal force (RCF), the gravitation or g-force. To test this assumption, we prepared PRF membranes with tubes where clotting is activated by a silicone-coated interior. Tubes underwent 210 g, 650 g and 1500 g for 12 min in a horizontal centrifuge. The respective PRF membranes, either in total or separated into a platelet-poor plasma and buffy coat fraction, were subjected to repeated freeze-thawing to prepare lysates. Gingival fibroblasts were exposed to the PRF lysates to provoke the expression of TGF-ß target genes. We show here that the expression of interleukin 11 (IL11) and NADPH oxidase 4 (NOX4), and Smad2/3 signaling were similarly activated by all lysates when normalized to the size of the PRF membranes. Notably, platelet-poor plasma had significantly less TGF-ß activity than the buffy coat fraction at both high-speed protocols. In contrast to our original assumption, the TGF-ß activity in PRF lysates produced using horizontal centrifugation follows a gradient with increasing concentration from the platelet-poor plasma towards the buffy coat layer.


Subject(s)
Blood Buffy Coat/metabolism , Fibroblasts/drug effects , Membranes, Artificial , Platelet-Rich Fibrin/chemistry , Transforming Growth Factor beta/pharmacology , Blood Coagulation , Cells, Cultured , Centrifugation/methods , Fibroblasts/metabolism , Gingiva/cytology , Gravitation , Humans , Interleukin-11/genetics , Interleukin-11/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Silicones/chemistry , Smad Proteins/genetics , Smad Proteins/metabolism
8.
Int J Mol Sci ; 21(12)2020 06 20.
Article in English | MEDLINE | ID: mdl-32575800

ABSTRACT

Platelet-released growth factor (PRGF) is a thrombocyte concentrate lysate which, like its clinically equivalent variations (e.g., Vivostat PRF® (platelet-rich fibrin)), is known to support the healing of chronic and hard-to-heal wounds. However, studies on the effect of PRGF on keratinocytes remain scarce. This study aims to identify genes in keratinocytes that are significantly influenced by PRGF. Therefore, we performed a whole transcriptome and gene ontology (GO) enrichment analysis of PRGF-stimulated human primary keratinocytes. This revealed an increased expression of genes involved in extracellular matrix (ECM) organization. Real-time polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA) analysis confirmed the PRGF-mediated induction of selected ECM-related factors such as transforming growth factor beta-induced protein, fibronectin 1, matrix metalloproteinase-9, transglutaminase 2, fermitin family member 1, collagen type I alpha 1 and collagen type XXII alpha 1. PRGF-induced expression of the above factors was influenced by blockade of the epidermal growth factor receptor (EGFR), a receptor playing a crucial role in wound healing. A differential induction of the investigated factors was also detected in skin explants exposed to PRGF and in experimentally generated in vivo wounds treated with Vivostat PRF®. Together, our study indicates that the induction of ECM-related factors may contribute to the beneficial wound-healing effects of PRGF-based formulations.


Subject(s)
Cytokines/pharmacology , Extracellular Matrix/genetics , Gene Expression Profiling/methods , Intercellular Signaling Peptides and Proteins/pharmacology , Keratinocytes/cytology , Cells, Cultured , Collagen Type I, alpha 1 Chain , Extracellular Matrix/drug effects , Gene Expression Regulation/drug effects , Gene Ontology , Gene Regulatory Networks/drug effects , High-Throughput Nucleotide Sequencing , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Platelet-Rich Fibrin/chemistry , Primary Cell Culture , Sequence Analysis, RNA , Wound Healing/drug effects
9.
Cytotherapy ; 21(1): 83-95, 2019 01.
Article in English | MEDLINE | ID: mdl-30509773

ABSTRACT

BACKGROUND: Several methods to cultivate limbal epithelial stem cells (LESCs) in vitro with the support of feeder layers and different growth medium formulations have been established for several years. The initial green medium consists of various ingredients that exhibit a non-optimal level of biosafety, therefore, different modifications have been made to suit it to safe clinical applications. However, the question of which formulation is the most appropriate remains to be answered. AIMS: This study evaluated the outgrowth kinetics and stemness of cells cultured from human limbal explants with the aim of preserving LESC characteristics in the human-derived platelet-rich fibrin (HPRF)-conditioned medium with no feeder cell layer or carrier for the first time. The final composition of the cell culture system included only human-derived products without any xenobiotic or chemical substances to minimize the potential risk for human health, which will be useful for clinical purposes. METHODS: To test our hypothesis, limbal explants were incubated with either Dulbecco's Modified Eagle's Medium (DMEM)/F12-10% human serum (HS), human-derived amniotic membrane (HAM)-conditioned DMEM/F12-10% HS or HPRF-conditioned DMEM/F12-10% HS to determine whether outgrowth kinetics and stemness of cells show any differences among groups. RESULTS: The results showed that the HPRF-conditioned medium showed higher concentration levels of growth factors, which may be involved in the promotion of LESC expansion while preserving the stem cell characteristics. HPRF-conditioned medium had significantly superior capacity to enhance the cell growth rate, the stem/progenitor cell phenotype and the expressions of putative stem cell markers. CONCLUSION: This novel xeno-feeder-chemical-free, completely human-derived and biologically safe culture system including HPRF and HS would be of interest to replace conventional cell culture strategies to meet safety requirements mandatory for clinical use in humans.


Subject(s)
Cell Culture Techniques/methods , Epithelium, Corneal/cytology , Feeder Cells , Limbus Corneae/cytology , Stem Cells/physiology , Adolescent , Adult , Aged , Amnion/chemistry , Cadaver , Cell Proliferation , Cell Survival , Culture Media, Conditioned/chemistry , Female , Humans , Intercellular Signaling Peptides and Proteins , Male , Middle Aged , Platelet-Rich Fibrin/chemistry , Pregnancy , Young Adult
10.
Clin Oral Investig ; 23(3): 1309-1318, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30003342

ABSTRACT

OBJECTIVES: The aim of the present study was to evaluate the blood cell content, morphological aspects, gene expression of type I collagen, and release of growth factors on an injectable platelet rich fibrin (i-PRF). MATERIALS AND METHODS: Blood samples were collected from 15 volunteers to prepare i-PRF samples. Peripheral blood was used as a control group. Blood clot and i-PRF samples were cultured for 10 days. The supernatant of the samples was collected for ELISA immunoassay quantification of PDGF and VEGF growth factors over periods of 1, 8, 24, 72, and 240 h. I-PRF and blood clot samples were biologically characterized using histological and immunohistochemistry analysis for IL-10, osteocalcin, and TGF-ß. Scanning electron microscopy (SEM) was used to inspect the fibrin network and distribution of blood platelets and leukocytes. Reverse transcriptase polymerase chain reaction (RT-PCR) method was used to evaluate gene expression for type I collagen. RESULTS: A higher concentration of platelets and lymphocytes was recorded in i-PRF than in peripheral blood (p < 0.05). The release of VEGF was higher in blood clot samples (1933 ± 704) than that for i-PRF (852 ± 376; p < 0.001). Immunohistochemistry showed upregulation of TGF-B, IL-10, and osteocalcin in the i-PRF group. RT-PCR showed increased type I collagen gene expression in i-PRF (p < 0.05). SEM images revealed agglomeration of platelets in some regions, while a fibrin networking was noticeable in the entire i-PRF sample. CONCLUSIONS: Injectable platelet rich fibrin becomes a good approach for soft and mineralized tissue healing considering the formation of a three-dimensional fibrin network embedding platelets, leukocytes, type I collagen, osteocalcin, and growth factors. Indeed, the injectable platelet rich fibrin can be indicated in several medical applications regarding bioactivity, simplied technique, and flowable mixing with other biomaterials. CLINICAL RELEVANCE: Morphological, cell, and protein characterization of platelet rich fibrin provides a better understanding of the clinical effects and improvement of clinical guidelines for several medical applications. Once well physicochemical and biologically characterized, the use of an injectable platelet rich fibrin can be extended to other applications in the field of orthopedics, periodontics, and implant dentistry on the repairing process of both soft and mineralized tissues.


Subject(s)
Platelet-Rich Fibrin/chemistry , Platelet-Rich Fibrin/cytology , Adult , Blood Platelets/cytology , Collagen Type I/chemistry , Fibrin/chemistry , Humans , Interleukin-10/chemistry , Leukocytes/cytology , Male , Osteocalcin/chemistry , Transforming Growth Factor beta1/chemistry , Vascular Endothelial Growth Factor A/chemistry
11.
Int J Mol Sci ; 20(7)2019 Apr 05.
Article in English | MEDLINE | ID: mdl-30959772

ABSTRACT

Nowadays, research in Tissue Engineering and Regenerative Medicine is focusing on the identification of instructive scaffolds to address the requirements of both clinicians and patients to achieve prompt and adequate healing in case of injury. Among biomaterials, hemocomponents, and in particular Platelet-rich Fibrin matrices, have aroused widespread interest, acting as delivery platforms for growth factors, cytokines and immune/stem-like cells for immunomodulation; their autologous origin and ready availability are also noteworthy aspects, as safety- and cost-related factors and practical aspects make it possible to shorten surgical interventions. In fact, several authors have focused on the use of Platelet-rich Fibrin in cartilage and tendon tissue engineering, reporting an increasing number of in vitro, pre-clinical and clinical studies. This narrative review attempts to compare the relevant advances in the field, with particular reference being made to the regenerative role of platelet-derived growth factors, as well as the main pre-clinical and clinical research on Platelet-rich Fibrin in chondrogenesis and tenogenesis, thereby providing a basis for critical revision of the topic.


Subject(s)
Cartilage/physiology , Platelet-Rich Fibrin/chemistry , Regenerative Medicine , Tendons/physiology , Tissue Scaffolds/chemistry , Translational Research, Biomedical , Humans
12.
Cell Tissue Bank ; 19(3): 341-356, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29264693

ABSTRACT

Fibrin-platelet glue (FPG) is a blood derivative, in which platelets and fibrinogen are concentrated in a small plasma volume, by differential centrifugation and precipitation. It can form a three-dimensional and biocompatible fibrin scaffold with a myriad of growth factors and proteins that are released progressively to the local environment and contribute to the accelerated postoperative bone healing. Gelatin (Gel) is a derivative of collagen and can promote cell adhesion and proliferation due to its unique sequence of amino acids, so it is suitable for bone tissue applications. This study examined the effects of Gel, FPG and their combinations as bone scaffold on the healing of surgically created critical-size defects in rat radius. Fifty critical size defects of 5 mm long were bilaterally created in the radial diaphysis of 25 rats. The animals were randomly divided into five equal groups as empty defect, autograft, Gel, FPG and Gel-FPG groups (n = 10 in each group). Radiographs of each forelimb were taken postoperatively on the 1st day and then at the 28th and 56th days post injury to evaluate bone formation, union and remodeling of the defect. After 56 days, the rats were euthanized and their harvested healing bone samples were evaluated by histopathology, scanning electron microscopy (SEM) and biomechanical testing. The results of present study showed that the Gel alone did not significantly affect bone healing and regeneration; however, the Gel treated defects promoted healing more than those that were left untreated (negative control). Furthermore, the FPG-enhanced grafts provided a good scaffold containing numerous growth factors for proliferation of osteoinduction and was effective in improving the structural and functional properties of the newly formed bone more than that of the untreated and also the Gel treated groups. Incorporation of Gel into the FPG scaffold improved healing potential of the FPG scaffold; however, it was still inferior to the autograft (positive control). Although the Gel-FPG scaffolds had best effectiveness during bone regeneration, it still needs to be further enhanced by incorporation of the ceramic and osteoinductive biomaterials.


Subject(s)
Biocompatible Materials/therapeutic use , Gelatin/therapeutic use , Osteogenesis , Platelet-Rich Fibrin , Radius/injuries , Tissue Scaffolds/chemistry , Animals , Biocompatible Materials/chemistry , Biomechanical Phenomena , Bone Regeneration , Disease Models, Animal , Gelatin/chemistry , Male , Platelet-Rich Fibrin/chemistry , Radius/pathology , Radius/physiology , Rats , Rats, Wistar
13.
J Contemp Dent Pract ; 19(7): 895-899, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-30066697

ABSTRACT

INTRODUCTION: Free gingival graft is a procedure that is used to increase keratinized tissue around teeth and edentulous sites for future dental implants. Keratinized tissue is critical for maintainability of surgical site and flap management. Platelet-rich fibrin consists of bioactive and biological components, mainly composed of growth factors. Growth factors attract stem cells to the site of release and stimulate cell proliferation. Moreover, growth factors promote angiogenesis, which accelerates wound healing. Site preparation is critical in implant dentistry, including soft tissue and hard tissue augmentation. AIM: To improve free gingival graft (FGG) healing by using platelet-rich fibrin (PRF) at the soft tissue defect area of extracted upper right first molar in order to restore keratinized tissue and prepare the site for bone grafting followed by dental implant placement. CASE REPORT: A healthy female patient, 59 years old, presented to the dental clinic at the University at Buffalo, School of Dental Medicine, seeking dental implants to restore missing teeth. The patient had an extraction for upper right first molar 4 months ago. The surgical extraction left severe soft and hard tissue defects at the site. Free gingival graft was placed at the site to increase keratinized tissue band followed by PRF to improve healing. Cyanoacrylate adhesive was used to stabilize PRF Two months later, a full-thickness flap was reflected, and tenting screws placed with bone grafting at the site. Titanium-reenforced membrane was placed over the bone graft. Three months later, tenting screws were removed and two dental implants were placed at the site. CONCLUSION: The use of PRF accelerates the healing of FGG. More tissue handling could be achieved by increasing the kera-tinized tissue during surgical procedures. CLINICAL SIGNIFICANCE: The combination of PRF and FGG could help in the healing process during soft tissue procedures.


Subject(s)
Dental Implantation, Endosseous/methods , Dental Implantation , Gingiva/transplantation , Molar/surgery , Platelet-Rich Fibrin , Soft Tissue Injuries/etiology , Soft Tissue Injuries/therapy , Therapy, Soft Tissue/methods , Tooth Extraction/adverse effects , Bone Transplantation/methods , Cell Proliferation , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Maxilla/surgery , Middle Aged , Neovascularization, Physiologic , Osseointegration , Platelet-Rich Fibrin/chemistry , Platelet-Rich Fibrin/physiology , Stem Cells , Wound Healing
14.
Carbohydr Polym ; 339: 122174, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38823938

ABSTRACT

Segmental bone defects can arise from trauma, infection, metabolic bone disorders, or tumor removal. Hydrogels have gained attention in the field of bone regeneration due to their unique hydrophilic properties and the ability to customize their physical and chemical characteristics to serve as scaffolds and carriers for growth factors. However, the limited mechanical strength of hydrogels and the rapid release of active substances have hindered their clinical utility and therapeutic effectiveness. With ongoing advancements in material science, the development of injectable and biofunctionalized hydrogels holds great promise for addressing the challenges associated with segmental bone defects. In this study, we incorporated lyophilized platelet-rich fibrin (LPRF), which contains a multitude of growth factors, into a genipin-crosslinked gelatin/hyaluronic acid (GLT/HA-0.5 % GP) hydrogel to create an injectable and biofunctionalized composite material. Our findings demonstrate that this biofunctionalized hydrogel possesses optimal attributes for bone tissue engineering. Furthermore, results obtained from rabbit model with segmental tibial bone defects, indicate that the treatment with this biofunctionalized hydrogel resulted in increased new bone formation, as confirmed by imaging and histological analysis. From a translational perspective, this biofunctionalized hydrogel provides innovative and bioinspired capabilities that have the potential to enhance bone repair and regeneration in future clinical applications.


Subject(s)
Bone Regeneration , Freeze Drying , Gelatin , Hyaluronic Acid , Hydrogels , Iridoids , Platelet-Rich Fibrin , Animals , Iridoids/chemistry , Iridoids/pharmacology , Gelatin/chemistry , Rabbits , Hydrogels/chemistry , Hydrogels/pharmacology , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Bone Regeneration/drug effects , Platelet-Rich Fibrin/chemistry , Tissue Engineering/methods , Cross-Linking Reagents/chemistry , Tissue Scaffolds/chemistry , Tibia/drug effects , Tibia/surgery
15.
Biomater Sci ; 12(9): 2418-2433, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38511973

ABSTRACT

Bone defects are a common complication of bone diseases, which often affect the quality of life and mental health of patients. The use of biomimetic bone scaffolds loaded with bioactive substances has become a focal point in the research on bone defect repair. In this study, composite scaffolds resembling bone tissue were created using nacre powder (NP) and sodium alginate (SA) through 3D printing. These scaffolds exhibit several physiological structural and mechanical characteristics of bone tissue, such as suitable porosity, an appropriate pore size, applicable degradation performance and satisfying the mechanical requirements of cancellous bone, etc. Then, platelet-rich fibrin (PRF), containing a mass of growth factors, was loaded on the NP/SA scaffolds. This was aimed to fully maximize the synergistic effect with NP, thereby accelerating bone tissue regeneration. Overall, this study marks the first instance of preparing a bionic bone structure scaffold containing NP by 3D printing technology, which is combined with PRF to further accelerate bone regeneration. These findings offer a new treatment strategy for bone tissue regeneration in clinical applications.


Subject(s)
Alginates , Bone Regeneration , Nacre , Platelet-Rich Fibrin , Powders , Printing, Three-Dimensional , Tissue Scaffolds , Alginates/chemistry , Alginates/pharmacology , Bone Regeneration/drug effects , Tissue Scaffolds/chemistry , Nacre/chemistry , Animals , Platelet-Rich Fibrin/chemistry , Tissue Engineering , Humans , Porosity , Bone and Bones/drug effects , Osteogenesis/drug effects
16.
J Appl Biomater Funct Mater ; 22: 22808000241289208, 2024.
Article in English | MEDLINE | ID: mdl-39369296

ABSTRACT

Craniofacial bone defects result from various disorders such as trauma, congenital malformations and infections. Cleft lip and palate are the most prevalent congenital craniofacial birth defect in humans. Growth factors (GFs) are soluble proteins secreted by cells that regulate various cellular processes and tissue regeneration. At present, developing three-dimensional scaffolds for delivering GFs to the site of injury has become an important aspect in craniofacial bone regeneration. This study aims to develop a novel 3D bone substitute using lyophilized-platelet-rich fibrin (LyPRF) biocomposite scaffolds for potential application for CLP repair. Collagen (C), bioglass (BG), and LyPRF were used to fabricate a biocomposite (C-BG-LyPRF) scaffold. The physical, chemical, and biocompatibility properties of the scaffold were evaluated. The C-BG-LyPRF scaffold demonstrated a mean pore diameter of 146 µm within a porosity of 87.26%. The FTIR spectra verified the presence of am-ide I, II, and III functional groups. The inorganic phase of the C-BG-LyPRF scaffold was composed of sodium, calcium, silicon, and phosphorus, as determined by EDX analysis. Furthermore, C-BG-LyPRF scaffold was biocompatible with MC3T3-E1 cells in both the Live/Dead and prolif-eration assays. Data demonstrate the developed C-BG-LyPRF scaffold exhibits biomimetic and biocompatibility properties, establishing it as a promising biomaterial for craniofacial regeneration.


Subject(s)
Cleft Lip , Cleft Palate , Freeze Drying , Platelet-Rich Fibrin , Tissue Scaffolds , Cleft Palate/surgery , Cleft Lip/surgery , Platelet-Rich Fibrin/chemistry , Tissue Scaffolds/chemistry , Mice , Animals , Humans , Ceramics/chemistry , Ceramics/pharmacology , Materials Testing , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Line , Porosity , Bone Regeneration/drug effects , Bone Substitutes/chemistry , Bone Substitutes/pharmacology
17.
J Mater Chem B ; 12(24): 5869-5883, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38775079

ABSTRACT

In recent years, metallic ion-doped magnesium phosphate (MgP)-based degradable bioceramics have emerged as alternative bone substitute materials owing to their excellent biocompatibility, bone-forming ability, bioactivity, and controlled degradability. Conversely, incorporating a biomolecule such as decellularized platelet-rich fibrin (d-PRF) on scaffolds has certain advantages for bone tissue regeneration, particularly in enhanced osteogenesis and angiogenesis. The present study focuses on the impact of d-PRF-loaded multiscale porous zinc-doped magnesium phosphate (Zn-MgP) scaffolds on biodegradability, biocompatibility, and bone regeneration. Scaffolds were fabricated through the powder-metallurgy route utilizing naphthalene as a porogen (porosity = 5-43%). With the inclusion of a higher porogen, a higher fraction of macro-porosity (>20 µm) and pore interconnectivity were observed. X-ray diffraction (XRD) studies confirmed the formation of the farringtonite phase. The developed scaffolds exhibited a minimum ultimate compressive strength (UCS) of 8.5 MPa (for 40 Naph), which lies within the range of UCS of the cancellous bone of humans (2-12 MPa). The in vitro assessment via immersion in physiological fluid yielded a higher deposition of the calcium phosphate (CaP) compound in response to increased macro-porosity and interconnectivity (40 Naph). Cytocompatibility assessed using MC3T3-E1 cells showed that the incorporation of d-PRF coupled with increased porosity resulted the highest cell attachment, proliferation, and viability. For further evaluation, the developed scaffolds were implanted in in vivo rabbit femur condylar defects. Radiography, SEM, OTC labelling, and histology analysis after 2 months of implantation revealed the better invasion of mature osteoblastic cells into the scaffolds with enhanced angiogenesis and superior and accelerated healing of bone defects in d-PRF-incorporated higher porosity scaffolds (40 Naph). Finally, it is hypothesized that the combination of d-PRF incorporation with multiscale porosity and increased interconnectivity facilitated better bone-forming ability, good biocompatibility, and controlled degradability within and around the Zn-doped MgP scaffolds.


Subject(s)
Bone Regeneration , Magnesium Compounds , Phosphates , Platelet-Rich Fibrin , Tissue Scaffolds , Zinc , Bone Regeneration/drug effects , Porosity , Animals , Zinc/chemistry , Zinc/pharmacology , Tissue Scaffolds/chemistry , Mice , Magnesium Compounds/chemistry , Magnesium Compounds/pharmacology , Platelet-Rich Fibrin/chemistry , Rabbits , Phosphates/chemistry , Phosphates/pharmacology , Humans , Cell Proliferation/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology
18.
Int J Biol Macromol ; 273(Pt 1): 133064, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38866288

ABSTRACT

Bone tissue regeneration strategies have incorporated the use of natural polymers, such as hydroxyapatite (nHA), chitosan (CH), gelatin (GEL), or alginate (ALG). Additionally, platelet concentrates, such as platelet-rich fibrin (PRF) have been suggested to improve scaffold biocompatibility. This study aimed to develop scaffolds composed of nHA, GEL, and CH, with or without ALG and lyophilized PRF, to evaluate the scaffold's properties, growth factor release, and dental pulp stem cells (DPSC), and osteoblast (OB) derived from DPSC viability. Four scaffold variations were synthesized and lyophilized. Then, degradation, swelling profiles, and morphological analysis were performed. Furthermore, PDGF-BB and FGF-B growth factors release were quantified by ELISA, and cytotoxicity and cell viability were evaluated. The swelling and degradation profiles were similar in all scaffolds, with pore sizes ranging between 100 and 250 µm. FGF-B and PDGF-BB release was evidenced after 24 h of scaffold immersion in cell culture medium. DPSC and OB-DPSC viability was notably increased in PRF-supplemented scaffolds. The nHA-CH-GEL-PRF scaffold demonstrated optimal physical-biological characteristics for stimulating DPSC and OB-DPSC cell viability. These results suggest lyophilized PRF improves scaffold biocompatibility for bone tissue regeneration purposes.


Subject(s)
Alginates , Cell Survival , Chitosan , Dental Pulp , Durapatite , Gelatin , Osteoblasts , Platelet-Rich Fibrin , Stem Cells , Tissue Scaffolds , Humans , Dental Pulp/cytology , Chitosan/chemistry , Chitosan/pharmacology , Gelatin/chemistry , Platelet-Rich Fibrin/chemistry , Platelet-Rich Fibrin/metabolism , Tissue Scaffolds/chemistry , Stem Cells/drug effects , Stem Cells/cytology , Stem Cells/metabolism , Cell Survival/drug effects , Durapatite/chemistry , Durapatite/pharmacology , Alginates/chemistry , Alginates/pharmacology , Osteoblasts/drug effects , Osteoblasts/cytology , Cell Adhesion/drug effects , Tissue Engineering/methods , Cells, Cultured
19.
F1000Res ; 12: 393, 2023.
Article in English | MEDLINE | ID: mdl-39291141

ABSTRACT

Background: Periodontal regeneration therapies frequently involve autologous platelet concentrates (APCs). They can be used in sinus lift surgeries and socket preservation, among other clinical settings. Platelet rich fibrin (PRF) membrane has been used to treat gingival recession in individuals or groups of individuals using a coronally progressed or lateral pedicle flap. In the treatment of mixed periodontic endodontic lesion/furcation defect, PRF functions as a healing and interpositional biomaterial, filling a cystic cavity. PRF is known to help the bone regeneration process. In the last few years, efforts have been made to enhance the PRFs characteristics and quality. One of them is titanium platelet rich fibrin (T-PRF). Third-generation platelet concentrate no longer contains silica, and its preparation in glass vacuum containers, that no longer creates any known concerns. The effectiveness PRF's has been evaluated in connective tissue and bone repair. The aim of this study is to compare T-PRF to other platelet concentrates and different treatment modalities for periodontal regenerative procedures. Methods: A protocol of this systematic review have been registered in prospero (CRD42022293545). The online database searched were PUBMED, COCHRANE for published articles up to November 2022 without language restrictions. Studies in trial registers, handsearching, bibliographic references of relevant articles were also checked. Data collection and analysis was done by individual authors. Independent eligibility assessments were conducted by four review authors. Then, using the standard Cochrane methodology, four review authors extracted the data and evaluated the risk of bias for individual studies. We developed "Summary of findings" tables and used GRADE to evaluate the evidence. Results: Three studies were included for meta-analysis. Results of meta-analysis supported that T-PRF is effective for correction of both hard and soft tissue defects. Conclusions: The overall qualitative and quantitative analysis suggest that T-PRF has superior structural properties and thicker fibrin network for ensuring predictable success periodontal regenerative procedures.


Subject(s)
Guided Tissue Regeneration, Periodontal , Platelet-Rich Fibrin , Titanium , Humans , Guided Tissue Regeneration, Periodontal/methods , Platelet-Rich Fibrin/chemistry , Regeneration , Titanium/chemistry , Treatment Outcome
20.
Int J Implant Dent ; 8(1): 36, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36098849

ABSTRACT

PURPOSE: Platelet-rich fibrin (PRF) has been proposed as promising biomaterials with the advantages of host accumulation of platelets and leukocytes with entrapment of growth factors and fibrin scaffold. However, limitations including fast resorption rate (~ 2 weeks) restricts its clinical application. Recent studies have demonstrated heating treatment can prolong PRF degradation. Current published articles used the method of 75 °C for 10 min to obtain longer degradation, while few studies investigated the most suitable temperature for heating horizontal PRF. Our present study was to discover and confirm the optimum temperature for heat treatment before obtaining H-PRF gels by investigating their structure, mechanical properties, and bioactivity of the H-PRF gels after heating treatment. METHODS: In the present study, 2-mL upper layer of horizontal PRF was collected and heated at 45 °C, 60 °C, 75 °C, and 90 °C to heat 2-mL upper layer of horizontal PRF for 10 min before mixing with the 2-mL lower layer horizontal PRF. The weight, solidification time and the degradation properties were subsequently recorded. Scanning electron microscopy (SEM) and rheologic tests were carried out to investigate the microstructure and rheologic properties of each H-PRF gel. The biological activity of each H-PRF gel was also evaluated using live/dead staining. RESULTS: H-PRF gel prepared at 75 °C for 10 min had the fast solidification period (over a tenfold increase than control) as well as the best resistance to degradation. The number of living cells in H-PRF gel is greater than 90%. SEM showed that H-PRF gel becomes denser as the heating temperature increases, and rheologic tests also revealed that the heat treatment improved the mechanical properties of H-PRF gels when compared to non-heated control group. Future clinical studies are needed to further support the clinical application of H-PRF gels in tissue regeneration procedures. CONCLUSIONS: Our results demonstrated that the H-PRF gel obtained at 75 °C for 10 min could produce a uniform, moldable gel with a short time for solidification time, great rheologic behavior and, high percent of live cells in PRF gel. A promising use of the commonly utilized PRF gel was achieved facilitating tissue regeneration and preventing degradation.


Subject(s)
Fibrin , Platelet-Rich Fibrin , Blood Platelets , Fibrin/analysis , Gels/analysis , Heating , Platelet-Rich Fibrin/chemistry , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL