1
|
Sun J, Jiang X, Luo J, Zhao L, Xu Z, Xiao W. Effect of platelet-derived growth factor (PDGF-BB) and bone morphogenic protein 2 (BMP-2) transfection of rBMSCs compounded with platelet-rich plasma on adipogenic differentiation. ACTA ACUST UNITED AC 2020; 54:e9944. [PMID: 33331538 PMCID: PMC7727098 DOI: 10.1590/1414-431x20209944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/21/2020] [Indexed: 01/08/2023]
Abstract
The aim of this study was to inhibit adipogenic differentiation by transfecting two growth factors, platelet-derived growth factor (PDGF-BB) and bone morphogenic protein 2 (BMP-2), into modified rat bone marrow mesenchymal stem cells (rBMSCs) and then compounded with platelet-rich plasma (PRP). To achieve rBMSCs, the osteoporosis model of rats was established, and then the rBMSCs from the rats were isolated and identified. Co-transfection of rBMSCs with PDGF-BB-GFP and BMP-2 and detection of PDGF-BB/BMP-2 expression in transfected BMSCs was assessed by qRT-PCR and western blot, respectively. Moreover, the effect of the two growth factors transfection of rBMSCs on adipogenic differentiation was evaluated by oil red O staining and western blot, respectively. Finally, construction of the two growth factors transfection of rBMSCs compounded with PRP and detection of adipogenic differentiation were assessed by oil red O staining, CCK-8, and western blot, respectively. In vitro studies revealed that the two growth factors transfection of rBMSCs compounded with PRP promoted cell viability and inhibited adipogenic differentiation and could be promising for inhibiting adipogenic differentiation.
Collapse
Affiliation(s)
- Jin Sun
- Department of Orthopedics, Huizhou Third People's Hospital, Guangzhou Medical University, Huizhou, Guangdong, China
| | - Xin Jiang
- Department of Orthopedics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Junnan Luo
- Department of Orthopedics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Liheng Zhao
- Department of Orthopedics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Zuhua Xu
- Department of Orthopedics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Wende Xiao
- Department of Orthopedics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Wong CC, Liao JH, Sheu SY, Lin PY, Chen CH, Kuo TF. Novel transplant of combined platelet-rich fibrin Releasate and bone marrow stem cells prevent bone loss in Ovariectomized osteoporotic mice. BMC Musculoskelet Disord 2020; 21:527. [PMID: 32770974 PMCID: PMC7415181 DOI: 10.1186/s12891-020-03549-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/30/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Osteoporosis is a metabolic bone disorder characterized by deterioration in the quantity and quality of bone tissue, with a consequent increase susceptibility to fracture. METHODS In this study, we sought to determine the efficacy of platelet-rich fibrin releasates (PRFr) in augmenting the therapeutic effects of stem cell-based therapy in treating osteoporotic bone disorder. An osteoporosis mouse model was established through bilateral ovariectomy on 12-week-old female ICR (Institute of Cancer Research) mice. Eight weeks postoperatively, the ovariectomized (OVX) mice were left untreated (control) or injected with PRFr, bone marrow stem cells (BMSCs), or the combination of BMSCs and PRFr. Two different injection (single versus quadruple) dosages were tested to investigate the accumulative effects of BMSCS and PRFr on bone quality. Eight weeks after injection, the changes in tibial microstructural profiles included the percentage of bone volume versus total tissue volume (BV/TV, %), bone mineral density (BMD, g/cm3), trabecular number (Tb.N, number/mm), and trabecular separation (Tb.Sp, mm) and bony histology were analyzed. RESULTS Postmenopausal osteoporosis model was successfully established in OVX mice, evidenced by reduced BMD, decreased BV/TV, lower Tb.N but increased Tb.Sp. Eight weeks after injection, there was no significant change to BMD and bone trabeculae could be detected in mice that received single-injection regimen. In contrast, in mice which received 4 doses of combined PRFr and BMSCs, the BMD, BV/TV, and TB.N increased, and the TB.Sp decreased significantly compared to untreated OVX mice. Moreover, the histological analysis showed the trabecular spacing become narrower in OVX-mice treated with quadruple injection of BMSCs and combined PRFr and BMSCs than untreated control. CONCLUSION The systemic administration of combined BMSCs and PRFr protected against OVX-induced bone mass loss in mice. Moreover, the improvement of bony profile scores in quadruple-injection group is better than the single-injection group, probably through the increase in effect size of cells and growth factors. Our data also revealed the combination therapy of BMSCs and PRFr has better effect in enhancing osteogenesis, which may provide insight for the development of a novel therapeutic strategy in osteoporosis treatment.
Collapse
Affiliation(s)
- Chin-Chean Wong
- Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan.,Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.,Research Center of Biomedical Devices, Taipei Medical University, Taipei, 11031, Taiwan.,International Ph.D. Program for Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.,Non-invasive Cancer Therapy Research Institute of Taiwan, Taipei, 10489, Taiwan
| | - Jeng-Hao Liao
- School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Shi-Yuan Sheu
- School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung, 84001, Taiwan. .,Department of Chinese Medicine, E-Da Cancer Hospital, Kaohsiung, 84001, Taiwan.
| | - Po-Yu Lin
- School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Chih-Hwa Chen
- Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan.,Research Center of Biomedical Devices, Taipei Medical University, Taipei, 11031, Taiwan.,School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan.,School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Tzong-Fu Kuo
- School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan. .,Department of Post-Baccalaureate Veterinary Medicine, Asia University, Taichung, 41354, Taiwan.
| |
Collapse
|
3
|
Salamanna F, Maglio M, Sartori M, Tschon M, Fini M. Platelet Features and Derivatives in Osteoporosis: A Rational and Systematic Review on the Best Evidence. Int J Mol Sci 2020; 21:ijms21051762. [PMID: 32143494 PMCID: PMC7084230 DOI: 10.3390/ijms21051762] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023] Open
Abstract
Background: With the increase in aging population, the rising prevalence of osteoporosis (OP) has become an important medical issue. Accumulating evidence showed a close relationship between OP and hematopoiesis and emerging proofs revealed that platelets (PLTs), unique blood elements, rich in growth factors (GFs), play a critical role in bone remodeling. The aim of this review was to evaluate how PLT features, size, volume, bioactive GFs released, existing GFs in PLTs and PLT derivatives change and behave during OP. Methods: A systematic search was carried out in PubMed, Scopus, Web of Science Core Collection and Cochrane Central Register of Controlled Trials databases to identify preclinical and clinical studies in the last 10 years on PLT function/features and growth factor in PLTs and on PLT derivatives during OP. The methodological quality of included studies was assessed by QUIPS tool for assessing risk of bias in the clinical studies and by the SYRCLE tool for assessing risk of bias in animal studies. Results: In the initial search, 2761 studies were obtained, only 47 articles were submitted to complete reading, and 23 articles were selected for the analysis, 13 on PLT function/features and growth factor in PLTs and 10 on PLT derivatives. Risk of bias of almost all animal studies was high, while the in the clinical studies risk of bias was prevalently moderate/low for the most of the studies. The majority of the evaluated studies highlighted a positive correlation between PLT size/volume and bone mineralization and an improvement in bone regeneration ability by using PLTs bioactive GFs and PLT derivatives. Conclusions: The application of PLT features as OP markers and of PLT-derived compounds as therapeutic approach to promote bone healing during OP need to be further confirmed to provide clear evidence for the real efficacy of these interventions and to contribute to the clinical translation.
Collapse
|
4
|
Agostini F, Rossi FM, Aldinucci D, Battiston M, Lombardi E, Zanolin S, Massarut S, Parodi PC, Da Ponte A, Tessitori G, Pivetta B, Durante C, Mazzucato M. Improved GMP compliant approach to manipulate lipoaspirates, to cryopreserve stromal vascular fraction, and to expand adipose stem cells in xeno-free media. Stem Cell Res Ther 2018; 9:130. [PMID: 29751821 PMCID: PMC5948766 DOI: 10.1186/s13287-018-0886-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/05/2018] [Accepted: 04/23/2018] [Indexed: 02/08/2023] Open
Abstract
Background The stromal vascular fraction (SVF) derived from adipose tissue contains adipose-derived stromal/stem cells (ASC) and can be used for regenerative applications. Thus, a validated protocol for SVF isolation, freezing, and thawing is required to manage product administration. To comply with Good Manufacturing Practice (GMP), fetal bovine serum (FBS), used to expand ASC in vitro, could be replaced by growth factors from platelet concentrates. Methods Throughout each protocol, GMP-compliant reagents and devices were used. SVF cells were isolated from lipoaspirates by a standardized enzymatic protocol. Cells were cryopreserved in solutions containing different albumin or serum and dimethylsulfoxide (DMSO) concentrations. Before and after cryopreservation, we analyzed: cell viability (by Trypan blue); immunophenotype (by flow cytometry); colony-forming unit-fibroblast (CFU-F) formation; and differentiation potential. ASC, seeded at different densities, were expanded in presence of 10% FBS or 5% supernatant rich in growth factors (SRGF) from platelets. The differentiation potential and cell transformation grade were tested in expanded ASC. Results We demonstrated that SVF can be obtained with a consistent yield (about 185 × 103 cells/ml lipoaspirate) and viability (about 82%). Lipoaspirate manipulation after overnight storage at +4 °C reduced cell viability (−11.6%). The relative abundance of ASC (CD34+CD45−CD31–) and endothelial precursors (CD34+CD45−CD31+) in the SVF product was about 59% and 42%, respectively. A period of 2 months cryostorage in autologous serum with added DMSO minimally affected post-thaw SVF cell viability as well as clonogenic and differentiation potentials. Viability was negatively affected when SVF was frozen at a cell concentration below 1.3 × 106 cells/ml. Cell viability was not significantly affected after a freezing period of 1 year. Independent of seeding density, ASC cultured in 5% SRGF exhibited higher growth rates when compared with 10% FBS. ASC expanded in both media showed unaltered identity (by flow cytometry) and were exempt from genetic lesions. Both 5% SRGF- and 10% FBS-expanded ASC efficiently differentiated to adipocytes, osteocytes, and chondrocytes. Conclusions This paper reports a GMP-compliant approach for freezing SVF cells isolated from adipose tissue by a standardized protocol. Moreover, an ASC expansion method in controlled culture conditions and without involvement of animal-derived additives was reported.
Collapse
Affiliation(s)
| | - Francesca Maria Rossi
- Clinical-Experimental Onco-Hematology Unit, CRO Aviano National Cancer Institute, Aviano, PN, Italy
| | - Donatella Aldinucci
- Molecular Oncology Unit, CRO Aviano National Cancer Institute, Aviano, PN, Italy
| | - Monica Battiston
- Stem Cell Unit, CRO Aviano National Cancer Institute, Aviano, PN, Italy
| | | | - Stefania Zanolin
- Stem Cell Unit, CRO Aviano National Cancer Institute, Aviano, PN, Italy
| | - Samuele Massarut
- Breast Surgery Unit; CRO Aviano National Cancer Institute, Aviano, PN, Italy
| | - Pier Camillo Parodi
- Department of Plastic and Reconstructive Surgery, University of Udine, Udine, Italy
| | | | - Giovanni Tessitori
- Cytogenetic Unit, AAS 5 Friuli Occidentale, "S. Maria degli Angeli" Hospital, Pordenone, Italy
| | - Barbara Pivetta
- Cytogenetic Unit, AAS 5 Friuli Occidentale, "S. Maria degli Angeli" Hospital, Pordenone, Italy
| | - Cristina Durante
- Stem Cell Unit, CRO Aviano National Cancer Institute, Aviano, PN, Italy
| | - Mario Mazzucato
- Stem Cell Unit, CRO Aviano National Cancer Institute, Aviano, PN, Italy.
| |
Collapse
|
5
|
Turajane T, Chaveewanakorn U, Fongsarun W, Aojanepong J, Papadopoulos KI. Avoidance of Total Knee Arthroplasty in Early Osteoarthritis of the Knee with Intra-Articular Implantation of Autologous Activated Peripheral Blood Stem Cells versus Hyaluronic Acid: A Randomized Controlled Trial with Differential Effects of Growth Factor Addition. Stem Cells Int 2017; 2017:8925132. [PMID: 29056974 PMCID: PMC5625803 DOI: 10.1155/2017/8925132] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/25/2017] [Accepted: 08/24/2017] [Indexed: 02/08/2023] Open
Abstract
In this randomized controlled trial, in early osteoarthritis (OA) that failed conservative intervention, the need for total knee arthroplasty (TKA) and WOMAC scores were evaluated, following a combination of arthroscopic microdrilling mesenchymal cell stimulation (MCS) and repeated intra-articular (IA) autologous activated peripheral blood stem cells (AAPBSCs) with growth factor addition (GFA) and hyaluronic acid (HA) versus IA-HA alone. Leukapheresis-harvested AAPBSCs were administered as three weekly IA injections combined with HA and GFA (platelet-rich plasma [PRP] and granulocyte colony-stimulating factor [hG-CSF]) and MCS in group 1 and in group 2 but without hG-CSF while group 3 received IA-HA alone. Each group of 20 patients was evaluated at baseline and at 1, 6, and, 12 months. At 12 months, all patients in the AAPBSC groups were surgical intervention free compared to three patients needing TKA in group 3 (p < 0.033). Total WOMAC scores showed statistically significant improvements at 6 and 12 months for the AAPBSC groups versus controls. There were no notable adverse events. We have shown avoidance of TKA in the AAPBSC groups at 12 months and potent, early, and sustained symptom alleviation through GFA versus HA alone. Differential effects of hG-CSF were noted with an earlier onset of symptom alleviation throughout.
Collapse
Affiliation(s)
- Thana Turajane
- Department of Orthopedic Surgery, Police General Hospital, Bangkok, Thailand
| | | | | | - Jongjate Aojanepong
- Department of Gynecology and Obstetrics, Police General Hospital, Bangkok, Thailand
| | | |
Collapse
|
6
|
Otero L, Carrillo N, Calvo-Guirado J, Villamil J, Delgado-Ruíz R. Osteogenic potential of platelet-rich plasma in dental stem-cell cultures. Br J Oral Maxillofac Surg 2017. [DOI: 10.1016/j.bjoms.2017.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
7
|
Scioli MG, Bielli A, Gentile P, Cervelli V, Orlandi A. Combined treatment with platelet-rich plasma and insulin favours chondrogenic and osteogenic differentiation of human adipose-derived stem cells in three-dimensional collagen scaffolds. J Tissue Eng Regen Med 2017; 11:2398-2410. [PMID: 27074878 DOI: 10.1002/term.2139] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 11/18/2015] [Accepted: 12/10/2015] [Indexed: 02/06/2023]
Abstract
Osteochondral lesions due to injury or other pathology commonly result in the development of osteoarthritis and progressive joint destruction. Bioengineered scaffolds are widely studied for regenerative surgery strategies in osteochondral defect management, also combining the use of stem cells, growth factors and hormones. The utility in tissue engineering of human adipose-derived stem cells (ASCs) isolated from adipose tissue has been widely noted. Autologous platelet-rich plasma (PRP) represents an alternative strategy in regenerative medicine for the local release of endogenous growth factors and hormones. Here we compared the effects of three-dimensional (3D) collagen type I scaffold culture and combined treatment with PRP and human recombinant insulin on the chondro-/osteogenic differentiation of ASCs. Histochemical and biomolecular analyses demonstrated that chondro-/osteogenic differentiation was increased in ASC-populated 3D collagen scaffolds compared with two-dimensional (2D) plastic dish culture. Chondro-/osteogenic differentiation was further enhanced in the presence of combined PRP (5% v/v) and insulin (100 nm) treatment. In addition, chondro-/osteogenic differentiation associated with the contraction of ASC-populated 3D collagen scaffold and increased β1/β3-integrin expression. Inhibition studies demonstrated that PRP/insulin-induced chondro-/osteogenic differentiation is independent of insulin-like growth factor 1 receptor (IGF-1R) and mammalian target of rapamycin (mTOR) signalling; IGF-R1/mTOR inhibition even enhanced ASC chondro-/osteogenic differentiation. Our findings underline that 3D collagen scaffold culture in association with platelet-derived growth factors and insulin favour the chondro-/osteogenic differentiation of ASCs, suggesting new translational applications in regenerative medicine for the management of osteochondral defects. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Maria Giovanna Scioli
- Institute of Anatomical Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Alessandra Bielli
- Institute of Anatomical Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Pietro Gentile
- Plastic and Reconstructive Surgery, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Valerio Cervelli
- Plastic and Reconstructive Surgery, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Augusto Orlandi
- Institute of Anatomical Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| |
Collapse
|
8
|
Badran Z, Abdallah MN, Torres J, Tamimi F. Platelet concentrates for bone regeneration: Current evidence and future challenges. Platelets 2017. [DOI: 10.1080/09537104.2017.1327656] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Zahi Badran
- Department of Periodontology (CHU/Rmes Inserm U1229/UIC11), Faculty of Dental Surgery, University of Nantes, Nantes, France
- Division of Biomedical Sciences, Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Mohamed-Nur Abdallah
- Division of Biomedical Sciences, Faculty of Dentistry, McGill University, Montreal, QC, Canada
| | - Jesus Torres
- Faculty of Dentistry, Universidad Complutense, Madrid, Spain
| | - Faleh Tamimi
- Division of Biomedical Sciences, Faculty of Dentistry, McGill University, Montreal, QC, Canada
| |
Collapse
|
9
|
Samuel S, Ahmad RE, Ramasamy TS, Karunanithi P, Naveen SV, Murali MR, Abbas AA, Kamarul T. Platelet-rich concentrate in serum free medium enhances osteogenic differentiation of bone marrow-derived human mesenchymal stromal cells. PeerJ 2016; 4:e2347. [PMID: 27651984 PMCID: PMC5018671 DOI: 10.7717/peerj.2347] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/19/2016] [Indexed: 12/22/2022] Open
Abstract
Previous studies have shown that platelet concentrates used in conjunction with appropriate growth media enhance osteogenic differentiation of human mesenchymal stromal cells (hMSCs). However, their potential in inducing osteogenesis of hMSCs when cultured in serum free medium has not been explored. Furthermore, the resulting osteogenic molecular signatures of the hMSCs have not been compared to standard osteogenic medium. We studied the effect of infrequent supplementation (8-day interval) of 15% non-activated platelet-rich concentrate (PRC) in serum free medium on hMSCs proliferation and differentiation throughout a course of 24 days, and compared the effect with those cultured in a standard osteogenic medium (OM). Cell proliferation was analyzed by alamar blue assay. Gene expression of osteogenic markers (Runx2, Collagen1, Alkaline Phosphatase, Bone morphogenetic protein 2, Osteopontin, Osteocalcin, Osteonectin) were analyzed using Q-PCR. Immunocytochemical staining for osteocalcin, osteopontin and transcription factor Runx2 were done at 8, 16 and 24 days. Biochemical assays for the expression of ALP and osteocalcin were also performed at these time-points. Osteogenic differentiation was further confirmed qualitatively by Alizarin Red S staining that was quantified using cetylpyridinium chloride. Results showed that PRC supplemented in serum free medium enhanced hMSC proliferation, which peaked at day 16. The temporal pattern of gene expression of hMSCs under the influence of PRC was comparable to that of the osteogenic media, but at a greater extent at specific time points. Immunocytochemical staining revealed stronger staining for Runx2 in the PRC-treated group compared to OM, while the staining for Osteocalcin and Osteopontin were comparable in both groups. ALP activity and Osteocalcin/DNA level were higher in the PRC group. Cells in the PRC group had similar level of bone mineralization as those cultured in OM, as reflected by the intensity of Alizarin red stain. Collectively, these results demonstrate a great potential of PRC alone in inducing proliferation of hMSCs without any influence from other lineage-specific growth media. PRC alone has similar capacity to enhance hMSC osteogenic differentiation as a standard OM, without changing the temporal profile of the differentiation process. Thus, PRC could be used as a substitute medium to provide sufficient pool of pre-differentiated hMSCs for potential clinical application in bone regeneration.
Collapse
Affiliation(s)
- Shani Samuel
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Orthopedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Raja Elina Ahmad
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Thamil Selvee Ramasamy
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Puvanan Karunanithi
- Department of Orthopedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Malliga Raman Murali
- Department of Orthopedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Azlina A Abbas
- Department of Orthopedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Tunku Kamarul
- Department of Orthopedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
10
|
Malhotra R, Kumar V, Garg B, Singh R, Jain V, Coshic P, Chatterjee K. Role of autologous platelet-rich plasma in treatment of long-bone nonunions: a prospective study. Musculoskelet Surg 2015; 99:243-248. [PMID: 26193983 DOI: 10.1007/s12306-015-0378-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/08/2015] [Indexed: 06/04/2023]
Abstract
PURPOSE Fracture union is a complex biological process, which depends upon several systemic and local factors. Disturbance of any of these factors may lead to nonunion of the fracture. These nonunions have a huge impact on quality of life as well as socioeconomical aspects. The platelets on activation release a number of growth factors and differentiation factors, which play important role in fracture healing. This study aimed to look for efficacy of platelet-rich plasma in the treatment of established fracture nonunions of long bones. METHODS A total of 94 patients with established nonunion of long bone (35 tibia, 30 femur, 11 humerus, 4 radius, 12 ulna, 2 with both radius and ulna) were included in this study. We injected 15-20 ml of autologous platelet-rich plasma (>2,000,000 platelets/μl) under image intensifier at each nonunion site. The fracture union was evaluated clinically and radiologically regularly at monthly interval till 4 months. RESULTS Eighty-two patients had their fracture united at the end of 4 months. Thirty-four patients showed bridging trabeculae on X-rays at the end of 2 months, while 41 patients showed bridging trabeculae at the end of third month. Twelve patients did not show any attempt of union at 4 months and were labeled as failure of treatment. There were no complications. CONCLUSION Platelet-rich plasma is a safe and effective treatment for the treatment of nonunions. More studies are needed to look into molecular mechanism of this fracture healing acceleration by platelet-rich plasma.
Collapse
Affiliation(s)
- R Malhotra
- Department of Orthopedics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - V Kumar
- Department of Orthopedics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - B Garg
- Department of Orthopedics, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - R Singh
- Department of Orthopedics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - V Jain
- Department of Orthopedics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - P Coshic
- Department of Transfusion Medicine, AIIMS, New Delhi, India
| | - K Chatterjee
- Department of Transfusion Medicine, AIIMS, New Delhi, India
| |
Collapse
|
11
|
New and emerging strategies in platelet-rich plasma application in musculoskeletal regenerative procedures: general overview on still open questions and outlook. BIOMED RESEARCH INTERNATIONAL 2015; 2015:846045. [PMID: 26075269 PMCID: PMC4436449 DOI: 10.1155/2015/846045] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 01/09/2015] [Accepted: 01/13/2015] [Indexed: 02/07/2023]
Abstract
Despite its pervasive use, the clinical efficacy of platelet-rich plasma (PRP) therapy and the different mechanisms of action have yet to be established. This overview of the literature is focused on the role of PRP in bone, tendon, cartilage, and ligament tissue regeneration considering basic science literature deriving from in vitro and in vivo studies. Although this work provides evidence that numerous preclinical studies published within the last 10 years showed promising results concerning the application of PRP, many key questions remain unanswered and controversial results have arisen. Additional preclinical studies are needed to define the dosing, timing, and frequency of PRP injections, different techniques for delivery and location of delivery, optimal physiologic conditions for injections, and the concomitant use of recombinant proteins, cytokines, additional growth factors, biological scaffolds, and stems cells to develop optimal treatment protocols that can effectively treat various musculoskeletal conditions.
Collapse
|
12
|
Song Z, Wu C, Sun S, Li H, Wang D, Gong J, Yan Z. Quantitative analysis of factors influencing tissue-engineered bone formation by detecting the expression levels of alkaline phosphatase and bone γ-carboxyglutamate protein 2. Exp Ther Med 2015; 9:1097-1102. [PMID: 25780393 PMCID: PMC4353779 DOI: 10.3892/etm.2015.2259] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 11/17/2014] [Indexed: 11/06/2022] Open
Abstract
Bone tissue engineering is a promising alternative approach that permits the efficient reconstruction of bone defects. There are four elements involved in bone tissue engineering technology, including the seed cells, growth factors, scaffolds and culture environment. The aim of the present study was to evaluate the effect of these factors on bone formation in tissue engineering technology by analyzing the expression of osteogenetic markers using polymerase chain reaction (PCR). Bone marrow mesenchymal stem cells (BMSCs) were extracted from the bone marrow of the bilateral tibial platform of New Zealand white rabbits. In addition, platelet-rich plasma (PRP) samples were prepared from blood extracted from the ear vein of the rabbits. A perfusion bioreactor was used to provide the culture environment, and β-tricalcium phosphate (β-TCP) was used to build the scaffolds. The β-TCP scaffolds were divided into five groups and each group was treated with a different combination of the factors. Next, the composites were implanted into the rabbits. After three months, the expression levels of the new bone formation markers, alkaline phosphatase and bone γ-carboxyglutamate protein 2, were detected using quantitative reverse transcription-PCR analysis. The expression levels of the markers in the experimental groups were higher compared with the negative control group. Comparisons between the experimental groups also revealed statistical significance. Scanning electron microscopy revealed good adhesion and distribution of the BMSCs on the β-TCP scaffold. In conclusion, the PCR results indicated that PRP, BMSCs and the bioreactor exhibited a promoting effect on bone formation.
Collapse
Affiliation(s)
- Zezhong Song
- Joint Surgery Department, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Changshun Wu
- Joint Surgery Department, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Shui Sun
- Joint Surgery Department, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Huibo Li
- Joint Surgery Department, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Dong Wang
- Joint Surgery Department, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Jianbao Gong
- Joint Surgery Department, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Zexing Yan
- Joint Surgery Department, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
13
|
Platelet rich concentrate promotes early cellular proliferation and multiple lineage differentiation of human mesenchymal stromal cells in vitro. ScientificWorldJournal 2014; 2014:845293. [PMID: 25436230 PMCID: PMC4243129 DOI: 10.1155/2014/845293] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 09/15/2014] [Indexed: 01/17/2023] Open
Abstract
Platelet rich concentrate (PRC) is a natural adjuvant that aids in human mesenchymal stromal cell (hMSC) proliferation in vitro; however, its role requires further exploration. This study was conducted to determine the optimal concentration of PRC required for achieving the maximal proliferation, and the need for activating the platelets to achieve this effect, and if PRC could independently induce early differentiation of hMSC. The gene expression of markers for osteocytes (ALP, RUNX2), chondrocytes (SOX9, COL2A1), and adipocytes (PPAR-γ) was determined at each time point in hMSC treated with 15% activated and nonactivated PRC since maximal proliferative effect was achieved at this concentration. The isolated PRC had approximately fourfold higher platelet count than whole blood. There was no significant difference in hMSC proliferation between the activated and nonactivated PRC. Only RUNX2 and SOX9 genes were upregulated throughout the 8 days. However, protein expression study showed formation of oil globules from day 4, significant increase in ALP at days 6 and 8 (P ≤ 0.05), and increased glycosaminoglycan levels at all time points (P < 0.05), suggesting the early differentiation of hMSC into osteogenic and adipogenic lineages. This study demonstrates that the use of PRC increased hMSC proliferation and induced early differentiation of hMSC into multiple mesenchymal lineages, without preactivation or addition of differentiation medium.
Collapse
|
14
|
Turajane T, Thitiset T, Honsawek S, Chaveewanakorn U, Aojanepong J, Papadopoulos KI. Assessment of chondrogenic differentiation potential of autologous activated peripheral blood stem cells on human early osteoarthritic cancellous tibial bone scaffold. Musculoskelet Surg 2014; 98:35-43. [PMID: 24178764 DOI: 10.1007/s12306-013-0303-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 10/17/2013] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Current therapeutic regimens in osteoarthritis (OA) address mainly pain but not the slow progressive degradation of the extracellular matrix (ECM) and the loss of a chondrogenic phenotype in articular cartilage. In the present study, using an early OA cancellous bone scaffold, we aimed to uncover evidence of the successful hyaline cartilage regenerative capacity of autologous human granulocyte colony-stimulating factor (hG-CSF)-activated peripheral blood stem cells (AAPBSC) with growth factor addition. MATERIALS AND METHODS AAPBSC were harvested in ten patients (median age 58 years, 8 females), and flow cytometry was performed for cell surface markers. Arthroscopically obtained cancellous bone scaffold specimens were seeded with AAPBSC. In Group 1, the scaffold was seeded with AAPBSC only, in Group 2, AAPBSC plus hyaluronic acid (HA), and in Group 3, AAPBSC plus HA, hG-CSF, and double-centrifuged platelet-rich plasma (PRP). The specimens were analyzed for cell attachment and proliferation by the fluorometric quantification of cellular DNA assay and scanning electron microscopy. Chondrogenic gene expression was determined by reverse transcriptase-polymerase chain reaction (RT-PCR) of Sox9, collagen type II (COL-2), and aggrecan. Histological sections of scaffold constructs for cartilaginous matrix formation were stained with toluidine blue (proteoglycan) and safranin O (sGAG) after 3 weeks. RESULTS AAPBSC displayed especially high levels of CD29 and CD44 surface markers, as well as CD90, and CD105, while only a small proportion expressed CD34. Almost half of the seeded cells attached on the bone scaffolds in all three groups (not statistically significant), whereas the means of cell proliferation on day 7 compared to day 1 were statistically significant difference with the order of increase as group 3 > group 2 > group 1. RT-PCR showed statistically significant sequential increases in Sox9, COL-2, and Aggrecan all being highest in group 3. Histological analysis demonstrated cells in the cancellous bone scaffold with a round morphology, and ECM was positively stained by toluidine blue and safranin O indicating increased proteoglycan and glycosaminoglycan content, respectively, in the newly formed cartilage matrix. CONCLUSIONS AAPBSC initiated chondrocyte differentiation on an autologous cancellous bone scaffold, and the addition of PRP and hG-CSF further stimulated cell proliferation toward a chondrocyte phenotype with potentiated Sox9 transcription resulting in sequential COL-2 and aggrecan mRNA increases that ultimately resulted in histologically confirmed increased proteoglycan and glucosaminoglycan content in newly formed hyaline cartilage.
Collapse
Affiliation(s)
- T Turajane
- Department of Orthopedic Surgery, Stem Cell Research and Treatment Center, Police General Hospital, Bangkok, Thailand
| | | | | | | | | | | |
Collapse
|
15
|
Machova Urdzikova L, Sedlacek R, Suchy T, Amemori T, Ruzicka J, Lesny P, Havlas V, Sykova E, Jendelova P. Human multipotent mesenchymal stem cells improve healing after collagenase tendon injury in the rat. Biomed Eng Online 2014; 13:42. [PMID: 24712305 PMCID: PMC4001357 DOI: 10.1186/1475-925x-13-42] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 04/01/2014] [Indexed: 01/30/2023] Open
Abstract
Background Mesenchymal stromal cells attract much interest in tissue regeneration because of their capacity to differentiate into mesodermal origin cells, their paracrine properties and their possible use in autologous transplantations. The aim of this study was to investigate the safety and reparative potential of implanted human mesenchymal stromal cells (hMSCs), prepared under Good Manufacturing Practice (GMP) conditions utilizing human mixed platelet lysate as a culture supplement, in a collagenase Achilles tendon injury model in rats. Methods Eighty-one rats with collagenase-induced injury were divided into two groups. The first group received human mesenchymal stromal cells injected into the site of injury 3 days after lesion induction, while the second group received saline. Biomechanical testing, morphometry and semiquantitative immunohistochemistry of collagens I, II and III, versican and aggrecan, neovascularization, and hMSC survival were performed 2, 4, and 6 weeks after injury. Results Human mesenchymal stromal cell-treated rats had a significantly better extracellular matrix structure and a larger amount of collagen I and collagen III. Neovascularization was also increased in hMSC-treated rats 2 and 4 weeks after tendon injury. MTCO2 (Cytochrome c oxidase subunit II) positivity confirmed the presence of hMSCs 2, 4 and 6 weeks after transplantation. Collagen II deposits and alizarin red staining for bone were found in 6 hMSC- and 2 saline-treated tendons 6 weeks after injury. The intensity of anti-versican and anti-aggrecan staining did not differ between the groups. Conclusions hMSCs can support tendon healing through better vascularization as well as through larger deposits and better organization of the extracellular matrix. The treatment procedure was found to be safe; however, cartilage and bone formation at the implantation site should be taken into account when planning subsequent in vivo and clinical trials on tendinopathy as an expected adverse event.
Collapse
Affiliation(s)
- Lucia Machova Urdzikova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Elder S, Thomason J. Effect of platelet-rich plasma on chondrogenic differentiation in three-dimensional culture. Open Orthop J 2014; 8:78-84. [PMID: 24843389 PMCID: PMC4023405 DOI: 10.2174/1874325001408010078] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 04/16/2014] [Accepted: 04/20/2014] [Indexed: 02/01/2023] Open
Abstract
Platelet-rich plasma (PRP) may have the potential to enhance articular cartilage regeneration through release of
growth factors including transforming growth factor isoforms. The purpose of this study was to investigate the potential
for PRP to stimulate chondrogenic differentiation in three-dimensional PRP hydrogel constructs. Allogenic PRP was
prepared using a double centrifugation protocol which resulted in a platelet concentration approximately 250% above
baseline. Canine marrow stromal cells were encapsulated at 6.8×106 cells/ml in either 2% sodium alginate or in a 3:1
mixture of freshly prepared PRP and 2% alginate. PRP and alginate beads were cultured in chemically defined
chondrogenic medium with and without 10 ng/ml TGF-β3. PRP cultures were additionally supplemented with frozen-thawed
PRP. In the absence of TGF-β3, PRP had a mild stimulatory effect on cell proliferation. PRP did not stimulate cell
proliferation in the presence of TGF-β3. Cells exposed to TGF-β3 accumulated significantly more GAG/DNA than those
which were not, but there was not a statistically significant difference between alginate and PRP. Total collagen content
was greater in PRP than in alginate, regardless of TGF-β3. Chondrogenesis in PRP was qualitatively and spatially
different than that which occurred in conventional alginate beads and was characterized by isolated centers of intense
chondrogenesis. Overall the results demonstrate that PRP alone weakly promotes chondroinduction of marrow stromal
cells, and the effect is greatly augmented by TGF-β3.
Collapse
Affiliation(s)
- Steven Elder
- Department of Agricultural & Biological Engineering, Bagley College of Engineering, Mississippi State University, Starkville, MS, USA
| | - John Thomason
- Department of Clinical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS, USA
| |
Collapse
|
17
|
Li H, Usas A, Poddar M, Chen CW, Thompson S, Ahani B, Cummins J, Lavasani M, Huard J. Platelet-rich plasma promotes the proliferation of human muscle derived progenitor cells and maintains their stemness. PLoS One 2013; 8:e64923. [PMID: 23762264 PMCID: PMC3676442 DOI: 10.1371/journal.pone.0064923] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 04/20/2013] [Indexed: 01/01/2023] Open
Abstract
Human muscle-derived progenitor cells (hMDPCs) offer great promise for muscle cell-based regenerative medicine; however, prolonged ex-vivo expansion using animal sera is necessary to acquire sufficient cells for transplantation. Due to the risks associated with the use of animal sera, the development of a strategy for the ex vivo expansion of hMDPCs is required. The purpose of this study was to investigate the efficacy of using platelet-rich plasma (PRP) for the ex-vivo expansion of hMDPCs. Pre-plated MDPCs, myoendothelial cells, and pericytes are three populations of hMDPCs that we isolated by the modified pre-plate technique and Fluorescence Activated Cell Sorting (FACS), respectively. Pooled allogeneic human PRP was obtained from a local blood bank, and the effect that thrombin-activated PRP-releasate supplemented media had on the ex-vivo expansion of the hMDPCs was tested against FBS supplemented media, both in vitro and in vivo. PRP significantly enhanced short and long-term cell proliferation, with or without FBS supplementation. Antibody-neutralization of PDGF significantly blocked the mitogenic/proliferative effects that PRP had on the hMDPCs. A more stable and sustained expression of markers associated with stemness, and a decreased expression of lineage specific markers was observed in the PRP-expanded cells when compared with the FBS-expanded cells. The in vitro osteogenic, chondrogenic, and myogenic differentiation capacities of the hMDPCs were not altered when expanded in media supplemented with PRP. All populations of hMDPCs that were expanded in PRP supplemented media retained their ability to regenerate myofibers in vivo. Our data demonstrated that PRP promoted the proliferation and maintained the multi-differentiation capacities of the hMDPCs during ex-vivo expansion by maintaining the cells in an undifferentiated state. Moreover, PDGF appears to be a key contributing factor to the beneficial effect that PRP has on the proliferation of hMDPCs.
Collapse
Affiliation(s)
- Hongshuai Li
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Arvydas Usas
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Minakshi Poddar
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Chien-Wen Chen
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Seth Thompson
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Bahar Ahani
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - James Cummins
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mitra Lavasani
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Johnny Huard
- Department of Orthopedic Surgery, Stem Cell Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
18
|
Odri GA, Hami A, Pomero V, Seite M, Heymann D, Bertrand-Vasseur A, Skalli W, Delecrin J. Development of a per-operative procedure for concentrated bone marrow adjunction in postero-lateral lumbar fusion: radiological, biological and clinical assessment. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2012; 21:2665-72. [PMID: 22639299 DOI: 10.1007/s00586-012-2375-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 04/07/2012] [Accepted: 05/10/2012] [Indexed: 12/13/2022]
Abstract
PURPOSE Addition of bone marrow to the bone graft in the postero-lateral lumbar arthrodesis is a widely used technique. Bone marrow brings stem cells and growth factors contained in the platelets, favorable for bone growth. Adjunction of concentrated bone marrow should create better conditions and may increase bone growth. METHODS Simple blind randomized clinical, prospective, monocentric trial was conducted. Fifteen patients underwent lumbar arthrodesis. During surgery, a fraction of the bone marrow harvested was centrifuged. One side received this concentrate with autologous bone and ceramics; the other side received the same graft with unconcentrated bone marrow. A quantitative study, realised with a volume calculating software on CT-scan images, determined the cortical bone volume in the graft post-operatively and at 3 months. The osteoprogenitor cells, nucleated cells and platelet concentrations were determined. RESULTS The biological study found an average concentration of six times for the nucleated cells, 3.5 times for the platelets and 2.2 times for the osteoprogenitor cells. The comparison of the mean cortical bone volumes post-operatively and at 3 months was not significantly different. CONCLUSIONS Despite the concentration obtained, there was no increase of bone growth by adding concentrated bone marrow. However, the number of stem cells in bone marrow was low and maybe a stronger concentration is needed to obtain a difference. The 3D reconstruction of the graft and the analysis of the graft's volume using a novel software was efficient according to the similarity of the graft's volume post-operatively in all patients.
Collapse
Affiliation(s)
- G A Odri
- Clinique Chirurgicale Orthopédique et Traumatologique, CHU Hôtel Dieu, 1 Place Alexis Ricordeau, 44093, Nantes Cedex 1, France.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Murphy MB, Blashki D, Buchanan RM, Yazdi IK, Ferrari M, Simmons PJ, Tasciotti E. Adult and umbilical cord blood-derived platelet-rich plasma for mesenchymal stem cell proliferation, chemotaxis, and cryo-preservation. Biomaterials 2012; 33:5308-16. [PMID: 22542609 DOI: 10.1016/j.biomaterials.2012.04.007] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 04/01/2012] [Indexed: 02/07/2023]
Abstract
Platelet-rich plasma (PRP) was prepared from human adult peripheral blood and from human umbilical cord (uc) blood and the properties were compared in a series of in vitro bioassays. Quantification of growth factors in PRP and platelet-poor plasma (PPP) fractions revealed increased levels of mitogenic growth factors PDGF-AB, PDGF-BB, and FGF-2, the angiogenic agent VEGF and the chemokine RANTES in ucPRP compared to adult PRP (aPRP) and PPP. To compare the ability of the various PRP products to stimulate proliferation of human bone marrow (BM), rat BM and compact bone (CB)-derived mesenchymal stem cells (MSC), cells were cultured in serum-free media for 4 and 7 days with varying concentrations of PRP, PPP, or combinations of recombinant mitogens. It was found that while all forms of PRP and PPP were more mitogenic than fetal bovine serum, ucPRP resulted in significantly higher proliferation by 7 days than adult PRP and PPP. We observed that addition of as little as 0.1% ucPRP caused greater proliferation of MSC effects than the most potent combination of recombinant growth factors tested, namely PDGF-AB + PDGF-BB + FGF-2, each at 10 ng/mL. Similarly, in chemotaxis assays, ucPRP showed greater potency than adult PRP, PPP from either source, or indeed than combinations of either recombinant growth factors (PDGF, FGF, and TGF-β1) or chemokines previously shown to stimulate chemotactic migration of MSC. Lastly, we successfully demonstrated that PRP and PPP represented a viable alternative to FBS containing media for the cryo-preservation of MSC from human and rat BM.
Collapse
Affiliation(s)
- Matthew B Murphy
- Department of Nanomedicine, The Methodist Hospital Research Institute, Houston, TX 77025, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Blackburn J, Mansell JP. The emerging role of lysophosphatidic acid (LPA) in skeletal biology. Bone 2012; 50:756-62. [PMID: 22193551 DOI: 10.1016/j.bone.2011.12.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 12/01/2011] [Accepted: 12/03/2011] [Indexed: 11/22/2022]
Abstract
Lysophosphatidic acid (LPA) is the simplest signalling lipid eliciting pleiotropic actions upon most mammalian cell types. Although LPA has an established role in many biological processes, particularly wound healing and cancer, the participation of LPA in skeletal biology is just beginning to emerge. Early studies, identified in this review, gave a solid indication that LPA, via binding to one of several cell surface receptors, activated multiple intracellular systems culminating in altered cell morphology, growth, motility and survival. More recently the ablation of murine LPA1 and 4 receptors implies that this lipid has a role in skeletal development and post natal bone accrual. Greater understanding of the ability of LPA to influence, for example, osteoblast growth, maturation and survival could be advantageous in developing novel strategies aimed at improving skeletal tissue repair and regeneration. Herein this review provides an insight into the diversity of studies exploring the actions of a small lipid on those major cell types key to skeletal tissue health and homeostasis.
Collapse
Affiliation(s)
- Julia Blackburn
- Musculoskeletal Research Unit, Avon Orthopaedic Centre, Southmead Hospital, Westbury-on-Trym, Bristol, BS10 5NB, UK
| | | |
Collapse
|
21
|
Hong J, Kurt S, Thor A. A Hydrophilic Dental Implant Surface Exhibit Thrombogenic Properties In Vitro. Clin Implant Dent Relat Res 2011; 15:105-112. [DOI: 10.1111/j.1708-8208.2011.00362.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jaan Hong
- Researcher, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory C5, Uppsala University, Uppsala, Sweden
| | - Seta Kurt
- research engineer, Department of Oncology, Radiology and Clinical Immunology, Division of Clinical Immunology, Rudbeck Laboratory C5, Uppsala University, Uppsala, Sweden,
| | - Andreas Thor
- consultant and researcher, Institute of Surgical Sciences, Department of Oral & Maxillofacial Surgery, Uppsala University, Uppsala, Sweden
| |
Collapse
|
22
|
Schroeder JE, Mosheiff R. Tissue engineering approaches for bone repair: concepts and evidence. Injury 2011; 42:609-13. [PMID: 21489529 DOI: 10.1016/j.injury.2011.03.029] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 03/17/2011] [Indexed: 02/02/2023]
Abstract
Over the last decades, the medical world has advanced dramatically in the understanding of fracture repair. The three components needed for fracture healing are osteoconduction, osteoinduction and osteogenesis. With newly designed scaffolds, ex vivo produced growth factors and isolated stem cells, most of the challenges of critical size bone defects have been resolved in vitro, and in some cases in animal models as well. However, there are still challenges needed to be overcome before these technologies can be fully converted from the bench to the bedside. These technological and biological advancements need to be converted to mass production of affordable products that can be used in every part of the world. Vascularity, full substation of scaffolds by native bone, and bio-safety are the three most critical steps to be challenged before reaching the clinical setting.
Collapse
Affiliation(s)
- Josh E Schroeder
- Orthopedic Surgery Department, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | |
Collapse
|
23
|
Yu YY, Lieu S, Lu C, Colnot C. Bone morphogenetic protein 2 stimulates endochondral ossification by regulating periosteal cell fate during bone repair. Bone 2010; 47:65-73. [PMID: 20348041 PMCID: PMC2891074 DOI: 10.1016/j.bone.2010.03.012] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Revised: 03/19/2010] [Accepted: 03/22/2010] [Indexed: 10/19/2022]
Abstract
Bone repair depends on the coordinated action of numerous growth factors and cytokines to stimulate new skeletal tissue formation. Among all the growth factors involved in bone repair, Bone Morphogenetic Proteins (BMPs) are the only molecules now used therapeutically to enhance healing. Although BMPs are known as strong bone inducers, their role in initiating skeletal repair is not entirely elucidated. The aim of this study was to define the role of BMP2 during the early stages of bone regeneration and more specifically in regulating the fate of skeletal progenitors. During healing of non-stabilized fractures via endochondral ossification, exogenous BMP2 increased the deposition and resorption of cartilage and bone, which was correlated with a stimulation of osteoclastogenesis but not angiogenesis in the early phase of repair. During healing of stabilized fractures, which normally occurs via intramembranous ossification, exogenous BMP2 induced cartilage formation suggesting a role in regulating cell fate decisions. Specifically, the periosteum was found to be a target of exogenous BMP2 as shown by activation of the BMP pathway in this tissue. Using cell lineage analyses, we further show that BMP2 can direct cell differentiation towards the chondrogenic lineage within the periosteum but not the endosteum, indicating that skeletal progenitors within periosteum and endosteum respond differently to BMP signals. In conclusion, BMP2 plays an important role in the early stages of repair by recruiting local sources of skeletal progenitors within periosteum and endosteum and by determining their differentiation towards the chondrogenic and osteogenic lineages.
Collapse
|
24
|
Felka T, Schäfer R, De Zwart P, Aicher WK. Animal serum-free expansion and differentiation of human mesenchymal stromal cells. Cytotherapy 2010; 12:143-53. [PMID: 20141338 DOI: 10.3109/14653240903470647] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSC) are attracting increasing interest for possible application in cell therapies. Fetal calf serum (FCS) is widely utilized for cell culture, but its use in the context of clinical applications is associated with too many risks. Therefore we tested FCS-free media for the expansion and differentiation of MSC in compliance with the European good manufacturing practice (GMP) regulations for medicinal products. METHODS MSC expansion medium was modified by replacing FCS with human plasma and platelet extract. Cells were characterized according to the defined minimal criteria for multipotent MSC. For chondrogenic differentiation, serum-free micromass cultures were employed. For adipogenic and osteogenic differentiation, the FCS was replaced by human plasma. After 28 days of incubation in differentiation media, cells were analyzed by cytochemical and immunohistochemical staining. Furthermore, mRNA expression of chondrogenic, adipogenic and osteogenic markers was investigated by quantitative reverse-transcription polymerase chain reaction (qRT-PCR). RESULTS Expansion and differentiation of MSC under FCS-free conditions yielded cells with chondrogenic, adipogenic and osteogenic phenotypes and a characteristic gene expression. Chondrocytes in micromass pellets revealed an accumulation of proteoglycans and type II collagen as well as a significantly increased mRNA expression of chondrogenic marker genes. The adipocytes displayed Oil red O staining and expressed peroxisome proliferator-activated receptor gamma(2) (ppARgamma2) and lipoprotein lipase (LPL) mRNA. The osteoblasts were positive for von Kossa staining and expressed mRNA of osteogenic marker genes. The results did not indicate any spontaneous differentiation. CONCLUSIONS Human plasma is a suitable FCS replacement for the expansion and differentiation of MSC, providing a feasible alternative for tissue engineering with GMP-compatible protocols.
Collapse
Affiliation(s)
- Tino Felka
- Center for Medical Research (ZMF), University of Tübingen Medical Center (UKT), 72072 Tübingen, Germany
| | | | | | | |
Collapse
|
25
|
Gociman B, Agko M, Agk M, Moran SL. Caption: a filtration-based platelet concentration system. Expert Rev Med Devices 2009; 6:607-10. [PMID: 19911872 DOI: 10.1586/erd.09.38] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Despite significant advances, wound management remains a medical challenge and a major health-expense problem. Currently, a wide range of therapeutic interventions are available for wound care. This is only partially explained by the variability in the presenting clinical situation and shows that there is no panacea for this complex medical problem. Over time, a multitude of interventions have been proposed as solutions for faster and more effective healing; however, only a few are still part of the wound care armamentarium today. Based on the fact that platelets contain a high concentration of growth factors, the application of platelet-enriched plasma over wound sites was recently introduced as a novel therapeutic modality. As increasing evidence started to validate its benefit, several devices for platelet-rich plasma preparation became available. Here, we present the most recently introduced platelet concentration system and provide insights on the current and possible future applications of this treatment modality.
Collapse
Affiliation(s)
- Barbu Gociman
- Department of Surgery, The University of Toledo Health Science Campus, 3000 Arlington Avenue, Toledo, OH 43614, USA
| | | | | | | |
Collapse
|
26
|
Interaction of platelet-rich concentrate with bone graft materials: an in vitro study. J Orthop Trauma 2009; 23:195-200; discussion 201-2. [PMID: 19516093 DOI: 10.1097/bot.0b013e31819b35db] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Platelet-rich concentrate (PRC) is in routine use for orthopaedic and maxilofacial surgery and is frequently combined with bone graft materials to fill bony defects and enhance healing. Numerous studies have been performed investigating the efficacy of PRC to enhance bone healing in which a variety of graft materials have been combined with varying degrees of success. Here, we sought to determine the effect of combining PRC with different graft materials on human bone marrow stromal cell (hBMSC) proliferation, osteoblastic differentiation, and bone formation. METHODS Our central hypothesis is that PRC is not a true osteogenic agent but rather is osteopromotive, with cell fate determination being dependent on additional signals derived from the microenvironment. Experiments were performed with low passage (maximum 3) hBMSCs that were maintained in the presence of ascorbic acid-2-phosphate and beta-glycerol phosphate. Dexamethasone was excluded from these studies. PRC and graft materials were retained within well inserts and clotted by addition of bovine thrombin. Cell proliferation was determined by DNA content, osteoblastic commitment, and differentiation by alkaline phosphatase activity and matrix mineralization. RESULTS Combining PRC with the graft materials increased proliferation above that seen with the graft materials alone; however, only demineralized bone matrix (DBM) and allograft were capable of increasing proliferation above that seen with PRC alone. The increased proliferation observed in the presence of PRC coincided with decreased normalized alkaline phosphatase activity, suggesting decreased osteoblastic differentiation. However, at later time points, PRC increased mineralization compared with DBM, collagen, or beta tricalcium phosphate alone. When compared with PRC alone, addition of DBM or allograft decreased mineralization. Collagen gave rise to a small increase in mineralization, whereas beta tricalcium phosphate yielded the same level of mineralization as PRC alone. CONCLUSIONS The data obtained from these in vitro investigations demonstrate that the cellular responses induced by PRC and bone graft materials in hBMSC can be significantly (positively or negatively) modified by adding the agents in combination. These in vitro data highlight the need to consider the potential interaction between biologic agents when added in combination.
Collapse
|