Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Arthritis Res Ther ; 24(1): 221, 2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-36096945

RESUMO

Osteoarthritis (OA) is a degenerative disease associated with cartilage degradation, osteophyte formation, and fibrillation. Autologous Protein Solution (APS), a type of autologous anti-inflammatory orthobiologic, is used for pain management and treatment of OA. Various compositions of autologous PRP formulations are in clinical use for musculoskeletal pathologies, by nature of their minimal processing and source of bioactive molecules. Currently, there is no consensus on the optimal composition of the complex mixture. In this study, we focused on elucidating the immune cell subtypes and phenotypes in APS. We identified the immune cell types in APS from healthy donors and investigated phenotypic changes in the immune cells after APS processing. Based on flow cytometric analysis, we found that neutrophils and T cells are the most abundant immune cell types in APS, while monocytes experience the largest fold change in concentration compared to WBCs. Gene expression profiling revealed that APS processing results in differential gene expression changes dependent on immune cell type, with the most significantly differentially regulated genes occurring in the monocytes. Our results demonstrate that the mechanical processing of blood, whose main purpose is enrichment and separation, can alter its protein and cellular composition, as well as cellular phenotypes in the final product.


Assuntos
Osteoartrite , Anti-Inflamatórios/uso terapêutico , Expressão Gênica , Humanos , Leucócitos , Monócitos , Osteoartrite/patologia
2.
Int J Mol Sci ; 22(5)2021 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-33800401

RESUMO

Osteoarthritis (OA) is a progressive degenerative disease that manifests as pain and inflammation and often results in total joint replacement. There is significant interest in understanding how intra-articular injections made from autologous blood or bone marrow could alleviate symptoms and potentially intervene in the progression of the disease. There is in vitro an in vivo evidence that suggests that these therapies, including platelet-rich plasma (PRP), autologous anti-inflammatories (AAIs), and concentrated bone marrow aspirate (cBMA), can interrupt cartilage matrix degradation driven by pro-inflammatory cytokines. This review analyzes the evidence for and against inclusion of white blood cells, the potential role of platelets, and the less studied potential role of blood plasma when combining these components to create an autologous point-of-care therapy to treat OA. There has been significant focus on the differences between the various autologous therapies. However, evidence suggests that there may be more in common between groups and perhaps we should be thinking of these therapies on a spectrum of the same technology, each providing significant levels of anti-inflammatory cytokines that can be antagonists against the inflammatory cytokines driving OA symptoms and progression. While clinical data have demonstrated symptom alleviation, more studies will need to be conducted to determine whether these preclinical disease-modifying findings translate into clinical practice.


Assuntos
Anti-Inflamatórios/uso terapêutico , Osteoartrite do Joelho/terapia , Plasma Rico em Plaquetas , Sistemas Automatizados de Assistência Junto ao Leito , Humanos , Injeções Intra-Articulares , Articulação do Joelho/imunologia , Articulação do Joelho/patologia , Osteoartrite do Joelho/imunologia , Osteoartrite do Joelho/patologia
3.
J Orthop Res ; 38(2): 253-257, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31469192

RESUMO

Understanding the molecular drivers and feedback loops of osteoarthritis (OA) may provide future therapeutic strategies to modulate the disease progression. The current paradigm of OA is evolving from a purely mechanical disease caused by cartilage wear toward a complex biological response connecting biomechanics, inflammation, and the immune system. The view of OA as a chronic wound highlights the role inflammation plays and also the body's attempts to repair an ongoing injury. Inflammatory signals, including cytokines such as interleukin-1 and tissue necrosis factor α, surface-expressed pattern recognition receptors such as toll-like receptors 2 and 4, complement factors such as C5, as well as pathogen-associated molecular patterns and damage-associated molecular patterns drive the enzymatic cascade that degrades cartilage matrix in OA. Considering the joint as an entire organ, interactions between the cells that reside in the synovium including macrophages and other immune cells, appear to drive enzymatic activity in cartilage, which, in turn, feeds signals back to the synovium that continues stimulating degradation in a feed-forward loop. This review will explore the potential roles of immune cells such as macrophages and T cells in the synovium in both stimulating and modulating the inflammatory response in OA. © 2019 Orthopedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:253-257, 2020.


Assuntos
Osteoartrite/imunologia , Imunidade Adaptativa , Animais , Humanos , Imunidade Inata , Macrófagos/fisiologia , Cicatrização
4.
Biomed Res Int ; 2018: 6510842, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30112414

RESUMO

There has been significant debate over the role of white blood cells (WBCs) in autologous therapies, with several groups suggesting that WBCs are purely inflammatory. Misconceptions in the practice of biologic orthopedics result in the simplified principle that platelets deliver growth factors, WBCs cause inflammation, and the singular value of bone marrow is the stem cells. The aim of this review is to address these common misconceptions which will enable better development of future orthopedic medical devices. WBC behavior is adaptive in nature and, depending on their environment, WBCs can hinder or induce healing. Successful tissue repair occurs when platelets arrive at a wound site, degranulate, and release growth factors and cytokines which, in turn, recruit WBCs to the damaged tissue. Therefore, a key role of even pure platelet-rich plasma is to recruit WBCs to a wound. Bone marrow contains a complex mixture of vascular cells, white blood cells present at much greater concentrations than in blood, and a small number of progenitor cells and stem cells. The negative results observed for WBC-containing autologous therapies in vitro have not translated to human clinical studies. With an enhanced understanding of the complex WBC biology, the next generation of biologics will be more specific, likely resulting in improved effectiveness.


Assuntos
Autoenxertos , Transplante de Medula Óssea , Medula Óssea , Contagem de Leucócitos , Plaquetas , Citocinas , Humanos , Leucócitos
5.
Biores Open Access ; 6(1): 151-158, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29279807

RESUMO

Osteoarthritis (OA) is a progressive and degenerative disease, which may result in significant pain and decreased quality of life. Recent updates in our understanding of OA have demonstrated that it is a whole joint disease that has many similarities to an unhealed wound containing inflammatory cytokines. The nSTRIDE Autologous Protein Solution (APS) Kit is a medical device under development for the treatment of OA. The APS Kit processes a patient's own blood at the point of care to contain high concentrations of anti-inflammatory cytokines and anabolic growth factors. This study assessed the safety and treatment effects of a single intra-articular injection of APS. Eleven patients were enrolled in this study. Sufficient blood could not be drawn from one patient who was subsequently withdrawn, leaving 10 patients treated. Minor adverse events (AEs) were experienced by seven subjects (63.6%). There was one serious AE (diverticulitis) unrelated to the device or procedure. One subject experienced AEs that were judged "likely" to be procedure related (arthralgia/musculoskeletal discomfort) and all resolved within 6 days of injection. All other AEs were unrelated to the device or procedure. Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) pain scores improved significantly over time (ANOVA, p < 0.0001, 12.0 ± 1.2 preinjection, 3.3 ± 2.9 one year postinjection, and 72.5% WOMAC pain improvement). There was significant positive correlation between white blood cell concentration in APS and improvement in WOMAC pain scores.

6.
J Orthop Res ; 35(10): 2260-2268, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28139016

RESUMO

Current osteoarthritis (OA) research searches for treatments that modify the course of disease progression. Autologous Protein Solution (APS) is derived from whole blood and is a unique autologous therapy that contains high concentrations of white blood cells, platelets, and concentrated plasma, providing cytokines that can target the underlying mechanisms of disease progression. The APS Kit is currently under investigation for clinical use in the USA (NCT02262364). The aim of this study was to determine if APS has disease-modifying properties in the well-characterized rat meniscal tear-induced model of OA. Thirty male athymic rats underwent surgery to induce OA by a complete tear of the medial meniscus of the right knee. Seven days later, rats were administered 50 µl intra-articular (IA) APS from a human donor or phosphate buffered saline (PBS) control. Rats were euthanized 3 weeks following treatment and knee joints were processed for histological analysis. Collagen and cartilage degeneration were decreased by APS treatment, resulting in a significantly improved total joint score in APS-treated rats compared to those treated with the PBS control. No significant variations in gait analysis, weight gain, osteophyte score, or synovitis score were observed between APS- and PBS-treated animals. There were no adverse effects of APS reported in the study. Treatment with a single IA injection of APS reduced the cartilage degeneration that characterizes the progression of OA. Further studies are necessary to determine if APS can modify OA disease progression in humans. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2260-2268, 2017.


Assuntos
Anti-Inflamatórios/uso terapêutico , Transfusão de Componentes Sanguíneos , Proteínas Sanguíneas/uso terapêutico , Osteoartrite do Joelho/prevenção & controle , Lesões do Menisco Tibial/complicações , Animais , Anti-Inflamatórios/química , Humanos , Injeções Intra-Articulares , Masculino , Osteoartrite do Joelho/etiologia , Ratos Nus
7.
J Exp Orthop ; 3(1): 9, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26915009

RESUMO

BACKGROUND: There has been debate on which blood components should be included in autologous therapies. Autologous Protein Solution (APS) is a unique blood-derived therapy composed of concentrated white blood cells, platelets, and plasma to contain high concentrations of anti-inflammatory cytokines and anabolic growth factors to potentially address osteoarthritis. The primary aim of the exploratory secondary analysis was to identify characteristics of an Autologous Protein Solution (APS) that may correlate with improved Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) scores and OMERACT-OARSI responder status after treatment of subjects with an intra-articular injection of APS. METHODS: Eleven subjects were enrolled in a pilot study of a single intra-articular injection of APS in subjects with knee osteoarthritis. Two APS kits were processed per patient. The output of the first APS kit was injected intra-articularly. White blood cell (WBC) and cytokine concentrations were measured from the output of the second APS kit. WOMAC surveys were completed at baseline and at follow up visits. Linear regression analyses were performed on the blood components of APS with subject outcomes. Anderson-Darling analysis was used to determine whether the cytokine concentrations in whole blood and APS had a normal distribution. Either paired t-test analyses or Wilcoxon signed-rank analyses were performed for normal and non-parametrically distributed data, respectively. RESULTS: The WBC concentration in APS was significantly (p < 0.05) and strongly (R(2) > 0.7) correlated with IL-1ra in APS but not significantly correlated with IL-1ß. The ratio of IL-1ra to IL-1ß in APS was significantly correlated with improved WOMAC pain scores one week and six months post-injection. 85.7 % of subjects whose APS had a IL-1ra:IL-1ß ratio greater than 1000 or a WBC count greater than 30 k/µl were OMERACT-OARSI responders six months post-injection. CONCLUSIONS: The correlations between the IL-1ra:IL-1ß ratio and WBC concentration in a subject's APS and their WOMAC pain scores and classification as OMERACT-OARSI responders suggest the potential utility of these characteristics as diagnostic markers. Additional studies are ongoing to determine whether APS is safe and effective and to further evaluate the relationship between APS composition and clinical outcomes. TRIAL REGISTRATION: ( NCT01773226 ).

8.
Biores Open Access ; 4(1): 417-24, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26634187

RESUMO

Critical limb ischemia (CLI) is a terminal disease with high morbidity and healthcare costs due to limb loss. There are no effective medical therapies for patients with CLI to prevent amputation. Cell-based therapies are currently being investigated to address this unmet clinical need and have shown promising preliminary results. The purpose of this study was to characterize the output of a point-of-care cell separator (MarrowStim P.A.D. Kit), currently under investigation for the treatment of CLI, and compare its output with Ficoll-based separation. The outputs of the MarrowStim P.A.D. Kit and Ficoll separation were characterized using an automated hematology analyzer, colony-forming unit (CFU) assays, and tubulogenesis assays. Hematology analysis indicated that the MarrowStim P.A.D. Kit concentrated the total nucleated cells, mononuclear cells, and granulocytes compared with baseline bone marrow aspirate. Cells collected were positive for VEGFR-2, CD3, CD14, CD34, CD45, CD56, CD105, CD117, CD133, and Stro-1 antigen. CFU assays demonstrated that the MarrowStim P.A.D. Kit output a significantly greater number of mesenchymal stem cells and hematopoietic stem cells compared with cells output by Ficoll separation. There was no significant difference in the number of endothelial progenitor cells output by the two separation techniques. Isolated cells from both techniques formed interconnected nodes and microtubules in a three-dimensional cell culture assay. This information, along with data currently being collected in large-scale clinical trials, will help instruct how different cellular fractions may affect the outcomes for CLI patients.

9.
J Orthop Res ; 32(10): 1349-55, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24981198

RESUMO

The objective of this clinical study was to test if blood from osteoarthritis (OA) patients (n = 105) could be processed by a device system to form an autologous protein solution (APS) with preferentially increased concentrations of anti-inflammatory cytokines compared to inflammatory cytokines. To address this objective, APS was prepared from patients exhibiting radiographic evidence of knee OA. Patient metrics were collected including: demographic information, medical history, medication records, and Knee Injury and Osteoarthritis Outcome Score (KOOS) surveys. Cytokine and growth factor concentrations in whole blood and APS were measured using enzyme-linked immunosorbent assays. Statistical analyses were used to identify relationships between OA patient metrics and cytokines. The results of this study indicated that anti-inflammatory cytokines were preferentially increased compared to inflammatory cytokines in APS from 98% of OA patients. APS contained high concentrations of anti-inflammatory proteins including 39,000 ± 20,000 pg/ml IL-1ra, 21,000 ± 5,000 pg/ml sIL-1RII, 2,100 ± 570 pg/ml sTNF-RI, and 4,200 ± 1,500 pg/ml sTNF-RII. Analysis of the 82 patient metrics indicated that no single patient metric was strongly correlated (R(2) > 0.7) with the key cytokine concentrations in APS. Therefore, APS can be prepared from a broad range of OA patients.


Assuntos
Transfusão de Sangue Autóloga , Citocinas/sangue , Mediadores da Inflamação/sangue , Peptídeos e Proteínas de Sinalização Intercelular/sangue , Osteoartrite/sangue , Regulação para Cima/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Citocinas/biossíntese , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Masculino , Pessoa de Meia-Idade , Osteoartrite/patologia , Osteoartrite/terapia , Adulto Jovem
10.
J Orthop Res ; 31(12): 1929-35, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23966313

RESUMO

Osteoarthritis (OA) is characterized by deterioration of articular cartilage driven by an imbalance of pro- and anti-inflammatory cytokines. To address the cartilage deterioration observed in OA, an autologous protein solution (APS) has been developed which has been shown to inhibit the production of destructive proteases and inflammatory cytokines from chondrocytes and monocytes, respectively. The purpose of this study was to determine the chondroprotective effect of APS on IL-1α- or TNFα-challenged bovine articular cartilage explants. Cartilage explants were cultured in the presence or absence of recombinant inflammatory cytokines, IL-1α and TNFα. Explants under equivalent inflammatory conditions were pretreated with recombinant antagonists IL-1ra, sTNF-RI, or APS to measure their inhibition of matrix degradation. Explants were further evaluated with Safranin-O, Masson's Trichrome, and Hematoxylin and Eosin histological staining. APS was more effective than recombinant antagonists in preventing cartilage matrix degradation and inhibited any measurable IL-1α-induced collagen release over a 21-day culture period. APS treatment reduced the degree of Safranin-O staining loss when cartilage explants were cultured with IL-1α or TNFα. Micrographs of APS treated cartilage explants showed an increase in observed cellularity and apparent cell division. APS may have the potential to prevent cartilage loss associated with early OA.


Assuntos
Proteínas Sanguíneas/farmacologia , Cartilagem Articular/efeitos dos fármacos , Interleucina-1alfa/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Proteínas Sanguíneas/uso terapêutico , Cartilagem Articular/metabolismo , Bovinos , Glicosaminoglicanos/análise , Humanos , Osteoartrite/tratamento farmacológico , Proteínas Recombinantes/farmacologia , Soluções
11.
Inflamm Res ; 60(10): 929-36, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21687998

RESUMO

OBJECTIVE: The purpose of this research was to determine if an autologous protein solution (APS), prepared from platelet-rich plasma (PRP), could reduce the deleterious effects of inflammatory cytokines in vitro. METHODS: APS was prepared by processing human blood in a tuned density buoy separation device (Platelet Separation System, Biomet Biologics, LLC) to produce platelet-rich plasma (PRP) and processing the PRP in a concentration device containing polyacrylimide beads to produce a highly concentrated anti-inflammatory solution. A functional assay was designed using recombinant interleukin (IL)-1ß to upregulate IL-8 production by human monocytes. Either recombinant human interleukin-1 receptor antagonist (rhIL-1ra) or APS was added to some samples to determine if a reduced inflammatory response could be identified in vitro. The enzyme-linked immunosorbent assay (ELISA) method was employed to perform cytokine analyses, and Student's t test (α = 0.05) was used for all statistical analyses. RESULTS: Both the rhIL-1ra and the APS reduced the effect of IL-1ß on human macrophages in vitro. This was measured by the reduced production of IL-8 and tumor necrosis factor (TNF)-α. Further analysis of the supernatants confirmed the presence of high concentrations of IL-1ra and soluble TNF receptor I (sTNF-RI) with the APS treatment. CONCLUSION: The ability of the APS to reduce the effect of IL-1ß and limit the expression of other inflammatory cytokines in vitro validates its potential use as an autologous treatment for osteoarthritis.


Assuntos
Anti-Inflamatórios/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Plaquetas/metabolismo , Diferenciação Celular , Citocinas/biossíntese , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Técnicas In Vitro , Inflamação , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Interleucina-8/metabolismo , Monócitos/citologia , Plasma/metabolismo , Proteínas Recombinantes/metabolismo
12.
J Orthop Res ; 29(9): 1320-6, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21437966

RESUMO

Catabolic inflammatory cytokines are prevalent in osteoarthritis (OA). The purpose of this study was to evaluate an autologous protein solution (APS) as a potential chondroprotective agent for OA therapy. APS was prepared from platelet-rich plasma (PRP). The APS solution contained both anabolic (bFGF, TGF-ß1, TGF-ß2, EGF, IGF-1, PDGF-AB, PDGF-BB, and VEGF) and anti-inflammatory (IL-1ra, sTNF-RI, sTNF-RII, IL-4, IL-10, IL-13, and IFNγ) cytokines but low concentrations of catabolic cytokines (IL-1α, IL-1ß, TNFα, IL-6, IL-8, IL-17, and IL-18). Human articular chondrocytes were pre-incubated with the antagonists IL-1ra, sTNF-RI, or APS prior to the addition of recombinant human IL-1ß or TNFα. Following exposure to inflammatory cytokines, the levels of MMP-13 in the culture medium were evaluated by ELISA. MMP-13 production stimulated in chondrocytes by IL-1ß or TNFα was reduced by rhIL-1ra and sTNF-RI to near basal levels. APS was also capable of inhibiting the production of MMP-13 induced by both IL-1ß and TNFα. The combination of anabolic and anti-inflammatory cytokines in the APS created from PRP may render this formulation to be a potential candidate for the treatment of inflammation in patients at early stages of OA.


Assuntos
Proteínas Sanguíneas/farmacologia , Condrócitos/efeitos dos fármacos , Interleucina-1beta/farmacologia , Metaloproteinase 13 da Matriz/metabolismo , Plasma Rico em Plaquetas/química , Fator de Necrose Tumoral alfa/farmacologia , Cartilagem Articular/citologia , Células Cultivadas , Condrócitos/metabolismo , Antagonismo de Drogas , Combinação de Medicamentos , Sinergismo Farmacológico , Humanos , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Interleucina-1beta/antagonistas & inibidores , Articulação do Joelho/citologia , Proteínas Recombinantes
13.
Cell Tissue Bank ; 12(2): 81-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20039143

RESUMO

Demineralization of allograft bone increases the bioavailability of matrix-associated bone morphogenetic proteins (BMPs), rendering these grafts osteoinductive. While osteoinductivity is related to BMP content, little is known about how the demineralization protocol, in particular, extended demineralization times, affects graft BMP levels. We characterized the BMP-7 content of <710 µm bovine bone powder demineralized under various conditions. Using 1 g of bone per 50 ml of 0.125 N, 0.25 N, or 0.5 N HCl, demineralization was performed at room temperature for 5 min to 24 h. Minimum residual calcium levels were obtained within 90 min and were <1 wt % using the 0.25 N and 0.5 N baths and 17 wt % using the 0.125 N bath. Measured peak BMP-7 levels were also obtained within 90 min and were 161-165 ng g(-1) using the 0.25 N and 0.5 N baths and 55.2 ng g(-1) using the 0.125 N bath. This compares to 5.1 ng g(-1) for undemineralized bone. Further acid bath exposure to 24 h resulted in BMP-7 decline to about 50% of the peak value, which was significant. The BMP-7 half-life was estimated to be 26 h. It is likely that the decline was due to diffusion of BMP-7 from the bone matrix into the acid. These results suggest the importance of not over demineralizing bone grafts and should stimulate further research that can be incorporated into the processing methodology followed by tissue banks.


Assuntos
Técnica de Desmineralização Óssea/métodos , Matriz Óssea/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Ácido Clorídrico/metabolismo , Animais , Proteína Morfogenética Óssea 7/metabolismo , Cálcio/metabolismo , Bovinos , Fatores de Tempo , Transplante Homólogo
14.
J Bone Joint Surg Am ; 91(6): 1459-70, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19487525

RESUMO

BACKGROUND: Demineralized bone matrix is an osteoinductive and osteoconductive material that is often used in orthopaedic surgery to induce bone formation. Autologous platelet-rich plasma, which contains proliferative and chemoattractant growth factors, has been used as a demineralized bone matrix adjuvant with mixed results. One variable during clinical use appears to be whether the platelet-rich plasma is activated with thrombin or is implanted in a liquid form with intact platelets. The objective of the present study was to determine if platelet-rich plasma can increase the osteoinductivity of demineralized bone matrix when used without thrombin activation. METHODS: The bioactivity of the demineralized bone matrix was evaluated in vitro by determining alkaline phosphatase production by C2C12 myoblast cells. The effect of thrombin activation on platelet-rich plasma was studied in vitro by evaluating osteosarcoma and bone marrow stromal cells for cell number and transforming growth factor-beta1 activation. Demineralized bone matrices supplemented with platelet-rich plasma, with or without thrombin activation, were implanted intramuscularly in athymic rats and were examined at fourteen, twenty-eight, and fifty-six days. Histological samples were analyzed for osteogenesis and chondrogenesis. Osteogenesis was further characterized on the basis of alkaline phosphatase activity. RESULTS: In vitro, thrombin triggered an immediate release of growth factors from the platelet-rich plasma, and the platelet-rich plasma increased the number of both osteosarcoma and stromal cells in a dose-dependent manner. Thrombin activation of the platelet-rich plasma eliminated such stimulatory effects. In vivo, the platelet-rich plasma stimulated chondrogenesis on Day 14 and osteogenesis on Days 28 and 56, whereas thrombin-activated platelet-rich plasma acted as an inhibitor of such events. In addition, inflammatory cells were detected in demineralized bone matrix samples that were mixed with thrombin-activated platelet-rich plasma. These cells were not present in matrix mixed with platelet-rich plasma alone. CONCLUSIONS: Platelet-rich plasma significantly increased in vivo demineralized bone matrix osteoinductivity only when used without thrombin activation.


Assuntos
Densidade Óssea/fisiologia , Matriz Óssea/transplante , Condrogênese/efeitos dos fármacos , Osteogênese/fisiologia , Plasma Rico em Plaquetas/fisiologia , Trombina/metabolismo , Adulto , Animais , Matriz Óssea/fisiologia , Proliferação de Células , Células Cultivadas , Condrogênese/fisiologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Humanos , Implantes Experimentais , Masculino , Camundongos , Camundongos Nus , Fotomicrografia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Sensibilidade e Especificidade , Fator A de Crescimento do Endotélio Vascular/sangue , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
J Craniofac Surg ; 17(1): 84-90, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16432413

RESUMO

Demineralized bone matrix (DBM) is a widely used bone graft material that derives its osteoinductive potential from matrix-associated bone morphogenetic proteins (BMPs). Prior investigations have shown that the osteoinductive potential can vary widely, with influence from both donor and processing sources. Although it is plausible that donor variance in the BMP profile can be an important consideration, the few published studies available have given inconsistent and incomplete information about this. The goal was to (1) characterize the variance of BMP-2, BMP-4, and BMP-7 in fully demineralized DBM derived from 20 appropriately screened (Food and Drug Administration and the American Association of Tissue Banks criteria) donors (male and female, 17-65 years) and (2) using literature review, infer the potential for this to be an important source of variability in graft function. BMPs were extracted with 4 M guanidine hydrochloride, and levels of BMP-2, BMP-4, and BMP-7 were measured using enzyme-linked immunosorbent assay. Measured levels were as follows: BMP-2 = 21.4 +/- 12.0 ng/g DBM, BMP-4 = 5.45 +/- 2.04 ng/g DBM, and BMP-7 = 84.1 +/- 34.4 ng/g DBM, which were significantly different (P < 0.05). There was a positive linear correlation between BMP-2 and BMP-7 (P = 0.0227). DBM derived from female donors had significantly greater concentrations of BMP-2 and BMP-7 than did that derived from male donors (P = 0.0257 and 0.0245, respectively). There was no significant correlation between donor age and the levels of any of the measured BMPs. The magnitude of variance of BMP profile appears to reasonably well correspond to the variance in osteoinductive potential cited by others, suggesting the possibility of using this as a method of donor screening.


Assuntos
Técnica de Desmineralização Óssea , Matriz Óssea/química , Proteínas Morfogenéticas Ósseas/análise , Preservação de Tecido , Fator de Crescimento Transformador beta/análise , Adolescente , Adulto , Idoso , Matriz Óssea/transplante , Proteína Morfogenética Óssea 2 , Proteína Morfogenética Óssea 4 , Proteína Morfogenética Óssea 7 , Cálcio/análise , Feminino , Liofilização , Humanos , Masculino , Pessoa de Meia-Idade , Fatores Sexuais , Doadores de Tecidos
16.
J Foot Ankle Surg ; 44(5): 345-53, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16210154

RESUMO

Osteoinductive demineralized bone matrix results from bone demineralization and is attributed to matrix-associated bone morphogenetic proteins. The osteoinductive potential can vary with donor. Many bioassay methods are available to screen donors, each with its own interpretation, so performance of more than one may be of value. Furthermore, little is known about the relationship between bioassay results and clinical outcomes. A study designed to meaningfully explore these issues would require assay of a large number of donors as well as clinical utilization in a large patient population. A preliminary study was undertaken to gain initial perspective. Using demineralized bone matrix derived from one 33-year-old female donor, 2 methods of bioassay and a clinical case study were performed. The levels of bone morphogenetic proteins 2, 4, and 7 in lyophilized demineralized bone matrix powder were measured (19.65 +/- 0.30 ng/g, 2.49 +/- 0.19 ng/g, and 82.03 +/- 6.89 ng/g, respectively). Also, putty (Osteostim DBM Putty), prepared from powder, was intramuscularly implanted in athymic rats and de novo bone formation quantified (6.7% +/- 3.5% new bone formation with 49% +/- 17% of the implant area associated with new bone formation). The putty, in conjunction with internal fixation, was used in the revision of a medial malleolar nonunion of an obese, 76-year-old woman. Radiographic union with excellent graft incorporation was achieved by 12 weeks postoperatively, with maintenance of an acceptable clinical result during the 14-month follow-up period. These results are interpreted in the broader context of demineralized bone grafting, in general, and an outline for further study is presented.


Assuntos
Matriz Óssea/transplante , Transplante Ósseo/métodos , Adulto , Idoso , Animais , Traumatismos do Tornozelo/cirurgia , Matriz Óssea/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Terapia por Estimulação Elétrica , Feminino , Fixação Interna de Fraturas , Fraturas Ósseas/cirurgia , Fraturas não Consolidadas/cirurgia , Humanos , Masculino , Osteogênese , Ratos , Ratos Sprague-Dawley , Reoperação , Transplante Heterólogo , Transplante Homólogo
17.
J Craniofac Surg ; 16(5): 749-56; discussion 757-9, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16192851

RESUMO

Platelet rich plasma (PRP) has been shown to clinically accelerate healing of both soft and hard tissues. As a result, it has gained increasing popularity. However, the clinical effectiveness of each type of PRP preparation method can vary in technique and efficiency, and current methods to evaluate the platelet concentration efficiency of PRP systems have several limitations. Therefore, the purpose of this study was to validate an automated hematology analyzer, the Cell-Dyn 3700, to accurately count platelets in concentration ranges of approximately 2,000,000-4,800,000 platelets/microL. PRP platelets were counted by way of a manual counting method and on the Cell-Dyn 3700, and the statistical evaluation indicated no difference between the groups (P > 0.05). Dilution of the PRP was not required, and accurate platelet counts could be achieved up to platelet concentrations of 4,800,000 platelets/microL. PRPs must be resuspended on a rocker for at least 5 minutes before platelet counts, and the entire PRP sample must be resuspended to allow for equal distribution of platelets before counting. With use of the validated Cell-Dyn 3700, a platelet concentrate system was used to prepare 153 PRPs. The baseline whole blood platelet concentration (328,000 platelets/microL +/- 69,000 platelets/microL) and the average PRP samples (2,645,000 platelets/microL +/- 680,000 platelets/microL) were compared, resulting in an eightfold increase in concentration and an average platelet percent recovery of approximately 76%. Automated hematology analyzers can be used to accurately count platelets in PRP given the system has been validated appropriately and the PRP samples are prepared properly to provide adequate platelet suspension.


Assuntos
Contagem de Plaquetas/instrumentação , Transfusão de Plaquetas/métodos , Animais , Anticoagulantes/química , Bovinos , Centrifugação , Ácido Edético/química , Desenho de Equipamento , Hematologia/instrumentação , Humanos , Plasma , Contagem de Plaquetas/métodos , Contagem de Plaquetas/normas , Transfusão de Plaquetas/instrumentação , Transfusão de Plaquetas/normas
18.
J Craniofac Surg ; 16(4): 579-85, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16077301

RESUMO

Allograft human bone is an integral part of the surgeons' armamentarium and will continue to be for the near future. The intraoperative handling and/or mechanical properties are critical to its function. These properties are significantly influenced by the composition and the structure of the bone, which varies from donor to donor. Published studies of human bone composition use bone derived from a population that may differ from the population of qualified donors from which allograft bone is derived and may not well represent the pool of clinical allograft bone. This study investigated the cortical bone composition from 20 donors (males and females, 17 to 65 years of age) that had passed the US Food and Drug Administration and American Association of Tissue Banks screening procedures for donor qualification. As such, this represents a subset of the general population. The analysis yielded the following composition: mineral (ash) = 67.0% +/- 1.3% (w/w); matrix (predominantly type I collagen and other proteins) = 31.9% +/- 1.1% (w/w); and lipid (hexane extractables) = 1.1% +/- 1.5% (w/w). In general, these results were well within the ranges specified in the literature, with the significance being the demonstration of low variability within the study population. No age or gender compositional dependency was evident in this series, possibly as a result of the relatively homogenous population, which may have limited the ability to observe trends. Visually, the bone powders ranged from nearly white to red-brown. The more intense colors appeared to be associated with greater lipid content, perhaps indicating the presence of residual oxidized lipids.


Assuntos
Composição Corporal , Transplante Ósseo , Osso e Ossos/química , Doadores Vivos , Adolescente , Adulto , Idoso , Matriz Óssea/química , Feminino , Humanos , Lipídeos/análise , Masculino , Pessoa de Meia-Idade , Minerais/análise , Valores de Referência , Estados Unidos , Água/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA