Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Stem Cell Rev Rep ; 17(4): 1362-1373, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33469783

RESUMO

Gathering a better grasp on the adipose stromal vascular fraction (SVF) is demanding among clinicians for osteoarthritis (OA) care because of its promising but multifaceted clinical outcomes. The aim of this preclinical in vitro study was to test whether the mechanical approach with Hy-Tissue SVF system, a class IIa CE marked device of adipose tissue micro-fragmentation, influences the biological features and functions of SVF. We compared mechanical generated-SVF (mSVF) with the enzymatic generated-SVF (eSVF) by testing cell survival, phenotype, differentiation, and paracrine properties using ELISA assays. Both adipose SVF showed 80% viable cells and enrichment for CD-44 marker. The mSVF product preserved the functions of cell populations within the adipose tissue; however, it displayed lowered nucleated cell recovery and CFU-F than eSVF. As for multipotency, mSVF and eSVF showed similar differentiation commitment for osteochondral lineages. Both adipose SVF exhibited an increased release of VEGF, HGF, IGF-1 and PDGF-bb, involved in pathways mediating osteochondral repair and cell migration. Both mSVF and eSVF also displayed high release for the anti-inflammatory cytokine IL-10. After in vitro culture, supernatants from both mSVF and eSVF groups showed a low release of cytokines except for IL-10, thereby giving evidence of functional changes after culture expansion. In this study, mSVF showed active cell populations in the adipose tissue comparable to eSVF with excellent survival, differentiation and paracrine properties under a new mechanical adipose tissue micro-fragmentation system; thereby suggesting its potential use as a minimally invasive technique for OA treatment.


Assuntos
Tecido Adiposo , Interleucina-10 , Osteoartrite , Fração Vascular Estromal , Animais , Diferenciação Celular , Osteoartrite/terapia , Coelhos
2.
Bone Joint Res ; 8(2): 101-106, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30915216

RESUMO

OBJECTIVES: Meniscal injuries are often associated with an active lifestyle. The damage of meniscal tissue puts young patients at higher risk of undergoing meniscal surgery and, therefore, at higher risk of osteoarthritis. In this study, we undertook proof-of-concept research to develop a cellularized human meniscus by using 3D bioprinting technology. METHODS: A 3D model of bioengineered medial meniscus tissue was created, based on MRI scans of a human volunteer. The Digital Imaging and Communications in Medicine (DICOM) data from these MRI scans were processed using dedicated software, in order to obtain an STL model of the structure. The chosen 3D Discovery printing tool was a microvalve-based inkjet printhead. Primary mesenchymal stem cells (MSCs) were isolated from bone marrow and embedded in a collagen-based bio-ink before printing. LIVE/DEAD assay was performed on realized cell-laden constructs carrying MSCs in order to evaluate cell distribution and viability. RESULTS: This study involved the realization of a human cell-laden collagen meniscus using 3D bioprinting. The meniscus prototype showed the biological potential of this technology to provide an anatomically shaped, patient-specific construct with viable cells on a biocompatible material. CONCLUSION: This paper reports the preliminary findings of the production of a custom-made, cell-laden, collagen-based human meniscus. The prototype described could act as the starting point for future developments of this collagen-based, tissue-engineered structure, which could aid the optimization of implants designed to replace damaged menisci.Cite this article: G. Filardo, M. Petretta, C. Cavallo, L. Roseti, S. Durante, U. Albisinni, B. Grigolo. Patient-specific meniscus prototype based on 3D bioprinting of human cell-laden scaffold. Bone Joint Res 2019;8:101-106. DOI: 10.1302/2046-3758.82.BJR-2018-0134.R1.

3.
Transfus Apher Sci ; 57(4): 549-555, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29929885

RESUMO

INTRODUCTION: Various blood-derived products have been proposed for the topical treatment of ocular surface diseases. The aim of the study was to compare the different content of Growth Factors (GFs) and Interleukins (ILs) in peripheral blood (PB-S) and Cord Blood (CB-S) sera. MATERIALS AND METHODS: Sera were obtained from 105 healthy adult donors (PB-S) and 107 umbilical/placental veins at the time of delivery (CB-S). The levels of epithelial-GF (EGF), fibroblast-GF (FGF), platelet-derived-GF (PDGF), insulin-GF (IGF), transforming-GF alpha (TGF-α,) and beta 1-2-3 (TGF-ß1-ß2-ß3), vascular endothelial-GF (VEGF), nerve-GF (NGF), Interleukin (IL)-1ß,IL-4,IL-6,IL-10, and IL-13 were assessed by Bio-Plex Protein Array System (Bio-Rad Laboratories, CA, USA). The Mann-Whitney test for unpaired data was applied to compare GFs and ILs levels in the two sources. The associations among each GF/IL level and the obstetric data for CB-S and hematological characteristics for PB-S were also investigated. RESULTS: The levels of EGF, TGF-α, TGF-ß2, FGF, PDGF, VEGF, NGF, IL-1B, IL-4, IL-6, IL-10, and IL-13 were significantly higher in CB-S compared to PB-S. Conversely, the levels of IGF-1, IGF-2, and TGF-ß1 were significantly higher in PB-S. The female sex and the weight of the child showed a significant association in predicting EGF and PDGF levels. CONCLUSION: A significantly different content in those GFs and ILs was demonstrated in the two blood sources. Since each GF/IL selectively regulates different cellular processes involved in corneal healing, the use of PB-S or CB-S should be chosen on the basis of the cellular mechanism to be promoted in each clinical case.


Assuntos
Córnea/efeitos dos fármacos , Infecções Oculares/tratamento farmacológico , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucinas/metabolismo , Adulto , Feminino , Sangue Fetal , Humanos , Masculino , Pessoa de Meia-Idade , Soluções Oftálmicas/uso terapêutico , Estudos Prospectivos , Soro
4.
J Tissue Eng Regen Med ; 12(8): 1835-1842, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29770602

RESUMO

Synovial inflammation plays an important role in osteoarthritis (OA) pathogenesis. Different biological compounds have been tested mainly on chondrocytes, to treat early stages of OA. However, because OA has been recently defined as "an organ" pathology, investigation on synoviocytes is also needed. Therefore, the aim of the present study was to validate a human fibroblast-like synoviocytes cell line (K4IM) to test the effects of platelet-rich plasma (PRP) and hyaluronan (HA) on anabolic and catabolic gene expression and on HA secretion from cell cultures. In order to determine the effect of PRP and HA, K4IM cells were maintained in culture with or without TNF-α stimulation. In the presence of PRP, unstimulated K4IM cells presented the same expression of IL1B, IL6, CXCL8, VEGF, TIMP1, and hyaluronic synthase isoform HAS3 as primary human synoviocytes, while HA addition did not change their expression pattern, which was similar to control cells. Stimulated cells expressed significantly higher values of IL1B, CXCL8, and VEGF compared with unstimulated ones. PRP did not show any modification, except for VEGF, while HA addition modulated IL1B expression. PRP did not modulate HA release of both stimulated and unstimulated cells. Our study showed the possibility to use K4IM synoviocytes as an in vitro model to test biological compounds useful for the treatment of early OA. Primary cells reflect the phenotype of cells in vivo, but limited recovery from biopsies and restricted lifespan makes experimental manipulation challenging. Therefore, despite cell lines present some limitations, they could be used as an alternative for preliminary experiments.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Hialurônico/farmacologia , Plasma Rico em Plaquetas , Sinoviócitos/metabolismo , Técnicas de Cultura de Células , Linhagem Celular Transformada , Citocinas/biossíntese , Humanos , Sinoviócitos/citologia , Inibidor Tecidual de Metaloproteinase-1/biossíntese
5.
Biotech Histochem ; 92(3): 201-211, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28318324

RESUMO

Although articular cartilage is the target of osteoarthritis (OA), its deterioration is not always clearly associated with patient symptoms. Because a functional interaction between cartilage and bone is crucial, the pathophysiology of OA and its treatment strategy must focus also on subchondral bone. We investigated whether adipose-derived stromal cells (ASCs) injected into a joint at two different concentrations could prevent subchondral bone damage after the onset of mild OA in a rabbit model. We measured both volumetric and densitometric aspects of bone remodeling. Although OA can stimulate bone remodeling either catabolically or anabolically over time, the accelerated turnover does not allow complete mineralization of new bone and therefore gradually reduces its density. We measured changes in morphometric and densitometric bone parameters using micro-CT analysis and correlated them with the corresponding parameters in cartilage and meniscus. We found that ASCs promoted cartilage repair and helped counteract the accelerated bone turnover that occurs with OA.


Assuntos
Adipócitos/transplante , Osso e Ossos/citologia , Osteoartrite/terapia , Células Estromais , Animais , Remodelação Óssea , Densitometria , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Masculino , Osteoartrite/patologia , Coelhos
6.
Osteoarthritis Cartilage ; 25(4): 589-599, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27836674

RESUMO

OBJECTIVE: Osteoarthritis (OA), the most common chronic degenerative joint disease, is characterized by joint structure changes and inflammation, both mediated by the IκB kinase (IKK) signalosome complex. The ability of N-acetyl phenylalanine derivative (NAPA) to increase cartilage matrix components and to reduce inflammatory cytokines, inhibiting IKKα kinase activity, has been observed in vitro. The present study aims to further clarify the effect of NAPA in counteracting OA progression, in an in vivo mouse model after destabilization of the medial meniscus (DMM). DESIGN: 26 mice were divided into three groups: (1) DMM surgery without treatment; (2) DMM surgery treated after 2 weeks with one intra-articular injection of NAPA (2.5 mM) and (3) no DMM surgery. At the end of experimental times, both knee joints of the animals were analyzed through histology, histomorphometry, immunohistochemistry and microhardness of subchondral bone (SB) tests. RESULTS: The injection of NAPA significantly improved cartilage thickness (CT) and reduced Chambers and Mankin modified scores and fibrillation index (FI), with weaker MMP13, ADAMTS5, MMP10 and IKKα staining. The microhardness measurements did not shown statistically significant differences between the different groups. CONCLUSIONS: NAPA markedly improved the physical structure of articular cartilage while reducing catabolic enzymes, extracellular matrix (ECM) remodeling and IKKα expression, showing to be able to exert a chondroprotective activity in vivo.


Assuntos
Cartilagem Articular/efeitos dos fármacos , Glucosamina/farmacologia , Articulação do Joelho/efeitos dos fármacos , Osteoartrite do Joelho/imunologia , Fenilalanina/análogos & derivados , Proteína ADAMTS5/efeitos dos fármacos , Proteína ADAMTS5/metabolismo , Animais , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Modelos Animais de Doenças , Quinase I-kappa B/efeitos dos fármacos , Quinase I-kappa B/metabolismo , Inflamação , Injeções Intra-Articulares , Articulação do Joelho/imunologia , Articulação do Joelho/metabolismo , Articulação do Joelho/patologia , Masculino , Metaloproteinase 10 da Matriz/efeitos dos fármacos , Metaloproteinase 10 da Matriz/metabolismo , Metaloproteinase 13 da Matriz/efeitos dos fármacos , Metaloproteinase 13 da Matriz/metabolismo , Meniscos Tibiais/cirurgia , Camundongos , Tamanho do Órgão , Osteoartrite do Joelho/metabolismo , Osteoartrite do Joelho/patologia , Fenilalanina/farmacologia
7.
J Biol Regul Homeost Agents ; 30(2): 409-20, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27358127

RESUMO

Osteochondral lesions are considered a challenge for orthopedic surgeons. Currently, the treatments available are often unsatisfactory and unable to stimulate tissue regeneration. Tissue engineering offers a new therapeutic strategy, taking into account the role exerted by cells, biomaterial and growth factors in restoring tissue damage. In this light, Mesenchymal Stem Cells (MSCs) have been indicated as a fascinating tool for regenerative medicine thanks to their ability to differentiate into bone, cartilage and adipose tissue. However, in vitro-cultivation of MSCs could be associated with some risks such as de-differentiation/reprogramming, infection and contaminations of the cells. To overcome these shortcomings, a new approach is represented by the use of Bone Marrow Concentrate (BMC), that could allow the delivery of cells surrounded by their microenvironment in injured tissue. For this purpose, cells require a tridimensional scaffold that can support their adhesion, proliferation and differentiation. This study is focused on the potentiality of BMC seeded onto a hyaluronan-based scaffold (Hyaff-11) to differentiate into osteogenic lineage. This process depends on the specific interaction between cells derived from bone marrow (surrounded by their niche) and scaffold, that create an environment able to support the regeneration of damaged tissue. The data obtained from the present study demonstrate that BMC grown onto Hyaff-11 are able to differentiate toward osteogenic sense, producing specific osteogenic genes and matrix proteins.


Assuntos
Células da Medula Óssea/citologia , Ácido Hialurônico/farmacologia , Osteogênese/efeitos dos fármacos , Alicerces Teciduais , Adulto , Diferenciação Celular/efeitos dos fármacos , Colágeno Tipo I/análise , Feminino , Humanos , Imuno-Histoquímica , Masculino , RNA Mensageiro/análise
8.
J Tissue Eng Regen Med ; 10(5): 374-91, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-23495253

RESUMO

Osteochondral lesions require treatment to restore the biology and functionality of the joint. A novel nanostructured biomimetic gradient scaffold was developed to mimic the biochemical and biophysical properties of the different layers of native osteochondral structure. The present results show that the scaffold presents important physicochemical characteristics and can support the growth and differentiation of mesenchymal stromal cells (h-MSCs), which adhere and penetrate into the cartilaginous and bony layers. H-MSCs grown in chondrogenic or osteogenic medium decreased their proliferation during days 14-52 on both scaffold layers and in medium without inducing factors used as controls. Both chondrogenic and osteogenic differentiation of h-MSCs occurred from day 28 and were increased on day 52, but not in the control medium. Safranin O staining and collagen type II and proteoglycans immunostaining confirmed that chondrogenic differentiation was specifically induced only in the cartilaginous layer. Conversely, von Kossa staining, osteocalcin and osteopontin immunostaining confirmed that osteogenic differentiation occurred on both layers. This study shows the specific potential of each layer of the biomimetic scaffold to induce chondrogenic or osteogenic differentiation of h-MSCs. These processes depended mainly on the media used but not the biomaterial itself, suggesting that the local milieu is fundamental for guiding cell differentiation. Copyright © 2013 John Wiley & Sons, Ltd.


Assuntos
Materiais Biomiméticos/química , Regeneração Óssea , Diferenciação Celular , Condrogênese , Células-Tronco Mesenquimais/metabolismo , Nanocompostos/química , Antígenos de Diferenciação/biossíntese , Humanos , Células-Tronco Mesenquimais/citologia
9.
Knee Surg Sports Traumatol Arthrosc ; 23(7): 2081-9, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24700335

RESUMO

PURPOSE: Bipolar fresh osteochondral allografts (BFOA) recently became a fascinating option for articular cartilage replacement, in particular in those young patients non-suitable for traditional replacement because of age. While the use of osteochondral allografts for the treatment of focal osteochondral lesions in the knee is well established, their use in the treatment of end-stage arthritis is far more controversial. The purpose of this paper is to describe our experience in a series of seven patients who underwent a resurfacing of both tibio-femoral and patello-femoral joints by BFOA. METHODS: From 2005 to 2007, seven patients (mean age 35.2 ± 6.3 years) underwent BFOA for end-stage arthritis of the knee. Patients were evaluated clinically, radiographically and by CT scan preoperatively and at established intervals up to the final follow-up. RESULTS: No intra-operative complications occurred. Nevertheless, joint laxity and aseptic effusion, along with a progressive chondrolysis, lead to early BFOA failure in six patients, which were revised by total knee arthroplasty at 19.5 ± 3.9 months follow-up. Only one patient, who received the allograft to convert a knee arthrodesis, gained a satisfactory result at the last follow-up control. CONCLUSIONS: BFOA in the knee joint still remains an inapplicable option in the treatment of post-traumatic end-stage arthritis of the young patient, due to the high rate of failure. Further studies are necessary in order to investigate the causes of failure and improve the applicability of this method. Still, after extensive counselling with the patient, BFOA may represent a salvage procedure aimed to revise scarcely tolerated knee arthrodesis. LEVEL OF EVIDENCE: Retrospective case series, Level IV.


Assuntos
Cartilagem Articular/cirurgia , Cartilagem/transplante , Articulação do Joelho/cirurgia , Osteoartrite do Joelho/cirurgia , Adulto , Aloenxertos , Artrodese , Feminino , Humanos , Masculino , Osteoartrite do Joelho/diagnóstico por imagem , Osteoartrite do Joelho/etiologia , Osteoartrite do Joelho/patologia , Articulação Patelofemoral/cirurgia , Radiografia , Estudos Retrospectivos , Falha de Tratamento
10.
Eur Rev Med Pharmacol Sci ; 18(23): 3702-11, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25535144

RESUMO

OBJECTIVE: A number of studies have shown the role of expanded Bone Marrow-derived Mesenchymal Stem Cells in the repair and regeneration of musculo-skeletal tissues. The current European regulations define in vitro expanded cells for clinical purposes as substantially manipulated and include them in the class of Advanced-Therapy Medicinal Products to be manufactured in compliance with current Good Manufacturing Practice. Among the characteristics that such cells should display, genomic stability has recently become a major safety concern. The aim of this study is to perform a chromosomal and genetic characterization of Bone Marrow-derived Mesenchymal Stem Cells expanded in compliance with Good Manufacturing Practice for a potential clinical use in orthopaedics. MATERIALS AND METHODS: Mesenchymal Stem Cells, isolated from bone marrow, were expanded for six weeks in compliance with current Good Manufacturing Practice. DNA profiling analyses were applied to test cross-contamination absence. Genomic stability was evaluated by means of karyotyping, sequencing of TP53, p21/CDKN1A and MDM2 genes and the expression analysis of c-MYC and H-RAS oncogenes, p21/CDKN1A, TP53, p16/CDKN2A, RB1 and p27/CDKN1B tumor suppressor genes and hTERT gene. RESULTS: The DNA profiling analysis showed a unique genetic profile for each Mesenchymal Stem Cell culture, indicating the absence of cross-contamination. Karyotyping evidentiated some chromosomal abnormalities within the 10% limit set by the Cell Products Working Party review, except for one patient. In all cases, the molecular biology analyses did not revealed DNA point mutations, acquisition or changes in gene expression. hTERT levels were undetectable. CONCLUSIONS: Cultured Mesenchymal Stem Cells do not seem to be prone to malignant transformation. In fact, although some chromosomal aberrations were found, molecular biology analyses demonstrated that the expansion phase did not induce the acquisition of de novo genetic changes.


Assuntos
Células da Medula Óssea/fisiologia , Células-Tronco Mesenquimais/fisiologia , Doenças Musculoesqueléticas/genética , Doenças Musculoesqueléticas/terapia , Adulto , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Células Cultivadas , Feminino , Humanos , Cariotipagem/métodos , Masculino , Doenças Musculoesqueléticas/patologia , Adulto Jovem
11.
J Biol Regul Homeost Agents ; 27(1): 165-75, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23489696

RESUMO

Bone marrow is one of the best characterized stem cell microenvironments that contains Mesenchymal Stem Cells (MSCs), a rare population of non-hematopoietic stromal cells. MSCs have been indicated as a new option for regenerative medicine because of their ability to differentiate into various lineages such as bone, cartilage and adipose tissue. However, isolation procedures are crucial for the functional activity of the transplanted cells. The use of concentrated bone marrow cells (BMCs) enables a cell population surrounded by its microenvironment (niche) to be implanted while avoiding all the complications related to the in vitro culture. The cells of the niche are able to regulate stem cell behavior through direct physical contact and secreting paracrine factors. The aim of this study was to characterize BMCs in vitro to evaluate their ability to differentiate toward mature cells and try to understand whether there are differences in the chondrogenic and osteogenic potential of cells from patients of different ages. Mononuclear Cells (MNCs) isolated by Ficoll were used as control. Both cell populations were grown in monolayers and differentiated with specific factors and analyzed by histological and molecular biology assays to evaluate the expression of some specific extracellular matrix molecules. The present investigations revealed the ability of BMCs to act as isolated cells. They are able to form colonies and differentiate toward chondrogenic and osteogenic lineages, the latter pathway appearing to be influenced by donor age. The results obtained by this study support the use of BMCs in clinical practice for the repair of osteochondral damage, which might be particularly useful for the one-step procedure allowing cells to be directly implanted in operating room.


Assuntos
Doenças Ósseas/terapia , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Adulto , Agrecanas/genética , Agrecanas/metabolismo , Azul Alciano/metabolismo , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Antraquinonas/metabolismo , Doenças Ósseas/patologia , Condrogênese/genética , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Ensaio de Unidades Formadoras de Colônias , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Leucócitos Mononucleares/metabolismo , Masculino , Osteogênese/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Coloração e Rotulagem
12.
J Biol Regul Homeost Agents ; 26(4): 701-11, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23241120

RESUMO

Articular cartilage lesions represent a challenging problem for orthopaedic surgeons. The purpose of this study was to evaluate the effect of a new pulsed Nd:YAG High Intensity Laser Therapy on the regeneration of cartilage tissue in patients with traumatic lesions. Clinical, histological and immunohistochemical evaluations were performed. Ten patients affected by chondral lesions scheduled for ACI procedure, were enrolled into the study. During the chondrocyte expansion for ACI procedure, cartilage from five patients was treated by Nd:YAG High Intensity Laser Therapy (HILT group). No laser treatment was performed in the remaining patients, who were used as controls. Cartilage repair was assessed by clinicians using two different scores: Cartilage Repair Assessment (CRA) and Overall Repair Assessment (ORA). Cartilage biopsy specimens were harvested to perform histological and immunohistochemical analyses at T0 (before laser treatment) and T1 (at the end of the treatment). A significant decrease in cartilage depth was noticed in the HILT group at T1. Histological and immunohistochemical evaluations showed some regenerative processes in cartilaginous tissue in terms of high amount of proteoglycans, integration with adjacent articular cartilage and good cellular arrangement in the HILT group. By contrast, a not well organized cartilaginous tissue with various fibrous features in the control group at T0 and T1 was observed. In conclusion, the use of this new pulsed Nd:YAG HILT resulted promising in the treatment of moderate cartilage lesions markedly in the young patients.


Assuntos
Cartilagem Articular/lesões , Terapia a Laser , Adolescente , Adulto , Cartilagem Articular/patologia , Cartilagem Articular/cirurgia , Colágeno Tipo II/análise , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Projetos Piloto
13.
J Biol Regul Homeost Agents ; 25(2 Suppl): S3-13, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22051166

RESUMO

The requirements for a successful regeneration of an osteo-chondral defect could effectively be met by using a bi-layered composite scaffold, able to support proliferation and differentiation of mesenchymal stem cells, while providing a biochemical environment promoting the formations of the two distinct tissues. The novel strategy here presented consists of developing a bio-mimetic scaffolds obtained by the combination of two integrated organic compounds (type I collagen and chitosan) with or without bioactive Mg-doped hydroxyapatite (Mg-HA) nanocrystals, depending on the specific layer, reproducing cartilaginous or subchondral bone tissue. An innovative patented methodology for scaffolds production, called - pH-dependent 3-phasic assembling -, allowed to development of a highly homogenous and chemically stable scaffold, presenting a very good integration among all three components, as confirmed by extensive SEM and thermogravimetric analyses. A preliminary in vitro evaluation was also carried out by seeding bi-layered scaffold with human bone marrow stromal cells (h-MSCs), by giving particular emphasis to cell viability and distribution at day 0, 7 and 14. Cells were viable and uniformly colonized the whole scaffold until day 14, indicating that the scaffold contributed to the maintenance of cell behaviour.


Assuntos
Materiais Biomiméticos/química , Células da Medula Óssea/citologia , Regeneração Óssea , Cartilagem , Teste de Materiais , Alicerces Teciduais/química , Células da Medula Óssea/metabolismo , Substitutos Ósseos/química , Células Cultivadas , Quitosana/química , Colágeno Tipo I/química , Durapatita/química , Humanos , Células Estromais/citologia , Células Estromais/metabolismo
14.
J Biol Regul Homeost Agents ; 25(2 Suppl): S53-62, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22051171

RESUMO

Association of biomaterials with autologous cells can provide a new generation of implantable devices for cartilage repair. An ideal scaffold should possess a preformed three-dimensional shape, fix the cells to the damaged area and prevent their migration into the articular cavity. Furthermore, the constructs should have sufficient mechanical strength to facilitate handling in a clinical setting and stimulate the uniform spreading of cells and a phenotype re-differentiation process. The aim of this study was to verify the ability of an equine collagen membrane to support the growth of human chondrocytes and to allow the re-expression of their original phenotype. This ability was assessed by the evaluation of collagen type I, II and aggrecan mRNA expression by Real-Time PCR. Immunohistochemical analyses were performed to evaluate collagen type I, II and proteoglycans synthesis. Electron microscopy was utilized to highlight the structure of the biomaterial and its interactions with the cells. Our data indicate that human chondrocytes seeded onto a collagen membrane express and produce collagen type II and aggrecan and downregulate the production of collagen type I during the experimental times analyzed. These results provide an in vitro demonstration for the therapeutic potential of autologous chondrocyte transplantation by an equine collagen membrane as a delivery vehicle in a tissue-engineered approach towards the repair of articular cartilage defects.


Assuntos
Cartilagem Articular , Condrócitos/metabolismo , Colágeno Tipo II/química , Colágeno Tipo I/química , Teste de Materiais , Membranas Artificiais , Alicerces Teciduais/química , Adulto , Agrecanas/biossíntese , Animais , Células Cultivadas , Condrócitos/citologia , Colágeno Tipo I/biossíntese , Colágeno Tipo II/biossíntese , Feminino , Humanos , Masculino , RNA Mensageiro/biossíntese , Ovinos , Engenharia Tecidual
15.
Injury ; 41(7): 693-701, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20035935

RESUMO

INTRODUCTION: Osteochondral articular defects are a key concern in orthopaedic surgery. Current surgical techniques to repair osteochondral defects lead to poor subchondral bone regeneration and fibrocartilage formation, which is often associated with joint pain and stiffness. The objective of this pilot clinical study is to evaluate the performance and the intrinsic stability of a newly developed biomimetic osteochondral scaffold and to test the safety and the feasibility of the surgical procedure. METHODS: A gradient composite osteochondral scaffold based on type I collagen-hydroxyapatite was obtained by nucleating collagen fibrils with hydroxyapatite nanoparticles. Thirteen patients (15 defect sites) were treated with scaffold implantation from January 2007 to July 2007: four at the medial femoral condyle, two at the lateral femoral condyles, five at the patellas and four at the trochleas. The mean size of the defects was 2.8 cm(2) (range: 1.5-5.9 cm(2)). All patients were followed up prospectively. High-resolution magnetic resonance imaging (MRI) was used to determine "the early postoperative adherence rate" at 4-5 weeks and 25-26 weeks after scaffold implantation. Moreover, the magnetic resonance observation of cartilage repair tissue (MOCART) score was performed on every MRI. Two second-looks were performed at 6 months; cartilage repair was assessed using the International Cartilage Repair Society (ICRS) visual scoring system and histological and immunohistochemical analysis of the two biopsies was carried out. RESULTS: A completely attached graft and repair tissue were found in 13 of 15 lesions (86.7%). A partial detachment was observed in two patients (13.3%). No detached grafts were found. Complete filling of the cartilage defect and congruency of the articular surface were seen in 10 lesions (66.7%) with MRI evaluation at 6 months. The complete integration of the grafted cartilage was detected in eight lesions (53.3%). Subchondral bone changes (oedema or sclerosis) were found in eight defects (53.3%). Statistical analysis showed a significant improvement in the International Knee Documentation Committee (IKDC) subjective and objective scores from preoperative to 6 months' follow-up (p<0.0005). Visual scoring of the repaired tissue at second-look revealed a normal repair score in one case and a near-normal repair score in the other case. Histological analysis showed the formation of subchondral bone without the presence of biomaterial. The cartilage repair tissue appeared to be engaged in an ongoing maturation process. CONCLUSIONS: The technique is safe and MRI evaluation at short-term follow-up has demonstrated good stability of the scaffold without any other fixation device. The preliminary clinical results at short-term follow-up are encouraging. A clinical and MRI study with longer follow-up and randomised studies will be done to confirm the high potential of this novel osteochondral scaffold.


Assuntos
Regeneração Óssea/fisiologia , Cartilagem Articular , Traumatismos do Joelho/terapia , Nanocompostos/uso terapêutico , Osteocondrite/terapia , Cicatrização , Adulto , Materiais Biocompatíveis/uso terapêutico , Cartilagem Articular/lesões , Cartilagem Articular/patologia , Estudos de Viabilidade , Feminino , Humanos , Imuno-Histoquímica , Traumatismos do Joelho/patologia , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Osteocondrite/patologia , Projetos Piloto , Estudos Prospectivos , Engenharia Tecidual/métodos , Alicerces Teciduais , Cicatrização/fisiologia
16.
J Biomed Mater Res A ; 84(1): 117-27, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17600335

RESUMO

The anterior cruciate ligament (ACL) is the most commonly injured tissue of the human knee. Its poor ability to regenerate after injury represents a challenge to ligament tissue engineering. An understanding of the molecular composition of the structures used for its repair is essential for clinical assessments and for the implementation of tissue engineering strategies. The objective of this study was to evaluate, both at gene and protein levels, the expression of characteristic molecules in human ACL, patellar, semitendinosus and gracilis tendons and in the ligament reconstructed with patellar or semitendinosus and gracilis tendons. We demonstrated that primary ACL and tendon tissues all express collagen I, II, Sox-9, tenascin-C and aggrecan. Collagen X expression was detected at very low levels or undetectable. Cathepsin B, MMP-1 and MMP-13 were expressed at higher levels in the ACL reconstructed by the two tendons, showing that a remodeling process occurs during "ligamentization". Both our molecular and immunohistochemical evaluations did not reveal significative differences between the tendons and ligaments analyzed. However, ACL reconstructed with semitendinosus and gracilis tendon seems to present a higher expression of collagen type II when compared to that reconstructed with patellar tendon. This study could give a reasonable identification of genetic and protein markers specific to tendon/ligament tissues and be helpful in testing tissue engineering approaches for ACL reconstruction.


Assuntos
Ligamento Cruzado Anterior/imunologia , Ligamento Cruzado Anterior/metabolismo , Adulto , Lesões do Ligamento Cruzado Anterior , Catepsina B/metabolismo , Colágeno/metabolismo , Regulação da Expressão Gênica/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Reação em Cadeia da Polimerase , Proteoglicanas/metabolismo , RNA Mensageiro/genética , Fatores de Transcrição SOX9 , Tenascina/metabolismo , Fatores de Tempo , Fatores de Transcrição/metabolismo
17.
Osteoarthritis Cartilage ; 15(11): 1311-7, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17548214

RESUMO

OBJECTIVE: To investigate the gene expression profile and the histological aspects of articular cartilage of patients affected by Morquio syndrome, a lysosomal storage disease characterized by the accumulation of glycosaminoglycans within the cells which result in abnormal formation and growth of the skeletal system. METHOD: Articular cartilage samples were obtained from the femoral condyle of two siblings with Morquio syndrome during surgery performed to treat valgus knee. As controls, four biopsy samples of healthy cartilage were obtained from four different male multiorgan donors. A Real-Time Polymerase Chain reaction (RT-PCR) analysis was performed to evaluate the expression of type I and II collagens and aggrecan mRNAs. Histological and immunohistochemical analyses for some matrix proteins were carried out on paraffin embedded sections. RESULTS: Type I collagen mRNA mean level was higher in the samples of patients with Morquio syndrome compared to controls. Type II collagen and aggrecan mRNAs' mean expression was instead lower. The morphological appearance of the cartilage showed a poorly organized tissue structure with not homogeneously distributed cells that were larger compared to normal chondrocytes due to the presence inside the vacuoles of proteoglycans which were not metabolized. Chondrocytes were negative for collagen II immunostaining while the extracellular matrix was weakly positive. Collagen type I immunostaining was positive at cellular level. Keratan sulfate showed diffuse positivity and chondroitin-6-sulfate was present throughout the cartilaginous thickness. CONCLUSION: In cartilage of patients with Morquio syndrome, a low expression of collagen type II and a high expression of collagen type I both at protein and molecular levels are evidentiated. This finding could give evidence of the reduction in ankle and knee joint movement observable in these patients.


Assuntos
Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo I/genética , Mucopolissacaridose IV/genética , Adulto , Agrecanas , Biomarcadores/análise , Condrócitos/patologia , Colágeno Tipo I/metabolismo , Colágeno Tipo II/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Mucopolissacaridose IV/metabolismo , Mucopolissacaridose IV/patologia , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Clin Exp Rheumatol ; 25(6): 847-52, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18173918

RESUMO

OBJECTIVES: Glucosamine (GlcN) is used in the treatment of osteoarthritis as symptomatic slow-acting drug, but its mode of action is not completely known. We analyzed the influence of GlcN and its N-acetyl-phenylalanine derivative (NAPA) on mRNA transcription level of TNF-alpha-stimulated genes in cell culture. METHODS: Human immortalized chondrocyte cell line lbpva55 was stimulated with TNF-alpha and treated with GlcN and NAPA. mRNA transcription level of several genes, identified by complementary DNA microarray (cDNA microarray), was validated by Quantitative Real-Time Polymerase Chain Reaction (Q-RT-PCR). RESULTS: Several genes, whose mRNA level was increased by TNF-alpha treatment and significantly reduced by GlcN and NAPA in lbpva55 cells, were identified. These include cytokine receptors TNF-R1 and TNF-R2, their associated factor TRAF-6, signaling intermediates IGFB-6 and Rnd1, as well as cell cycle regulating proteins CUL-2 and G1S protein 1. Down- regulation of mRNA expression level of some of these genes is in accordance with inactivation of NF-kB transcription factor. Moreover, we found down-regulation of c-jun mRNA level, a component of AP-1 transcription factor. CONCLUSIONS: Our study suggests that GlcN and NAPA interfere with activation of NF-kB and AP-1 transcription factors, which are responsible for the expression of genes involved in diverse biological processes, such as cell growth and death, inflammatory and stress responses, accounting for the beneficial effects of GlcN in osteoarthritis.


Assuntos
Condrócitos/efeitos dos fármacos , Condrócitos/fisiologia , Glucosamina/farmacologia , Ativação Transcricional/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Linhagem Celular , Proteínas Culina/análise , Regulação para Baixo , Expressão Gênica , Humanos , NF-kappa B/análise , Análise de Sequência com Séries de Oligonucleotídeos , Osteoartrite , RNA Mensageiro/análise , Receptores do Fator de Necrose Tumoral/análise , Receptores Tipo II do Fator de Necrose Tumoral/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator 6 Associado a Receptor de TNF/análise , Proteínas rho de Ligação ao GTP/análise
20.
Osteoarthritis Cartilage ; 13(7): 601-7, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15979012

RESUMO

OBJECTIVE: Autologous chondrocyte implantation (ACI) has been successfully used for the treatment of osteochondral lesions of the talus. One of the main problems of this surgical strategy is related to the harvesting of the cartilage slice from a healthy knee. The aim of this study was to examine the capacity of chondrocytes harvested from a detached osteochondral fragment to proliferate and to serve as a source of viable cells for ACI in the repair of ankle cartilage defects. METHODS: Detached osteochondral fragments harvested from the ankle joint of 20 patients with osteochondral lesions of the talus served as the source of human articular cartilage specimens. All of the osteochondral lesions were chronic and of traumatic origin. In all cases, the fragments were utilized to evaluate the viability and proliferation of the cells, the histological appearance of the cartilage tissue and the expression of specific cartilage markers by real-time polymerase chain reaction (PCR). In the 16 patients scheduled for ACI, the expanded chondrocytes were used for chondrocyte implantation. In the other 4 patients, with lesion size <1.5cm(2), microfractures were created during the initial arthroscopic step. As a control group, 7 patients with comparable osteochondral lesions underwent the same surgery, but received chondrocytes harvested from the ipsilateral knee. RESULTS: According to the American Orthopaedic Foot and Ankle Scoring (AOFAS) system, patients in the experimental group had a preoperative score of 54.2+/-16 points and a postoperative one of 89+/-9.6 points after a minimum follow-up time of 12 months (P<0.0005). The control group of patients had a preoperative score of 54.6+/-11.7 points and a postoperative one of 90.2+/-9.7 points at a minimum follow-up time of 12 months (P<0.0005). The clinical results of the two groups did not differ significantly from each other. Chondrocytes isolated from the detached fragments were highly viable, phenotypically stable, proliferated in culture and redifferentiated when grown within the three-dimensional scaffold used for ACI. The morphological and molecular characteristics of the cartilage samples obtained from the detached osteochondral fragments were similar to those of healthy hyaline articular cartilage. CONCLUSIONS: The good results achieved with this strategy indicate that cells derived from the lesioned area may be useful in the treatment of osteochondral defects of the talus.


Assuntos
Articulação do Tornozelo/cirurgia , Cartilagem Articular/cirurgia , Condrócitos/transplante , Osteocondrite/cirurgia , Adulto , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Células Cultivadas , Feminino , Humanos , Imuno-Histoquímica , Masculino , Osteocondrite/patologia , Reação em Cadeia da Polimerase , Tálus/cirurgia , Transplante Autólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA