Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Cell Physiol ; 232(12): 3574-3585, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28121025

RESUMO

Mesenchymal stromal cells (MSCs) are key players in the repair or regeneration of the damaged bone tissue. However, heterogeneity exists between MSCs derived from different donors in their bone formation ability both in vitro and in vivo. The identification of markers defining MSCs with different functional phenotypes is fundamental to maximize their clinical potential. In our previous in vivo study, impaired expression in MSCs of cystathionine-ß-synthase (CBS) and cystathionine-γ-lyase (CSE), the two key enzymes in the catabolic pathway of homocysteine, was associated to decreased bone formation and to the onset of osteoporosis in mice. Here, we investigated whether osteogenic differentiation of human MSCs (hMSCs) modulates the expression of CBS and CSE. The expression of CBS and CSE was also assessed during chondrogenesis to confirm the specificity of their expression during osteogenesis. hMSCs displayed a heterogeneous mineralizing capacity between donors (70% of the samples mineralized, while 30% did not mineralize). Inducible expression of CBS and CSE was found to be associated with a mineralizing phenotype in hMSCs. In particular, up-regulation of CSE was restricted to hMSCs undergoing mineralization. During chondrogenesis, CBS was significantly up-regulated while CSE expression was not affected. Ex-vivo findings confirmed that mature h-osteoblasts (hOBs) show consistently higher expression of CBS and CSE than hMSCs. Our data provide the first evidence that the expression of CBS and CSE in hMSCs closely correlates with the transition of hMSCs toward the osteoblastic phenotype and that CSE may constitute a novel marker of osteogenic differentiation.


Assuntos
Calcificação Fisiológica , Diferenciação Celular , Cistationina beta-Sintase/metabolismo , Cistationina gama-Liase/metabolismo , Células-Tronco Mesenquimais/enzimologia , Osteoblastos/enzimologia , Osteogênese , Biomarcadores/metabolismo , Proliferação de Células , Células Cultivadas , Condrogênese , Humanos , Fenótipo , Fatores de Tempo
2.
Arthritis Rheum ; 65(5): 1271-81, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23613363

RESUMO

OBJECTIVE: To examine the effect of different sources of good manufacturing practice clinical grade adipose-derived mesenchymal stem cells (AD-MSCs) on inflammatory factors in osteoarthritic (OA) chondrocytes and synoviocytes. METHODS: AD-MSCs from infrapatellar Hoffa fat, subcutaneous (SC) hip fat, and SC abdominal fat were cocultured in Transwells with chondrocytes or synoviocytes. Inflammatory factors (interleukin-1ß [IL-1ß], tumor necrosis factor α, IL-6, CXCL1/growth-related oncogene α, CXCL8/IL-8, CCL2/monocyte chemotactic protein 1, CCL3/macrophage inflammatory protein 1α, and CCL5/RANTES) were evaluated by quantitative reverse transcription-polymerase chain reaction or multiplex bead-based immunoassay. The role of different immunomodulators was analyzed. RESULTS: All the inflammatory factors analyzed were down-modulated at the messenger RNA or protein level independently by all 3 AD-MSC sources or by allogeneic AD-MSCs used in coculture with chondrocytes or synoviocytes. Inflammatory factor down-modulation was observed only when AD-MSCs were cocultured with chondrocytes or synoviocytes that produced high levels of inflammatory factors, but no effect was observed in cells that produced low levels of those factors, thus highlighting a dependence of the AD-MSC effect on existing inflammation. The immunomodulators IL-10, IL-1 receptor antagonist, fibroblast growth factor 2, indoleamine 2,3-dioxygenase 1, and galectin 1 were not involved in AD-MSC effects, whereas the cyclooxygenase 2 (COX-2)/prostaglandin E2 (PGE2 ) pathway exerted a role in the mechanism of antiinflammatory AD-MSC action. CONCLUSION: The antiinflammatory effects of AD-MSCs are probably not dependent on AD-MSC adipose tissue sources and donors but rather on the inflammatory status of OA chondrocytes and synoviocytes. AD-MSCs seem to be able to sense and respond to the local environment. Even though a combination of different molecules may be involved in AD-MSC effects, the COX-2/PGE2 pathway may play a role, suggesting that AD-MSCs may be useful for therapies in osteoarticular diseases.


Assuntos
Adipócitos/citologia , Condrócitos/citologia , Dinoprostona/metabolismo , Células-Tronco Mesenquimais/citologia , Osteoartrite/patologia , Membrana Sinovial/citologia , Idoso , Biomarcadores/metabolismo , Cartilagem Articular/patologia , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Condrócitos/metabolismo , Técnicas de Cocultura , Regulação para Baixo , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Membrana Sinovial/metabolismo
3.
J Cell Physiol ; 227(8): 3151-61, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22034088

RESUMO

Fluctuation in extracellular calcium (Ca(2+)) concentration occurs during bone remodeling. Free ionized Ca(2+) plays a critical role in regulating osteoblast functions. We analyzed the effects of different concentrations of free ionized Ca(2+) (0.5, 1.3, and 2.6 mM) on human osteoblasts and we evaluated osteoblastic phenotype (marker expression and cell morphology) and functions (osteogenic differentiation, cell proliferation, and cell signaling). Our data show human osteoblasts that chronically stimulated with 0.5, 1.3, or 2.6 mM Ca(2+) significantly increase intracellular content of alkaline phosphatase, collagen type I, osteocalcin, and bone sialoprotein, whereas collagen type XV was down-modulated and RUNX2 expression was not affected. We also found a Ca(2+) concentration-dependent increase in osteogenic differentiation and cell proliferation, associated to an increase of signaling protein PLCß1 and p-ERK. Human osteoblast morphology was affected by Ca(2+) as seen by the presence of numerous nucleoli, cells in mitosis, cell junctions, and an increased number of vacuoles. In conclusion, our data show a clear phenotypical and functional effect of extracellular Ca(2+) on human osteoblasts and support the hypothesis of a direct role of this cation in the bone remodeling processes.


Assuntos
Cálcio/metabolismo , Cálcio/farmacologia , Colágeno/metabolismo , Osteoblastos/efeitos dos fármacos , Osteocalcina/metabolismo , Idoso , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Remodelação Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno/genética , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Sialoproteína de Ligação à Integrina/genética , Sialoproteína de Ligação à Integrina/metabolismo , Osteoblastos/metabolismo , Osteocalcina/genética
4.
Exp Cell Res ; 317(8): 1159-68, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21182836

RESUMO

CXCL12/CXCR4 chemokine/receptor axis signaling has recently been found to play an important role in the remodeling of bone tissue, but little is known about the molecular mechanisms that are involved. The present study shows that CXCL12 is present at high levels both in human mesenchymal stem cells (hMSCs) and primary osteoblasts (hOBs). When osteogenesis was induced, CXCL12 expression was strictly confined to mineralized nodules. To investigate what mechanisms contribute to the maintenance of a correct expression of CXCL12 in bone cellular context, we analyzed the relationship between CXCL12 and Slug, a transcription factor recently associated with osteoblast maturation. By gene silencing and chromatin immunoprecipitation assay, we showed that both proteins are required for the mineralization process and CXCL12 is transcriptionally and functionally regulated by Slug, which is recruited at specific sites to its gene promoter in vivo. These findings showed for the first time a positive correlation between CXCL12 signaling and Slug activity, thus corroborating the role of these two proteins in bone cellular context and suggesting a new potential target for bone tissue repair and regeneration.


Assuntos
Quimiocina CXCL12/metabolismo , Osteoblastos/fisiologia , Fatores de Transcrição/metabolismo , Células Cultivadas , Quimiocina CXCL12/genética , Imunoprecipitação da Cromatina , Inativação Gênica , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Osteoblastos/citologia , Regiões Promotoras Genéticas , Transdução de Sinais/fisiologia , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética
5.
J Cell Physiol ; 226(4): 982-90, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20857429

RESUMO

T cells are critical regulators of osteoclast differentiation and function in bone, but whether osteoclasts can, in turn, regulate T cell homing, and response to stimuli is unclear. To investigate whether osteoclasts are immune competent cells, the expression of HLA Class II and costimulatory receptors was evaluated by RT-PCR and immunohistochemistry by comparing osteoclast precursors and mature osteoclasts. T-cell-attracting chemokines were measured in the supernatants of confluent cultures of osteoclasts and compared with mesenchymal stromal cells and osteoblasts. T cell proliferation, cytokine production, and apoptosis were assayed in co-cultures with osteoclasts in the presence or absence of mitogenic stimuli. To define the mechanism of action of osteoclasts, cytokine-blocking experiments were performed. Our findings revealed that mature osteoclasts constitutively expressed Class II HLA in the membrane and upregulate the expression of CD40 and CD80 during differentiation. Osteoclasts secreted high levels of most T cell chemoattractants and effectively retained T cells in adhesion assays. Moreover, the osteoclasts potently blunted T cell response to PHA and CD3/CD28 stimulation, thus inhibiting proliferation, suppressing T cell TNFα and IFNγ production and decreasing T cell apoptosis by a mostly cell-contact independent mechanism. In conclusion, osteoclasts are immune-competent cells which can retain T cells and suppress in vitro T cell response to proliferative stimuli.


Assuntos
Osteoclastos/citologia , Linfócitos T/citologia , Antígenos/imunologia , Apoptose/imunologia , Biomarcadores , Diferenciação Celular/imunologia , Proliferação de Células , Fragmentação do DNA , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Interferon gama/biossíntese , Osteoclastos/metabolismo , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/biossíntese
6.
J Cell Physiol ; 226(10): 2675-82, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21302278

RESUMO

Human bone cells used for in vitro studies are mainly derived from bone marrow (BM) or trabecular bone (TB). There are no specific markers or procedures for isolation and growth of these cells. To validate the potentiality of these cells, we isolated human mesenchymal stromal cells (MSCs) and osteoblasts (OBs) from the tibial plateau of the same subject, grown in two different media (α-MEM and DMEM/F12) and analyzed for cell growth, proliferation, phenotype and osteogenic potential. We found that OBs grew well in both media tested, but MSCs were able to grow only in α-MEM medium. OBs in DMEM/F12 showed reduced proliferation capability and expressed a low level of alkaline phosphatase (AP), RUNX-2, osteocalcin (OC), bone sialoprotein (BSP), collagen type I (Col.I) compared with OBs in α-MEM but high level of collagen type XV (Col.XV). Compared with MSCs in α-MEM, OBs have an increased ability to proliferate and express more OC and BSP at molecular level but less AP, RUNX-2 and Col.I than MSCs. Time-course experiments to analyze the osteogenic potential of these cells showed that OBs were more efficient than MSCs. However, these cells obtained from tibial plateau showed a different trend of AP, OC and Col.I osteogenic markers compared to control MSCs from the iliac crest. This study shows that bone-adherent OBs grown in α-MEM medium are more efficient for osteogenic differentiation than BM MSCs and contribute to defining their phenotypic and functional characteristics, so providing a rationale for their use in bone tissue engineering or therapeutic purposes.


Assuntos
Células da Medula Óssea/citologia , Osteogênese/fisiologia , Células-Tronco/citologia , Tíbia/citologia , Idoso , Células da Medula Óssea/fisiologia , Separação Celular/métodos , Células Cultivadas , Humanos , Pessoa de Meia-Idade , Osteoartrite do Joelho/cirurgia , Osteoblastos/citologia , Osteoblastos/fisiologia , Osteogênese/genética , Células-Tronco/fisiologia , Células Estromais/citologia , Células Estromais/fisiologia , Coleta de Tecidos e Órgãos/métodos
7.
J Cell Physiol ; 221(1): 154-60, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19492413

RESUMO

Subchondral bone remodeling in osteoarthritis (OA) and rheumatoid arthritis (RA) is mainly characterized by the formation of osteophytes/fibrosis and by the presence of infiltrating cells associated to bone resorption. In this study we analyzed CC (cysteine cysteine motif) chemokine ligand (CCL)20 and CC chemokine receptor (CCR)6 function in subchondral bone tissue and osteoblasts isolated from OA and RA patients. CCL20/CCR6 expression was evaluated by immunohistochemical techniques in bone tissue from OA and RA patients. CCL20-functional tests were performed on osteoblasts isolated from OA and RA patients to evaluate enzymatic response and cell proliferation. Moreover, we assessed Akt phosphorylation as the major signaling pathway for CCL20. In bone tissue biopsies we found that osteoblasts from both OA and RA patients expressed CCR6 while CCL20 was expressed only by RA osteoblasts. Both CCR6 and CCL20 were highly expressed in osteocytes and mononuclear cells from only RA patients. CCL20-stimulated OA osteoblasts showed a significant increase in beta-N-acetylhexosaminidase release compared to RA. Conversely, a significant increase in cellular proliferation was found only in CCL20-stimulated RA osteoblasts associated to Akt phosphorylation. These data were confirmed in bone tissue biopsies. This study demonstrates a different expression of CCL20-positive osteoblasts in OA versus RA disease that seem to be associated with the presence of infiltrating mononuclear cells. Moreover, CCL20 stimulation resulted in a greater proliferative response in RA osteoblasts compared to OA osteoblasts, mediated by Akt signaling, while OA osteoblasts showed increased enzymatic activity, thus suggesting a differential role of this chemokine in OA and RA.


Assuntos
Artrite Reumatoide/metabolismo , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Quimiocina CCL20/metabolismo , Osteoartrite/metabolismo , Osteoblastos/metabolismo , Receptores CCR6/metabolismo , Idoso , Artrite Reumatoide/enzimologia , Artrite Reumatoide/patologia , Biópsia , Osso e Ossos/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Separação Celular , Quimiocina CCL20/farmacologia , Exocitose/efeitos dos fármacos , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/enzimologia , Leucócitos Mononucleares/patologia , Masculino , Osteoartrite/enzimologia , Osteoartrite/patologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/enzimologia , Osteoblastos/patologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , beta-N-Acetil-Hexosaminidases/metabolismo
8.
J Cell Physiol ; 220(2): 401-9, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19365806

RESUMO

Bone marrow stromal cells (MSCs) and osteoblasts are the two main non-haematopoietic cellular components of human bone tissue. To identify novel osteoblast-related molecules, we performed a gene expression profiling analysis comparing MSCs and osteoblasts isolated from the same donors. Genes differentially overexpressed in osteoblasts were mainly related to the negative control of cell proliferation, pro-apoptotic processes, protein metabolism and bone remodelling. Notably, we also identified the collagen XV (COL15A1) gene as the most up-regulated gene in osteoblasts compared with MSCs, previously described as being expressed in the basement membrane in other cell types. The expression of collagen type XV was confirmed at the protein level on isolated osteoblasts and we demonstrated that it significantly increases during the osteogenic differentiation of MSCs in vitro and that free ionised extracellular calcium significantly down-modulates its expression. Moreover, light and electron microscopy showed that collagen type XV is expressed in bone tissue biopsies mainly by working osteoblasts forming new bone tissue or lining bone trabeculae. To our knowledge, these data represent the first evidence of the expression of collagen type XV in human osteoblasts, a calcium-regulated protein which correlates to a specific functional state of these cells.


Assuntos
Colágeno/metabolismo , Matriz Extracelular/metabolismo , Perfilação da Expressão Gênica , Análise em Microsséries , Osteoblastos/metabolismo , Idoso , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Cálcio/metabolismo , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Colágeno/genética , Matriz Extracelular/química , Humanos , Pessoa de Meia-Idade , Osteoblastos/citologia , Osteogênese/fisiologia , Fenótipo , Células Estromais/citologia , Células Estromais/metabolismo
9.
J Biomed Mater Res A ; 77(3): 497-506, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16482549

RESUMO

Chondrogenesis is a complex process characterized by a sequence of different steps that start with the condensation of the cells, followed by the expression of specific components, such as collagens and proteoglycans. We evaluated in vitro chondrogenic differentiation of C3H10T1/2 murine mesenchymal cells and compared them with human mesenchymal stromal cells (h-MSCs) in a hyaluronic acid scaffold. We analyzed (from day 0 to day 28) cellular morphology, proliferation, and chondrogenic/osteogenic gene expression at different time points. Our data demonstrate that, during chondrogenic differentiation, murine cells proliferate both in the absence and presence of TGFbeta, while h-MSCs require the presence of this activating factor. Murine cells, even if viable, differentiate on hyaluronan scaffold, maintain a fibroblastic morphology, and form a capsule outside the scaffold. At the mRNA level, murine cells showed a decrease in collagen type I combined with a significant increase in collagen type II (from day 0), and aggrecan (on day 28), as found for h-MSCs. Immunohistochemical data confirmed that chondrogenic differentiation of murine cells, induced by TGFbeta, occurred only in some restricted areas inside the scaffold that were positive to collagen type II, but did not show a cartilage-like tissue structure, as we had found using h-MSCs. These data demonstrate that C3H10T1/2 murine cells, widely used as a chondrogenic model, show a different sequence of chondrogenic events in hyaluronic acid scaffold, compared with primary h-MSCs.


Assuntos
Materiais Biocompatíveis , Cartilagem/citologia , Diferenciação Celular/fisiologia , Expressão Gênica/fisiologia , Ácido Hialurônico , Animais , Condrogênese/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C3H , Células Estromais/citologia , Células Estromais/fisiologia
10.
J Tissue Eng Regen Med ; 10(4): 305-14, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23653421

RESUMO

T lymphocytes play a key role in the regulation of bone homeostasis and bone healing. The inflammatory response at the site of bone injury is essential to the initiation of the bone repair program; however, an uncontrolled exposure to inflammatory environment has a negative effect on tissue regeneration - indeed, activated T cells were shown to inhibit osteogenic differentiation on human mesenchymal stromal cells (MSCs). Whether resting T cells can induce osteogenic differentiation of MSCs and what role specific T cells subset play in this process is still elusive. In this study, we sought to analyse the osteogenic gene expression profile of whole T cells, CD4 and CD8 T cells isolated from healthy donors and investigated whether secreted factors from each group modulate osteogenic differentiation of human MSCs. Gene expression profiling identified a pool of 51 genes involved at various stages in bone growth which are expressed above detectable levels in CD4 and CD8 T cells. Most genes of this pool were expressed at higher levels in the CD4 subset. In vitro mineralization assays revealed that conditioned medium from CD4 T cells, but not from CD8 cells, significantly increased mineralization in osteogenic cultures of human MSCs; furthermore, mRNA expression of Runt-related transcription factor 2 (RUNX-2), osteocalcin (OC), bone sialoprotein (BSP) and alkaline phosphatase (ALP) in MSCs was significantly upregulated in the presence of CD4-conditioned medium but not with that obtained from CD8. The results show a differential role for CD4 and CD8 T cells in supporting bone formation and identify an osteogenic gene signature of each subset.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Osteogênese , Subpopulações de Linfócitos T/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/genética , Separação Celular , Análise por Conglomerados , Perfilação da Expressão Gênica , Humanos , Células-Tronco Mesenquimais/metabolismo , Osteogênese/genética , Fenótipo , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
11.
Biomaterials ; 26(28): 5677-86, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15878373

RESUMO

Mesenchymal stromal cells (MSCs) seem to be a good alternative to chondrocytes for cartilage regeneration. To obtain new information on the sequence of cellular and molecular events during in vitro chondrogenic differentiation we analysed MSCs on a widely used hyaluronic acid biomaterial (Hyaff-11). Cellular differentiation was induced using two different concentrations of TGFbeta1 (10 and 20 ng/ml) and the process was analysed at different time points (24 h, and 7, 14, 21 and 28 days) using techniques of light and electron microscopy, real-time PCR and immunohistochemistry. We found that without TGFbeta MSCs did not survive while in the presence of TGFbeta the cells significantly proliferated from day 7 until day 28. Light and electron microscopy showed that TGFbeta at 20 ng/ml better induced the formation of cartilage-like tissue. Real-time PCR showed an increased expression of collagen type II, IX and aggrecan associated to a down-regulation of collagen type I. Immunohistochemical analysis confirmed that collagen type I was down-modulated while collagen type II increased from day 14 to day 28. These data clearly showed that higher concentrations of TGFbeta (20 ng/ml) induce chondrogenesis of MSCs on Hyaff-11 scaffold better than 10 ng/ml of TGFbeta. This process is characterized by a sequence of cellular and molecular events that deal with the in vitro formation of a cartilage-like tissue.


Assuntos
Condrócitos/citologia , Condrócitos/fisiologia , Condrogênese/fisiologia , Ácido Hialurônico/química , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Engenharia Tecidual/métodos , Materiais Biocompatíveis/química , Diferenciação Celular/fisiologia , Proliferação de Células , Sobrevivência Celular/fisiologia , Células Cultivadas , Humanos , Teste de Materiais , Células Estromais/citologia , Células Estromais/fisiologia
12.
J Bone Joint Surg Am ; 87(1): 46-57, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15634813

RESUMO

BACKGROUND: There are only a few studies concerning the cellular, biochemical, and genetic processes that occur during the remodeling of graft tissue after autologous chondrocyte transplantation. The purpose of the present study was to quantify the expression of genes encoding extracellular matrix proteins and regulatory factors that are essential for cell differentiation in cartilage biopsy specimens from patients who had this treatment two years previously. METHODS: Two cartilage biopsy specimens from each of four patients who had been treated with autologous chondrocyte transplantation and from two multiorgan donors were used. Real-time reverse transcriptase-polymerase chain reaction analysis was performed to evaluate the expression of types I, II, and X collagen; aggrecan; cathepsin B; and early growth response protein-1 (Egr-1) and Sry-type high-mobility-group box transcription factor-9 (Sox-9) mRNAs. Immunohistochemical analysis for matrix proteins and regulatory proteins was carried out on paraffin-embedded sections. RESULTS: Type-I collagen mRNA was expressed in all of the samples evaluated. Type-II collagen was present in autologous chondrocyte transplantation samples but at lower levels than in the controls. Type-X collagen messenger was undetectable. Aggrecan mRNA was present in all of the samples at lower levels than in the controls, while cathepsin-B messenger levels were higher and Egr-1 and Sox-9 mRNAs were expressed at lower levels. The immunohistochemical analysis showed slight positivity for type-I collagen in all of the sections. Type-II collagen was found in all of the samples with positivity confined inside the cells, while the controls displayed a positivity that was diffuse in the extracellular matrix. Cathepsin B was slightly positive in all of the samples, while the controls were negative. Egr-1 protein was particularly evident in the areas negative for type-II collagen. Sox-9 was positive in all samples, with evident localization in the superficial and middle layers. CONCLUSIONS: In biopsy specimens from autologous chondrocyte transplantation tissue at two years, there is evidence of the formation of new tissue, which displays varying degrees of organization with some fibrous and fibrocartilaginous features. Long-term follow-up investigations are needed to verify whether, once all of the remodeling processes are completed, the newly formed tissue will acquire the more typical features of articular cartilage.


Assuntos
Cartilagem/citologia , Condrócitos/transplante , Adulto , Agrecanas , Cartilagem Articular/cirurgia , Catepsina B/genética , Proteoglicanas de Sulfatos de Condroitina/genética , Colágeno/genética , Proteínas de Ligação a DNA/genética , Proteína 1 de Resposta de Crescimento Precoce , Proteínas da Matriz Extracelular/genética , Feminino , Proteínas de Grupo de Alta Mobilidade/genética , Humanos , Proteínas Imediatamente Precoces/genética , Imuno-Histoquímica , Lectinas Tipo C , Masculino , Proteoglicanas/genética , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOX9 , Fatores de Tempo , Fatores de Transcrição/genética , Transplante Autólogo , Dedos de Zinco/genética
13.
Exp Gerontol ; 39(4): 659-65, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15050303

RESUMO

Bone homeostasis is regulated by cells at different stages of maturation that are influenced by soluble factors. The modulatory function of two pro-inflammatory cytokines, IL-1beta and TNF-alpha, on the expression of CXCL13 chemokine was evaluated in osteoblasts (OB) and bone marrow stromal cells (BMSC) from osteoarthritis (OA) and post-traumatic (PT) patients. In basal condition, CXCL13 production by both BMSC and OB was significantly higher in OA than in PT patients. IL1beta, significantly induced CXCL13 production in differentiated OB, both from OA and PT patients, but not in BMSC from both either group. TNFalpha reduced CXCL13 production only in BMSC from OA patients. The combination of IL1beta and TNFalpha increased CXCL13 production only in OB in the same amount as for IL-1beta alone. OB from OA released a higher amount of CXCL13 compared to PT in all conditions tested. CD121a (IL1 receptor type I) was highly expressed only by OB. Moreover, in bone tissue biopsies CXCL13 was expressed by mesenchymal and mononuclear cells. This study demonstrates that cells at different stages of maturation (BMSC and OB) and derived from physiological (PT) or pathological conditions (OA) respond in different ways to inflammatory stimuli. These data may contribute to understand the basic maturation processes of bone cells in old patients.


Assuntos
Quimiocinas CXC/biossíntese , Osteoartrite/metabolismo , Osteoblastos/metabolismo , Células Estromais/metabolismo , Adulto , Idoso , Antígenos CD/metabolismo , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Quimiocina CXCL13 , Feminino , Humanos , Interleucina-1/farmacologia , Masculino , Pessoa de Meia-Idade , Osteoartrite/patologia , Osteoblastos/efeitos dos fármacos , Receptores de Interleucina-1/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral , Células Estromais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
14.
Biomaterials ; 24(10): 1751-7, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12593957

RESUMO

The clinical need for improved human autologous chondrocyte transplantation has motivated the use of different biomaterials, which are aimed at fixing the cells in the defect area and permit their proliferation and differentiation. The maintenance of the original phenotype by isolated chondrocytes grown in vitro is an important requisite for their use in repairing damaged articular cartilage. The methods to verify the expression of cartilage-specific molecules usually involve destructive procedures to recover the cells from the scaffolds for tests. The aim of our study was to find a soluble marker able to attest the occurrence of a differentiation process by chondrocytes grown onto a biomaterial used for cell transplantation. We turned our attention to cathepsin B which is known to be abnormally synthesized in de-differentiated chondrocytes and scarcely produced in the differentiated ones. The production of cathepsin B by human articular chondrocytes expanded in vitro and then grown onto a hyaluronan-based polymer derivative (Hyaff-11) three-dimensional scaffold was evaluated with a specific enzyme-immunoassay at different experimental times together with the expression of mRNA by real-time PCR. We showed that cathepsin B, which is abundantly produced by chondrocytes grown in a monolayer culture, decreases significantly after the cells are seeded onto the scaffold, giving further evidence of a re-differentiation process. This result suggests cathepsin B a practical soluble marker to evaluate the "good" quality of transplantable constructs.


Assuntos
Cartilagem Articular/transplante , Catepsina B/metabolismo , Engenharia Tecidual/métodos , Materiais Biocompatíveis , Biomarcadores , Fenômenos Biomecânicos , Catepsina B/genética , Diferenciação Celular , Divisão Celular , Condrócitos/citologia , Condrócitos/metabolismo , Condrócitos/transplante , Expressão Gênica , Humanos , Ácido Hialurônico/análogos & derivados , Imuno-Histoquímica , Teste de Materiais , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Solubilidade , Transplante Autólogo
15.
Biomaterials ; 23(4): 1187-95, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11791922

RESUMO

Association of biomaterials with autologous cells can provide a new generation of implantable devices for cartilage repair. Such scaffolds should provide a preformed three-dimensional shape and prevent cells from escaping into the articular cavity. Furthermore, these constructs should have sufficient mechanical strength to facilitate handling in a clinical setting and stimulate the uniform spreading of cells and their phenotype redifferentiation. The aim of this study was to verify the ability of HYAFF 11, a recently developed hyaluronic-acid-based biodegradable polymer, to support the growth of human chondrocytes and to maintain their original phenotype. This capability was assessed by the evaluation of collagen types I, II and aggrecan mRNA expression. Immunohistochemical analyses were also performed to evaluate collagen types I, II and proteoglycans synthesis. A field emission in lens scanning microscopy was utilized to verify the interactions between the cells and the biomaterial. Our data indicate that human chondrocytes seeded on HYAFF 11 express and produce collagen type II and aggrecan and downregulate the production of collagen type I. These results provide an in vitro demonstration for the therapeutic potential of HYAFF 11 as a delivery vehicle in a tissue-engineered approach towards the repair of articular cartilage defects.


Assuntos
Materiais Biocompatíveis , Condrócitos/citologia , Proteínas da Matriz Extracelular , Ácido Hialurônico/análogos & derivados , Agrecanas , Diferenciação Celular , Divisão Celular , Células Cultivadas , Condrócitos/metabolismo , Condrócitos/ultraestrutura , Colágeno Tipo I/genética , Colágeno Tipo II/genética , Humanos , Imuno-Histoquímica , Lectinas Tipo C , Teste de Materiais , Microscopia Eletrônica de Varredura , Proteoglicanas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
16.
J Biomed Mater Res A ; 89(3): 687-96, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18442123

RESUMO

Surface properties affect the biological properties of cells modulating the expression of different factors. Osteoblasts contribute both to new bone formation and controlling haematopoiesis through cytokines and growth factors. We analyzed the effect of bone (calcium-phosphate bone slides), cartilaginous (hyaluronan-based scaffold), and plastic substrate culture on human osteoblast proliferation, bone matrix molecule, and inflammatory factor expression. Osteoblast proliferation increased to a greater extent when the cells were grown for 14 days on plastic and bone slides, whereas hyaluronan-based scaffold (HA-scaffold) induced only a minimal increase. Collagen type I, osteonectin, alkaline phosphatase and osteocalcin were expressed on osteoblasts grown on plastic and bone slides and down-modulated at mRNA and protein level by HA-scaffold. Bone slides showed the ability to increase osteopontin mRNA expression. The expression of CXCR4 and CXCL13 was upregulated by bone slides and HA-scaffold, while CXCL12 and CXCR5 expression was down-modulated. These data suggest a substrate-dependent modulation of human osteoblast proliferation, bone matrix and inflammatory factor expression, which might help to understand the active role played by osteoblasts in bone microenvironment by coupling bone extracellular matrix, chemokines and the haematopoietic system.


Assuntos
Matriz Óssea/metabolismo , Mediadores da Inflamação/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Idoso , Matriz Óssea/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quimiocinas/metabolismo , Durapatita/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Osteoblastos/efeitos dos fármacos , Plásticos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Propriedades de Superfície/efeitos dos fármacos , Alicerces Teciduais
17.
Tissue Eng Part A ; 14(1): 97-105, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18333808

RESUMO

Chemokines contribute to the maintenance of cartilage homeostasis. To evaluate the role of CXC chemokines CXCL8 (interleukin-8), CXCL10 (interferon-gamma-inducible protein-10), CXCL12 (stroma-derived factor-1) and CXCL13 (B-cell attracting chemokine-1) and their receptors, respectively CXCR1-2, CXCR3, CXCR4, and CXCR5, during chondrogenic differentiation of human mesenchymal stromal cells (h-MSCs), we used a well-defined in vitro model. Chondrogenic differentiation was analyzed on h-MSCs grown on hyaluronic acid-based biomaterial in the presence or absence of transforming growth factor-beta, and the expression and modulation of CXC chemokines and receptors were evaluated at different time points. Real-time polymerase chain reaction was performed to analyze their expression at the messenger ribonucleic acid (mRNA) level, and immunohistochemistry and enzyme-linked immunosorbent assay were used to evaluate their expression at the protein level. Human articular cartilage biopsies were used to evaluate chemokine and receptor expression in normal tissue. We found no expression of CXCR1, CXCR2, CXCR3, or CXCL10 at the mRNA level. CXCL8 mRNA was down-modulated, whereas at the protein level, we found greater release of this chemokine. CXCR4 and its ligand CXCL12 were down-modulated during chondrogenesis. By contrast, CXCR5 was up-regulated, whereas its ligand CXCL13 was lower. These data were also confirmed on human articular cartilage. These findings show that, during in vitro h-MSC chondrogenic differentiation, chemokine and receptor expression was specifically induced or repressed. This was in line with what the authors also found in normal articular cartilage, suggesting a role in differentiation and maturation of a cartilage-like structure in vitro and consequently the regulation of cartilage homeostasis.


Assuntos
Cartilagem Articular/metabolismo , Diferenciação Celular/fisiologia , Quimiocinas CXC/biossíntese , Condrócitos/metabolismo , Condrogênese/fisiologia , Células-Tronco Mesenquimais/metabolismo , Receptores CXCR/biossíntese , Cartilagem Articular/citologia , Diferenciação Celular/efeitos dos fármacos , Condrócitos/citologia , Condrogênese/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Humanos , Ácido Hialurônico , Imuno-Histoquímica , Células-Tronco Mesenquimais/citologia , RNA Mensageiro/biossíntese , Fator de Crescimento Transformador beta/farmacologia
18.
J Cell Physiol ; 210(3): 798-806, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17133360

RESUMO

We evaluated the role of CCL20 (MIP-3alpha) chemokine in cells directly involved in the remodeling of bone tissue (osteoblasts and osteoclasts) and we confirmed its expression in the subchondral bone tissue of rheumatoid arthritis (RA) patients. The expression of CCL20 and of its receptor CCR6 was evaluated in osteoblasts isolated from bone tissue of post-traumatic (PT) patients. Functional tests were performed to evaluate osteoblast proliferation and matrix protein modulation. Immunohistochemical analysis for CCR6, CCL20, and RANKL was performed on bone samples from RA patients. The role of CCL20 was then analyzed in osteoclast differentiation. We found that in basal conditions CCR6, but not its ligand CCL20, was highly expressed by osteoblasts. Functional analysis on osteoblasts showed that CCL20 significantly increased cellular proliferation but did not affect matrix protein expression. Pro-inflammatory cytokines significantly induced the release of CCL20 and RANKL by human osteoblasts but did not modulate CCR6 expression. Increased expression of CCR6, CCL20, and RANKL was confirmed in RA subchondral bone tissue biopsies. We demonstrated that CCL20 was also an earlier inducer of osteoclast differentiation by increasing the number of pre-osteoclasts, thus favoring cell fusion and MMP-9 release. Our results add new insight to the important role of the CCL20/CCR6, RANKL system in the bone tissue of RA. The contemporary action of CCL20 on osteoblasts and osteoclasts involved in the maintenance of bone tissue homeostasis demonstrates the important role of this compartment in the evolution of RA, by showing a clear uncoupling between new bone formation and bone resorption.


Assuntos
Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Proliferação de Células , Quimiocinas CC/fisiologia , Proteínas Inflamatórias de Macrófagos/fisiologia , Osteoblastos/patologia , Osteoclastos/patologia , Idoso , Biópsia , Remodelação Óssea , Diferenciação Celular , Células Cultivadas , Quimiocina CCL20 , Quimiocinas CC/genética , Regulação da Expressão Gênica , Homeostase , Humanos , Proteínas Inflamatórias de Macrófagos/genética , Metaloproteinase 9 da Matriz/metabolismo , Pessoa de Meia-Idade , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Ligante RANK/genética , Ligante RANK/metabolismo , Receptores CCR6 , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo
19.
J Cell Physiol ; 206(1): 78-85, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15965952

RESUMO

To evaluate the role of CXC chemokines CXCL8 (IL8), CXCL10 (IP-10), CXCL12 (SDF-1), and CXCL13 (BCA-1) in bone remodeling, we analyzed their effects on osteoblasts (OBs) obtained from subchondral trabecular bone tissue of osteoarthritis (OA) and post-traumatic (PT) patients. The expression of CXC receptors/ligands (CXCR1/CXCL8, CXCR2/CXCL8, CXCR3/CXCL10, CXCR4/CXCL12, and CXCR5/CXCL13) was analyzed in cultured OBs by flow cytometry and immunocytochemistry. Functional assays on CXC chemokine-treated-OBs in the presence or absence of their specific inhibitors were performed to analyze cellular proliferation and the enzymatic response to chemokine activation. The expression of chemokine ligands/receptors was also confirmed in bone tissue samples by immunohistochemical analysis. Collagen type I and alkaline phosphatase mRNA expression were analyzed on CXCL12- and CXCL13-treated OBs by real-time PCR. OBs from both OA and PT patients expressed high levels of CXCR3 and CXCR5 and lower amounts of CXCR1 and CXCR4. CXCL12 and CXCL13, only in OBs from OA patients, induced a significant proliferation that was also confirmed by specific blocking experiments. Moreover, OBs from OA patients released a higher amount of CXCL13 than those of PT patients while no differences were found for CXCL12. In the remodeling area of bone tissue samples, immunohistochemical analysis confirmed that OBs expressed CXCL12/CXCR4 and CXCL13/CXCR5 both in OA and PT samples. CXCL12 and CXCL13 upregulated collagen type I mRNA expression in OBs from OA patients. These data suggest that CXCL12 and CXCL13 may directly modulate cellular proliferation and collagen type I in OA patients, so contributing to the remodeling process that occurs in the evolution of this disease.


Assuntos
Proliferação de Células , Quimiocinas CXC/metabolismo , Colágeno Tipo I/metabolismo , Osteoartrite/metabolismo , Osteoblastos/fisiologia , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Células Cultivadas , Quimiocina CXCL10 , Quimiocina CXCL12 , Quimiocina CXCL13 , Colágeno Tipo I/genética , Exocitose/fisiologia , Humanos , Interleucina-8/metabolismo , Osteoartrite/patologia , Osteoblastos/citologia , Fenótipo , Ligação Proteica , Receptores de Quimiocinas/metabolismo , Tíbia/citologia , Tíbia/lesões , beta-N-Acetil-Hexosaminidases/metabolismo
20.
J Cell Physiol ; 207(2): 364-73, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16331675

RESUMO

Hyaluronan (HA), in the bone marrow stroma, is the major non-protein glycosaminoglycan component of extracellular matrix (ECM) involved in cell positioning, proliferation, differentiation as well as in receptor-mediated changes in gene expression. Repair of bone and regeneration of bone marrow is dependent on ECM, inflammatory factors, like chemokines and degradative factors, like metalloproteinases. We analyzed the interaction between human mesenchymal stem cells (h-MSCs) and a three-dimensional (3-D) HA-based scaffold in vitro. The expression of CXC chemokines/receptors, CXCL8 (IL-8)/CXCR1-2, CXCL10 (IP-10)/CXCR3, CXCL12 (SDF-1)/CXCR4, and CXCL13 (BCA-1)/CXCR5, and metalloproteinases/inhibitors MMP-1, MMP-3, MMP-13/TIMP-1 were evaluated in h-MSCs grown on plastic or on HA-based scaffold by Real-time PCR, ELISA, and immunocytochemical techniques. Moreover, the expression of two HA receptors, CD44 and CD54, was analyzed. We found both at mRNA and protein levels that HA-based scaffold induced the expression of CXCR4, CXCL13, and MMP-3 and downmodulated the expression of CXCL12, CXCR5, MMP-13, and TIMP-1 while HA-based scaffold induced CD54 expression but not CD44. We found that these two HA receptors were directly involved in the modulation of CXCL12, CXCL13, and CXCR5. This study demonstrates a direct action of a 3-D HA-based scaffold, widely used for cartilage and bone repair, in modulating both h-MSCs inflammatory and degradative factors directly involved in the engraftment of specific cell types in a damaged area. Our data clearly demonstrate that HA in this 3-D conformation acts as a signaling molecule for h-MSCs.


Assuntos
Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/farmacologia , Mediadores da Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Anticorpos Monoclonais/farmacologia , Células Cultivadas , Quimiocina CXCL12 , Quimiocina CXCL13 , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Colagenases/genética , Colagenases/metabolismo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Humanos , Receptores de Hialuronatos/imunologia , Imuno-Histoquímica , Molécula 1 de Adesão Intercelular/imunologia , Interleucina-1/farmacologia , Metaloproteinase 13 da Matriz , Metaloproteinase 3 da Matriz/genética , Metaloproteinase 3 da Matriz/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Receptores CXCR5 , Receptores de Quimiocinas , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Engenharia Tecidual/métodos , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA