Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Bone Joint Res ; 10(9): 619-628, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34569806

RESUMO

AIMS: To investigate whether idiopathic osteonecrosis of the femoral head (ONFH) is related to impaired osteoblast activities. METHODS: We cultured osteoblasts isolated from trabecular bone explants taken from the femoral head and the intertrochanteric region of patients with idiopathic ONFH, or from the intertrochanteric region of patients with osteoarthritis (OA), and compared their viability, mineralization capacity, and secretion of paracrine factors. RESULTS: Osteoblasts from the intertrochanteric region of patients with ONFH showed lower alkaline phosphatase (ALP) activity and mineralization capacity than osteoblasts from the same skeletal site in age-matched patients with OA, as well as lower messenger RNA (mRNA) levels of genes encoding osteocalcin and bone sialoprotein and higher osteopontin expression. In addition, osteoblasts from patients with ONFH secreted lower osteoprotegerin (OPG) levels than those from patients with OA, resulting in a higher receptor activator of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) ligand (RANKL)-to-OPG ratio. In patients with ONFH, osteoblasts from the femoral head showed reduced viability and mineralized nodule formation compared with osteoblasts from the intertrochanteric region. Notably, the secretion of the pro-resorptive factors interleukin-6 and prostaglandin E2 as well as the RANKL-to-OPG ratio were markedly higher in osteoblast cultures from the femoral head than in those from the intertrochanteric region. CONCLUSION: Idiopathic ONFH is associated with a reduced mineralization capacity of osteoblasts and increased secretion of pro-resorptive factors. Cite this article: Bone Joint Res 2021;10(9):619-628.

2.
Stem Cell Res Ther ; 11(1): 57, 2020 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-32054534

RESUMO

BACKGROUND: The mechanisms by which macrophage phenotype contributes to mesenchymal stem cells (MSC)-mediated bone repair remain unclear. In this work, we investigated the influence of factors released by human macrophages polarized to a pro-inflammatory or an anti-inflammatory phenotype on the ability of human MSC to attach, migrate, and differentiate toward the osteoblastic lineage. We focused on the role of TNF-α and IL-10, key pro-inflammatory and anti-inflammatory cytokines, respectively, in regulating MSC functions. METHODS: MSC were treated with media conditioned by pro-inflammatory or anti-inflammatory macrophages to study their influence in cell attachment, migration, and osteogenic differentiation. The involvement of TNF-α and IL-10 in the regulation of MSC functions was investigated using neutralizing antibodies and recombinant cytokines. RESULTS: Treatment of MSC with media conditioned by pro-inflammatory or anti-inflammatory macrophages promoted cell elongation and enhanced MSC ability to attach and migrate. These effects were more noticeable when MSC were treated with media from pro-inflammatory macrophages. Interestingly, MSC osteogenic activity was enhanced by factors released by anti-inflammatory macrophages, but not by pro-inflammatory macrophages. Significant IL-10 levels originated from anti-inflammatory macrophages enhanced MSC osteogenesis by increasing ALP activity and mineralization in MSC layers cultured under osteogenic conditions. Moreover, macrophage-derived IL-10 regulated the expression of the osteogenic markers RUNX2, COL1A1, and ALPL. Notably, low TNF-α levels secreted by anti-inflammatory macrophages increased ALP activity in differentiating MSC whereas high TNF-α levels produced by pro-inflammatory macrophages had no effects on osteogenesis. Experiments in which MSC were treated with cytokines revealed that IL-10 was more effective in promoting matrix maturation and mineralization than TNF-α. CONCLUSIONS: Factors secreted by pro-inflammatory macrophages substantially increased MSC attachment and migration whereas those released by anti-inflammatory macrophages enhanced MSC osteogenic activity as well as cell migration. IL-10 was identified as an important cytokine secreted by anti-inflammatory macrophages that potentiates MSC osteogenesis. Our findings provide novel insights into how environments provided by macrophages regulate MSC osteogenesis, which may be helpful to develop strategies to enhance bone regeneration.


Assuntos
Expressão Gênica/genética , Inflamação/metabolismo , Macrófagos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteogênese/genética , Diferenciação Celular , Proliferação de Células , Humanos
3.
Stem Cell Res Ther ; 10(1): 58, 2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30760316

RESUMO

BACKGROUND: Immunoregulatory capacity of mesenchymal stem cells (MSC) is triggered by the inflammatory environment, which changes during tissue repair. Macrophages are essential in mediating the inflammatory response after injury and can adopt a range of functional phenotypes, exhibiting pro-inflammatory and anti-inflammatory activities. An accurate characterization of MSC activation by the inflammatory milieu is needed for improving the efficacy of regenerative therapies. In this work, we investigated the immunomodulatory functions of MSC primed with factors secreted from macrophages polarized toward a pro-inflammatory or an anti-inflammatory phenotype. We focused on the role of TNF-α and IL-10, prototypic pro-inflammatory and anti-inflammatory cytokines, respectively, as priming factors for MSC. METHODS: Secretion of immunoregulatory mediators from human MSC primed with media conditioned by human macrophages polarized toward a pro-inflammatory or an anti-inflammatory phenotype was determined. Immunomodulatory potential of primed MSC on polarized macrophages was studied using indirect co-cultures. Involvement of TNF-α and IL-10 in priming MSC and of PGE2 in MSC-mediated immunomodulation was investigated employing neutralizing antibodies. Collagen hydrogels were used to study MSC and macrophages interactions in a more physiological environment. RESULTS: Priming MSC with media conditioned by pro-inflammatory or anti-inflammatory macrophages enhanced their immunomodulatory potential through increased PGE2 secretion. We identified the pro-inflammatory cytokine TNF-α as a priming factor for MSC. Notably, the anti-inflammatory IL-10, mainly produced by pro-resolving macrophages, potentiated the priming effect of TNF-α. Collagen hydrogels acted as instructive microenvironments for MSC and macrophages functions and their crosstalk. Culturing macrophages on hydrogels stimulated anti-inflammatory versus pro-inflammatory cytokine secretion. Encapsulation of MSC within hydrogels increased PGE2 secretion and potentiated immunomodulation on macrophages, attenuating macrophage pro-inflammatory state and sustaining anti-inflammatory activation. Priming with inflammatory factors conferred to MSC loaded in hydrogels greater immunomodulatory potential, promoting anti-inflammatory activity of macrophages. CONCLUSIONS: Factors secreted by pro-inflammatory and anti-inflammatory macrophages activated the immunomodulatory potential of MSC. This was partially attributed to the priming effect of TNF-α and IL-10. Immunoregulatory functions of primed MSC were enhanced after encapsulation in hydrogels. These findings may provide insight into novel strategies to enhance MSC immunoregulatory potency.


Assuntos
Inflamação/genética , Interleucina-10/genética , Macrófagos/imunologia , Células-Tronco Mesenquimais/imunologia , Fator de Necrose Tumoral alfa/genética , Animais , Células Cultivadas , Técnicas de Cocultura , Citocinas/genética , Dinoprostona/genética , Humanos , Hidrogéis/farmacologia , Imunomodulação/genética , Inflamação/imunologia , Inflamação/patologia , Macrófagos/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Medicina Regenerativa
4.
Sci Rep ; 8(1): 4278, 2018 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-29511300

RESUMO

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

5.
Sci Rep ; 8(1): 364, 2018 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-29305583

RESUMO

A correction to this article has been published and is linked from the HTML version of this paper. The error has been fixed in the paper.

6.
Sci Rep ; 7(1): 14618, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-29097745

RESUMO

Mesenchymal stem cells (MSC) modulate the macrophage-mediated inflammatory response through the secretion of soluble factors. In addition to its classical effects on calcium homeostasis, 1,25-dihydroxyvitamin D3 (1,25D3) has emerged as an important regulator of the immune system. The present study investigates whether 1,25D3 modulates the paracrine interactions between MSC and macrophages. 1,25D3 stimulated MSC to produce PGE2 and VEGF and regulated the interplay between macrophages and MSC toward reduced pro-inflammatory cytokine production. Conditioned media (CM) from co-cultures of macrophages and MSC impaired MSC osteogenesis. However, MSC cultured in CM from 1,25D3-treated co-cultures showed increased matrix maturation and mineralization. Co-culturing MSC with macrophages prevented the 1,25D3-induced increase in RANKL levels, which correlated with up-regulation of OPG secretion. MSC seeding in three-dimensional (3D) substrates potentiated their immunomodulatory effects on macrophages. Exposure of 3D co-cultures to 1,25D3 further reduced the levels of soluble factors related to inflammation and chemotaxis. As a consequence of 1,25D3 treatment, the recruitment of monocytes toward CM of 3D co-cultures decreased, while the osteogenic maturation of MSC increased. These data add new insights into the pleiotropic effects of 1,25D3 on the crosstalk between MSC and macrophages and highlight the role of the hormone in bone regeneration.


Assuntos
Conservadores da Densidade Óssea/farmacologia , Calcitriol/farmacologia , Macrófagos/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Comunicação Parácrina/efeitos dos fármacos , Adolescente , Adulto , Prótese Vascular , Técnicas de Cocultura , Expressão Gênica , Humanos , Macrófagos/citologia , Macrófagos/fisiologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Osteogênese/efeitos dos fármacos , Osteogênese/fisiologia , Comunicação Parácrina/fisiologia , Ligante RANK/metabolismo , Células THP-1 , Alicerces Teciduais , Adulto Jovem
7.
Biomed Mater ; 11(5): 055011, 2016 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-27716631

RESUMO

We developed biodegradable polymeric coatings loaded with increasing amounts of dexamethasone on composites based on polylactic acid and Mg particles for bone repair. Incorporation of Mg particles into the polymeric matrix improves the compressive behaviour of the polymer. Mg-containing composites release Mg2+ ions into the culture medium and improve mesenchymal stem cell (MSC) viability, enhance their osteogenic potential and promote the release of angiogenic factors. Dexamethasone-loaded coatings deposited on composites delay Mg2+ ion dissolution while releasing controlled amounts of the drug, which are highly dependent on initial payload. Release kinetic of dexamethasone from the coatings exhibits a fast initial release of the drug followed by a slower secondary release. Bioactivity of the released dexamethasone was explored by monitoring dose-dependent responses of MSCs and macrophages. Biological effects exerted by the released drug are similar to those observed in cells treated with solutions of the glucocorticoid, indicating that the method employed for inclusion of dexamethasone into the coatings does not impair its bioactive behaviour. Culturing MSCs on dexamethasone-releasing coatings enhances extracellular matrix production and initial induction to osteogenic commitment as a function of drug payload. Dexamethasone incorporated into the coatings presents anti-inflammatory activity, as shown by the decrease in the production of cytokines and angiogenic factors by macrophages and MSCs. Deposition of dexamethasone-releasing coatings on polymer/Mg composites appears to be a promising approach to delay composite degradation at the early stage of implantation and may be useful to attenuate inflammation and adverse foreign body reactions.


Assuntos
Materiais Revestidos Biocompatíveis/química , Dexametasona/química , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Anti-Inflamatórios/química , Células da Medula Óssea/citologia , Sobrevivência Celular , Força Compressiva , Análise Custo-Benefício , Citocinas/metabolismo , Dexametasona/administração & dosagem , Reação a Corpo Estranho , Glucocorticoides/química , Humanos , Inflamação , Macrófagos/metabolismo , Magnésio/química , Microscopia Confocal , Neovascularização Patológica , Poliésteres/química , Ácido Poliglicólico/química , Polímeros/química , Estresse Mecânico
8.
J Biomed Mater Res A ; 104(4): 866-78, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26662548

RESUMO

In this work, we investigated a new approach to incorporate Mg particles within a PDLLA matrix using a solvent-free commercially available process. PDLLA/Mg composites were manufactured by injection moulding and the effects of Mg incorporated into PDLLA on MSC and macrophage responses were evaluated. Small amounts of Mg particles (≤ 1 wt %) do not cause thermal degradation of PDLLA, which retains its mechanical properties. PDLLA/Mg composites release hydrogen, alkaline products and Mg(2+) ions without changing pH of culture media. Mg-containing materials provide a noncytotoxic environment that enhances MSC viability. Concentration of Mg(2+) ions in extracts of MSCs increases with the increment of Mg content in the composites. Incorporation of Mg particles into PDLLA stimulates FN production, ALP activity, and VEGF secretion in MSCs, an effect mediated by degradation products dissolved from the composites. Degradation products of PDLLA induce an increase in MCP-1, RANTES, and MIP-1α secretion in macrophages while products of composites have minimal effect on these chemokines. Regulation of MSC behavior at the biomaterial's interface and macrophage-mediated inflammatory response to the degradation products is related to the incorporation of Mg in the composites. These findings suggest that including small amounts of Mg particles into polymeric devices can be a valuable strategy to promote osseointegration and reduce host inflammatory response.


Assuntos
Materiais Biocompatíveis/metabolismo , Macrófagos/citologia , Magnésio/metabolismo , Células-Tronco Mesenquimais/citologia , Poliésteres/metabolismo , Fosfatase Alcalina/metabolismo , Materiais Biocompatíveis/química , Linhagem Celular , Sobrevivência Celular , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CCL3/metabolismo , Quimiocina CCL5/metabolismo , Humanos , Macrófagos/metabolismo , Magnésio/química , Células-Tronco Mesenquimais/metabolismo , Poliésteres/química , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Biomaterials ; 37: 124-33, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25453943

RESUMO

Implantation of scaffolds may elicit a host foreign body response triggered by monocyte/macrophage lineage cells. Growing evidence suggests that topographical cues of scaffolds play an important role in MSC functionality. In this work, we examined whether surface topographical features can regulate paracrine interactions that MSCs establish with macrophages. Three-dimensional (3D) topography sensing drives MSCs into a spatial arrangement that stimulates the production of the anti-inflammatory proteins PGE2 and TSG-6. Compared to two-dimensional (2D) settings, 3D arrangement of MSCs co-cultured with macrophages leads to an important decrease in the secretion of soluble factors related with inflammation and chemotaxis including IL-6 and MCP-1. Attenuation of MCP-1 secretion in 3D co-cultures correlates with a decrease in the accumulation of its mRNA levels in MSCs and macrophages. Using neutralizing antibodies, we identified that the interplay between PGE2, IL-6, TSG-6 and MCP-1 in the co-cultures is strongly influenced by the micro-architecture that supports MSCs. Local inflammatory milieu provided by 3D-arranged MSCs in co-cultures induces a decrease in monocyte migration as compared to monolayer cells. This effect is partially mediated by reduced levels of IL-6 and MCP-1, proteins that up-regulate each other's secretion. Our findings highlight the importance of topographical cues in the soluble factor-guided communication between MSCs and macrophages.


Assuntos
Comunicação Celular , Macrófagos/citologia , Células-Tronco Mesenquimais/citologia , Anticorpos Bloqueadores/farmacologia , Comunicação Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Dinoprostona/metabolismo , Humanos , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Modelos Biológicos , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Tempo
10.
J Biomed Mater Res A ; 102(1): 128-40, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23613185

RESUMO

The interactions between bone tissue and orthopedic implants are strongly affected by mechanical forces at the bone-implant interface, but the interplay between surface topographies, mechanical stimuli, and cell behavior is complex and not well understood yet. This study reports on the influence of mechanical stretch on human mesenchymal stem cells (hMSCs) attached to metallic substrates with different roughness. Controlled forces were applied to plasma membrane of hMSCs cultured on smooth and rough stainless steel surfaces using magnetic collagen-coated particles and an electromagnet system. Degree of phosphorylation of focal adhesion kinase (p-FAK) on the active form (Tyr-397), prostaglandin E2 (PGE2) and vascular endothelial growth factor (VEGF) levels increased on rough samples under static conditions. Cell viability and fibronectin production decreased on rough substrates, while hMSCs maturated to the osteoblastic lineage to a similar extent on both surfaces. PGE2 production and osteoprotegerin/receptor activator of nuclear factor kappa-B ligand ratio increased after force application on both surfaces, although to a greater extent on smooth substrates. p-FAK on Tyr-397 was induced fairly rapidly by mechanical stimulation on rough surfaces while cells cultured on smooth samples failed to activate this kinase in response to tensile forces. Mechanical forces enhanced VEGF secretion and reduced cell viability, fibronetin levels and osteoblastic maturation on smooth surfaces but not on rough samples. The magnetite beads model used in this study is well suited to characterize the response of hMSCs cultured on metallic surfaces to tensile forces and collected data suggest a mechanism whereby mechanotransduction driven by FAK is essential for stem cell growth and functioning on metallic substrates.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Estresse Mecânico , Células Cultivadas , Quinase 1 de Adesão Focal/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Subunidade p50 de NF-kappa B/metabolismo , Osteoblastos/citologia , Fosforilação , Propriedades de Superfície , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Acta Biomater ; 7(12): 4210-21, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21827875

RESUMO

Osteosarcoma-derived cells have been routinely used for studying osteoblastic functions, but it remains unclear to what extent they mimic the behavior of primary osteoblasts in the study of cells and materials interactions. This study reports comparatively on the responses of three human osteosarcoma cell lines, MG-63, Saos-2 and U-2 OS, and human primary osteoblasts cultured on Ti6Al4V surfaces or exposed to Ti particles. Phenotypic characterization of the cell lines revealed that Saos-2 cells and primary osteoblasts displayed similar expression patterns of Cbfa1, SP7 and osteocalcin. Unlike primary cells, the cell lines expressed markers of undifferentiated cells, had high proliferative rates and poor fibronectin matrix assembly. None of the three cell lines faithfully reproduced the adhesive behavior of primary osteoblasts when cultured on Ti6Al4V surfaces or exposed to Ti particles. Differences in cell growth between the cell lines and primary osteoblasts cultured on Ti6Al4V surfaces were also observed. Ti particles inhibited the growth of Saos-2 cells and primary osteoblasts to a similar extent, while no such effect was observed in U-2 OS and MG-63 cells. Saos-2 cells reproduced the alkaline phosphatase (ALP) activity profile of primary osteoblasts cultured on metallic surfaces or exposed to particles. Altogether, these results show that none of the osteoblast-like cells studied perfectly mimic the behavior of human osteoblast cells (hOB) on Ti6Al4V surfaces or exposed to Ti particles. Saos-2 cells reproduce some of the hOB responses such as the profile of enzymatic ALP activity when cultured on the surfaces or treated with particles as well as cell growth inhibition when exposed to Ti particles. Although in vitro cytocompatibility studies involve the evaluation of multiple parameters, Saos-2 cells may be used as representative of human osteoblasts when these standard tests are evaluated.


Assuntos
Materiais Biocompatíveis , Modelos Teóricos , Osteoblastos/citologia , Ligas , Sequência de Bases , Diferenciação Celular , Linhagem Celular , Primers do DNA , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Titânio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...