Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Small ; 10(21): 4340-51, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-24990430

RESUMO

Mesenchymal stromal cells (MSCs) are promising candidates in regenerative cell-therapies. However, optimizing their number and route of delivery remains a critical issue, which can be addressed by monitoring the MSCs' bio-distribution in vivo using super-paramagnetic iron-oxide nanoparticles (SPIONs). In this study, amino-polyvinyl alcohol coated (A-PVA) SPIONs are introduced for cell-labeling and visualization by magnetic resonance imaging (MRI) of human MSCs. Size and surface charge of A-PVA-SPIONs differ depending on their solvent. Under MSC-labeling conditions, A-PVA-SPIONs have a hydrodynamic diameter of 42 ± 2 nm and a negative Zeta potential of 25 ± 5 mV, which enable efficient internalization by MSCs without the need to use transfection agents. Transmission X-ray microscopy localizes A-PVA-SPIONs in intracellular vesicles and as cytosolic single particles. After identifying non-interfering cell-assays and determining the delivered and cellular dose, in addition to the administered dose, A-PVA-SPIONs are found to be non-toxic to MSCs and non-destructive towards their multi-lineage differentiation potential. Surprisingly, MSC migration is increased. In MRI, A-PVA-SPION-labeled MSCs are successfully visualized in vitro and in vivo. In conclusion, A-PVA-SPIONs have no unfavorable influences on MSCs, although it becomes evident how sensitive their functional behavior is towards SPION-labeling. And A-PVA-SPIONs allow MSC-monitoring in vivo.


Assuntos
Rastreamento de Células/métodos , Dextranos/química , Imageamento por Ressonância Magnética/métodos , Nanopartículas de Magnetita/química , Células-Tronco Mesenquimais/citologia , Álcool de Polivinil/química , Idoso , Animais , Diferenciação Celular , Rastreamento de Células/instrumentação , Células Cultivadas , Meios de Contraste/química , Dextranos/síntese química , Feminino , Humanos , Imageamento por Ressonância Magnética/instrumentação , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Pessoa de Meia-Idade , Ratos , Ratos Endogâmicos Lew
2.
J Transl Med ; 11: 236, 2013 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-24074138

RESUMO

BACKGROUND: Clinical cardiac cell therapy using autologous somatic stem cells is restricted by age and disease-associated impairment of stem cell function. Juvenile cells possibly represent a more potent alternative, but the impact of patient-related variables on such cell products is unknown. We therefore evaluated the behavior of neonatal cord blood mesenchymal stem cells (CB-MSC) in the presence of serum from patients with advanced heart failure (HF). METHODS: Human serum was obtained from patients with severe HF (n = 21) and from healthy volunteers (n = 12). To confirm the systemic quality of HF in the sera, TNF-α and IL-6 were quantified. CB-MSC from healthy neonates were cultivated for up to 14 days in medium supplemented with 10% protein-normalized human HF or control serum or fetal calf serum (FCS). RESULTS: All HF sera contained increased cytokine concentrations (IL-6, TNF-α). When exposed to HF serum, CB-MSC maintained basic MSC properties as confirmed by immunophenotyping and differentiation assays, but clonogenic cells were reduced in number and gave rise to substantially smaller colonies in the CFU-F assay. Cell cycle analysis pointed towards G1 arrest. CB-MSC metabolic activity and proliferation were significantly impaired for up to 3 days as measured by MTS turnover, BrdU incorporation and DAPI + nuclei counting. On day 5, however, CB-MSC growth kinetics approached control serum levels, though protein expression of cell cycle inhibitors (p21, p27), and apoptosis marker Caspase 3 remained elevated. Signal transduction included the stress and cytokine-induced JNK and ERK1/2 MAP kinase pathways. CONCLUSIONS: Heart failure temporarily inhibits clonality and proliferation of "healthy" juvenile MSC in vitro. Further studies should address the in vivo and clinical relevance of this finding.


Assuntos
Insuficiência Cardíaca/patologia , Células-Tronco Mesenquimais/citologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose , Biomarcadores/sangue , Estudos de Casos e Controles , Ciclo Celular , Proliferação de Células , Células Clonais , Ensaio de Unidades Formadoras de Colônias , Citocinas/sangue , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sangue Fetal/citologia , Insuficiência Cardíaca/sangue , Humanos , Recém-Nascido , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Soro/metabolismo , Transdução de Sinais , Adulto Jovem
4.
Biomaterials ; 98: 31-40, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27179133

RESUMO

In hip arthroplasty the implants' articulating surfaces can be made of a cobalt-chromium-molybdenum (CoCrMo) alloy. The use of these metal-on-metal (MoM) pairings can lead to the release of wear products such as metallic particles and dissociated metal species, raising concerns regarding their safety amongst orthopedic surgeons and the public. MoM-wear particles are reported to be heterogeneous in their physicochemical properties, are capable of inducing adverse effects on a cellular level and are thought to be involved in relevant clinical problems like aseptic osteolysis. Yet, it remains elusive how MoM-wear affects bone forming cells and their progenitors: bone marrow residing mesenchymal stromal cells (MSCs). This study introduces an assessment of the in vivo exposure to particulate and dissociated Co and Cr and evaluates the effects of MoM-wear on MSCs. The exposure to MoM-wear products in vivo and in vitro leads to a decrease in MSCs' osteogenic matrix mineralization and alkaline phosphatase activity on a cellular and systemic level. In conclusion, MoM-wear products are released in the periprosthetic region and elevate bone marrow Co and Cr concentrations towards levels that impair osteogenic differentiation of MSCs. Therefore, the ongoing use of CoCrMo alloys for articulating surfaces in joint replacement implants needs critical reconsideration.


Assuntos
Prótese de Quadril/efeitos adversos , Células-Tronco Mesenquimais/metabolismo , Próteses Articulares Metal-Metal/efeitos adversos , Material Particulado/efeitos adversos , Idoso , Fosfatase Alcalina/sangue , Matriz Óssea/metabolismo , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/patologia , Pessoa de Meia-Idade , Minerais/metabolismo , Osteogênese
5.
Nanomedicine (Lond) ; 10(14): 2139-51, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26214354

RESUMO

AIMS: First, it will be investigated if amino-polyvinyl alcohol-coated superparamagnetic iron oxide nanoparticles (A-PVA-SPIONs) are suitable for MRI contrast enhancement in bone marrow. Second, the impact of A-PVA-SPION exposure in vivo on the viability and key functions of local bone marrow stromal cells (BMSCs) will be investigated. MATERIAL & METHODS: Animals were systemically injected with A-PVA-SPIONs, followed by a 7-day survival time. Accumulation of A-PVA-SPIONs was confirmed by MRI, histology and inductively coupled plasma optical emission spectrometry. BMSCs were isolated from bone marrow for in vitro assessment of their viability and regenerative key functions. RESULTS: In this study, A-PVA-SPIONs were found to accumulate in bone marrow and increase the BMSCs' metabolic activity and migration rate. CONCLUSION: A-PVA-SPIONs appear suitable for contrast enhancement in bone marrow while our data suggest an influence on the BMSCs biology that necessitates future research.


Assuntos
Medula Óssea/metabolismo , Compostos Férricos/química , Células-Tronco Mesenquimais/metabolismo , Nanopartículas/química , Nanopartículas/metabolismo , Álcool de Polivinil/química , Animais , Meios de Contraste/química , Meios de Contraste/metabolismo , Ratos
6.
PLoS One ; 9(9): e106462, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25187955

RESUMO

Among other stressors, age and mechanical constraints significantly influence regeneration cascades in bone healing. Here, our aim was to identify genes and, through their functional annotation, related biological processes that are influenced by an interaction between the effects of mechanical fixation stability and age. Therefore, at day three post-osteotomy, chip-based whole-genome gene expression analyses of fracture hematoma tissue were performed for four groups of Sprague-Dawley rats with a 1.5-mm osteotomy gap in the femora with varying age (12 vs. 52 weeks - biologically challenging) and external fixator stiffness (mechanically challenging). From 31099 analysed genes, 1103 genes were differentially expressed between the six possible combinations of the four groups and from those 144 genes were identified as statistically significantly influenced by the interaction between age and fixation stability. Functional annotation of these differentially expressed genes revealed an association with extracellular space, cell migration or vasculature development. The chip-based whole-genome gene expression data was validated by q-RT-PCR at days three and seven post-osteotomy for MMP-9 and MMP-13, members of the mechanosensitive matrix metalloproteinase family and key players in cell migration and angiogenesis. Furthermore, we observed an interaction of age and mechanical stimuli in vitro on cell migration of mesenchymal stromal cells. These cells are a subpopulation of the fracture hematoma and are known to be key players in bone regeneration. In summary, these data correspond to and might explain our previously described biomechanical healing outcome after six weeks in response to fixation stiffness variation. In conclusion, our data highlight the importance of analysing the influence of risk factors of fracture healing (e.g. advanced age, suboptimal fixator stability) in combination rather than alone.


Assuntos
Consolidação da Fratura/fisiologia , Fraturas Ósseas/genética , Hematoma/metabolismo , Fatores Etários , Animais , Feminino , Fraturas Ósseas/fisiopatologia , Hematoma/fisiopatologia , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Cell Mol Immunol ; 10(2): 151-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23396474

RESUMO

The initial inflammatory phase of bone fracture healing represents a critical step for the outcome of the healing process. However, both the mechanisms initiating this inflammatory phase and the function of immune cells present at the fracture site are poorly understood. In order to study the early events within a fracture hematoma, we established an in vitro fracture hematoma model: we cultured hematomas forming during an osteotomy (artificial bone fracture) of the femur during total hip arthroplasty (THA) in vitro under bioenergetically controlled conditions. This model allowed us to monitor immune cell populations, cell survival and cytokine expression during the early phase following a fracture. Moreover, this model enabled us to change the bioenergetical conditions in order to mimic the in vivo situation, which is assumed to be characterized by hypoxia and restricted amounts of nutrients. Using this model, we found that immune cells adapt to hypoxia via the expression of angiogenic factors, chemoattractants and pro-inflammatory molecules. In addition, combined restriction of oxygen and nutrient supply enhanced the selective survival of lymphocytes in comparison with that of myeloid derived cells (i.e., neutrophils). Of note, non-restricted bioenergetical conditions did not show any similar effects regarding cytokine expression and/or different survival rates of immune cell subsets. In conclusion, we found that the bioenergetical conditions are among the crucial factors inducing the initial inflammatory phase of fracture healing and are thus a critical step for influencing survival and function of immune cells in the early fracture hematoma.


Assuntos
Artroplastia de Quadril/métodos , Metabolismo Energético/imunologia , Fraturas Ósseas/imunologia , Fraturas Ósseas/patologia , Hematoma/imunologia , Hematoma/patologia , Idoso , Sobrevivência Celular/imunologia , Células Cultivadas , Quimiocina CCL2/metabolismo , Feminino , Fêmur/cirurgia , Fraturas Ósseas/metabolismo , Hematoma/metabolismo , Humanos , Interferon gama/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Masculino , Pessoa de Meia-Idade , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização/imunologia
8.
PLoS One ; 7(12): e52700, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23285157

RESUMO

Mesenchymal stromal cells (MSCs) are of high relevance for the regeneration of mesenchymal tissues such as bone and cartilage. The promising role of MSCs in cell-based therapies and tissue engineering appears to be limited due to a decline of their regenerative potential with increasing donor age, their limited availability in human tissues and the need of in vitro expansion prior to treatment. We therefore aimed to determine to which degree in vitro aging and chronological aging may be similar processes or if in vitro culture-related changes at the cellular and molecular level are at least altered as a function of donor age. For that purpose we established MSCs cultures from young (yMSCs) and aged (aMSCs) rats that were cultured for more than 100 passages. These long-term MSCs cultures were non-tumorigenic and exhibited similar surface marker patterns as primary MSCs of passage 2. During in vitro expansion, but not during chronological aging, MSCs progressively lose their progenitor characteristics, e.g., complete loss of osteogenic differentiation potential, diminished adipogenic differentiation, altered cell morphology and increased susceptibility towards senescence. Transcriptome analysis revealed that long-term in vitro MSCs cultivation leads to down-regulation of genes involved in cell differentiation, focal adhesion organization, cytoskeleton turnover and mitochondria function. Accordingly, functional analysis demonstrated altered mitochondrial morphology, decreased antioxidant capacities and elevated ROS levels in long-term cultivated yMSCs as well as aMSCs. Notably, only the MSC migration potential and their antioxidative capacity were altered by in vitro as well as chronological aging. Based on specific differences observed between the impact of chronological and in vitro MSC aging we conclude that both are distinct processes.


Assuntos
Senescência Celular , Citoesqueleto/metabolismo , Células-Tronco Mesenquimais/metabolismo , Mitocôndrias/metabolismo , Animais , Diferenciação Celular , Movimento Celular , Células Cultivadas , Senescência Celular/genética , Citoesqueleto/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Masculino , Células-Tronco Mesenquimais/citologia , Mitocôndrias/genética , Osteogênese , Ratos
9.
PLoS One ; 7(9): e46483, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23029528

RESUMO

BACKGROUND: Bone fracture initiates a series of cellular and molecular events including the expression of hypoxia-inducible factor (HIF)-1. HIF-1 is known to facilitate recruitment and differentiation of multipotent human mesenchymal stromal cells (hMSC). Therefore, we analyzed the impact of hypoxia and HIF-1 on the competitive differentiation potential of hMSCs towards adipogenic and osteogenic lineages. METHODOLOGY/PRINCIPAL FINDINGS: Bone marrow derived primary hMSCs cultured for 2 weeks either under normoxic (app. 18% O(2)) or hypoxic (less than 2% O(2)) conditions were analyzed for the expression of MSC surface markers and for expression of the genes HIF1A, VEGFA, LDHA, PGK1, and GLUT1. Using conditioned medium, adipogenic or osteogenic differentiation as verified by Oil-Red-O or von-Kossa staining was induced in hMSCs under either normoxic or hypoxic conditions. The expression of HIF1A and VEGFA was measured by qPCR. A knockdown of HIF-1α by lentiviral transduction was performed, and the ability of the transduced hMSCs to differentiate into adipogenic and osteogenic lineages was analyzed. Hypoxia induced HIF-1α and HIF-1 target gene expression, but did not alter MSC phenotype or surface marker expression. Hypoxia (i) suppressed adipogenesis and associated HIF1A and PPARG gene expression in hMSCs and (ii) enhanced osteogenesis and associated HIF1A and RUNX2 gene expression. shRNA-mediated knockdown of HIF-1α enhanced adipogenesis under both normoxia and hypoxia, and suppressed hypoxia-induced osteogenesis. CONCLUSIONS/SIGNIFICANCE: Hypoxia promotes osteogenesis but suppresses adipogenesis of human MSCs in a competitive and HIF-1-dependent manner. We therefore conclude that the effects of hypoxia are crucial for effective bone healing, which may potentially lead to the development of novel therapeutic approaches.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Células-Tronco Mesenquimais/fisiologia , Adipogenia , Antígenos CD/metabolismo , Hipóxia Celular , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteogênese , PPAR gama/genética , PPAR gama/metabolismo , Fenótipo , Interferência de RNA , Ativação Transcricional , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
J Biomed Mater Res A ; 95(4): 1114-24, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20878902

RESUMO

Bone defect treatments can be augmented by mesenchymal stem cell (MSC) based therapies. MSC interaction with the extracellular matrix (ECM) of the surrounding tissue regulates their functional behavior. Understanding of these specific regulatory mechanisms is essential for the therapeutic stimulation of MSC in vivo. However, these interactions are presently only partially understood. This study examined in parallel, for the first time, the effects on the functional behavior of MSCs of 13 ECM components from bone, cartilage and hematoma compared to a control protein, and hence draws conclusions for rational biomaterial design. ECM components specifically modulated MSC adhesion, migration, proliferation, and osteogenic differentiation, for example, fibronectin facilitated migration, adhesion, and proliferation, but not osteogenic differentiation, whereas fibrinogen enhanced adhesion and proliferation, but not migration. Subsequently, the integrin expression pattern of MSCs was determined and related to the cell behavior on specific ECM components. Finally, on this basis, peptide sequences are reported for the potential stimulation of MSC functions. Based on the results of this study, ECM component coatings could be designed to specifically guide cell functions.


Assuntos
Materiais Biomiméticos/farmacologia , Regeneração Óssea/efeitos dos fármacos , Matriz Extracelular/metabolismo , Teste de Materiais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Biomarcadores/metabolismo , Bovinos , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Feminino , Humanos , Integrinas/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Dados de Sequência Molecular , Osteogênese/efeitos dos fármacos , Peptídeos/química , Peptídeos/metabolismo , Reprodutibilidade dos Testes
11.
Tissue Eng Part A ; 16(10): 3139-48, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20486790

RESUMO

Angiogenesis is essential to tissue reconstitution, is sensitive to mechanical stresses, and currently represents one of the major challenges in tissue engineering. The pro-angiogenic matrix metalloprotease-2 (MMP-2) is upregulated in mechanically loaded mesenchymal stem cells (MSCs). Therefore, MMP-2 may provide a regulating link between angiogenesis and the surrounding mechanical conditions. This study aimed to modulate MMP-2 levels by mechanical loading of MSCs embedded in a three-dimensional matrix as well as to investigate the mechanism of MMP-2 regulation along with its contribution to angiogenesis stimulation. MMP-2-inducing conditions (30% compression, 1 Hz, 72 h) were defined after varying loading parameters. Addition of the Golgi-disturbing agent Brefeldin A suppressed this mechanical upregulation of MMP-2. Analysis of enzymatic activities demonstrated an enhancement of pro-MMP-2, mature MMP-2, and tissue inhibitor of metalloproteases-2. Further, mechano-regulation of MMP-14 and mature MMP-2 was dependent upon the activity of furin, a proprotein processing endoprotease. Angiogenesis was stimulated by conditioned media from MSCs loaded at inducing conditions. This augmentation of angiogenesis was hindered by inhibition of pro-MMP-2 and mature MMP-2. In conclusion, mechanical stimulation of MSCs in a three-dimensional matrix induces pro-MMP-2 secretion and MMP-2 activation, potentially via the activation complex consisting of MMP-2/-14/tissue inhibitor of metalloproteases-2. Mechano-regulated pro-MMP-2 and mature MMP-2 seem to contribute to angiogenesis stimulation. Thus, an application of these loading parameters could augment vascularization of tissue-engineered constructs based on the described MMP-2-dependent mechanism.


Assuntos
Fenômenos Biomecânicos/fisiologia , Metaloproteinase 2 da Matriz/metabolismo , Células-Tronco Mesenquimais/citologia , Neovascularização Fisiológica/fisiologia , Reatores Biológicos , Proliferação de Células , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Masculino , Engenharia Tecidual , Alicerces Teciduais
12.
Bone ; 45(6): 1065-72, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19679210

RESUMO

INTRODUCTION: The clinically known importance of patient sex as a major risk factor for compromised bone healing is poorly reflected in animal models. Consequently, the underlying cellular mechanisms remain elusive. Because mesenchymal stem cells (MSCs) are postulated to regulate tissue regeneration and give rise to essential differentiated cell types, they may contribute to sex-specific differences in bone healing outcomes. METHODS: We investigated sex-specific variations in bone healing and associated differences in MSC populations. A 1.5 mm osteotomy gap in the femora of 8 male and 8 female 12-month-old Sprague-Dawley rats was stabilized by an external fixator. Healing was analyzed in terms of biomechanical testing, bridging and callus size over time (radiography at 2, 4, and 6 weeks after surgery), and callus volume and geometry by microCT at final follow-up. MSCs were obtained from bone marrow samples of an age-matched group of 12 animals (6 per gender) and analyzed for numbers of colony-forming units (CFUs) and their capacity to differentiate and proliferate. The proportion of senescent cells was determined by beta-galactosidase staining. RESULTS: Sex-specific differences were indicated by a compromised mechanical competence of the callus in females compared with males (maximum torque at failure, p=0.028). Throughout the follow-up, the cross-sectional area of callus relative to bone was reduced in females (p< or =0.01), and the bridging of callus was delayed (p(2weeks)=0.041). microCT revealed a reduced callus size (p=0.003), mineralization (p=0.003) and polar moment of inertia (p=0.003) in female animals. The female bone marrow contained significantly fewer MSCs, represented by low CFU numbers in both femora and tibiae (p(femur)=0.017, p(tibia)=0.010). Functional characteristics of male and female MSCs were similar. CONCLUSION: Biomechanically compromised and radiographically delayed bone formation were distinctive in female rats. These differences were concomitant with a reduced number of MSCs, which may be causative for the suboptimal bone healing.


Assuntos
Osso e Ossos/patologia , Células-Tronco Mesenquimais/citologia , Caracteres Sexuais , Cicatrização , Animais , Fenômenos Biomecânicos , Osso e Ossos/diagnóstico por imagem , Calo Ósseo/diagnóstico por imagem , Calo Ósseo/patologia , Contagem de Células , Ensaio de Unidades Formadoras de Colônias , Feminino , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Microtomografia por Raio-X
13.
Stem Cells ; 25(8): 1985-94, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17495113

RESUMO

Progenitor cells are involved in the regeneration of the musculoskeletal system, which is known to be influenced by mechanical boundary conditions. Furthermore, matrix metalloproteases (MMPs) and tissue-specific inhibitors of metalloproteases (TIMPs) are crucial for matrix remodelling processes that occur during regeneration of bone and other tissues. This study has therefore investigated whether MMP activity affects mesenchymal stem cell (MSC) behavior and how MMP activity is influenced by the mechanical stimulation of these cells. Broad spectrum inhibition of MMPs altered the migration, proliferation, and osteogenic differentiation of MSCs. Expression analysis detected MMP-2, -3, -10, -11, -13, and -14, as well as TIMP-2, in MSCs at the mRNA and protein levels. Mechanical stimulation of MSCs led to an upregulation of their extracellular gelatinolytic activity, which was consistent with the increased protein levels seen for MMP-2, -3, -13, and TIMP-2. However, mRNA expression levels of MMPs/TIMPs showed no changes in response to mechanical stimulation, indicating an involvement of post-transcriptional regulatory processes such as alterations in MMP secretion or activation. One potential regulatory molecule might be the furin protease. Specific inhibition of MMP-2, -3, and -13 showed MMP-13 to be involved in osteogenic differentiation. The results of this study suggest that MSC function is controlled by MMP activity, which in turn is regulated by mechanical stimulation of cells. Thus, MMP/TIMP balance seems to play an essential role in transferring mechanical signals into MSC function. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Metaloproteinases da Matriz/fisiologia , Mecanotransdução Celular , Células-Tronco Mesenquimais/fisiologia , Fenômenos Fisiológicos Musculoesqueléticos , Regeneração , Idoso , Idoso de 80 Anos ou mais , Células Cultivadas , Dipeptídeos/farmacologia , Furina/fisiologia , Regulação da Expressão Gênica , Humanos , Metaloproteinase 13 da Matriz/fisiologia , Metaloproteinase 2 da Matriz/genética , Inibidores de Metaloproteinases de Matriz , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Mecanotransdução Celular/genética , Pessoa de Meia-Idade , Modelos Biológicos , Fenômenos Fisiológicos Musculoesqueléticos/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Regeneração/efeitos dos fármacos , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Inibidores Teciduais de Metaloproteinases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA