Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Cell Sci ; 131(21)2018 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-30301777

RESUMO

Mechanical loading is a potent stimulus of bone adaptation, requiring the replenishment of the osteoblast from a progenitor population. One such progenitor is the mesenchymal stem cell (MSC), which undergoes osteogenic differentiation in response to oscillatory fluid shear. Yet, the mechanism mediating stem cell mechanotransduction, and thus the potential to target this therapeutically, is poorly understood. In this study, we demonstrate that MSCs utilise cAMP as a second messenger in mechanotransduction, which is required for flow-mediated increases in osteogenic gene expression. Furthermore, we demonstrate that this mechanosignalling is dependent on the primary cilium and the ciliary localised adenylyl cyclase 6. Finally, we also demonstrate that this mechanotransduction mechanism can be targeted therapeutically to enhance cAMP signalling and early osteogenic signalling, mimicking the beneficial effect of physical loading. Our findings therefore demonstrate a novel mechanism of MSC mechanotransduction that can be targeted therapeutically, demonstrating a potential mechanotherapeutic for bone-loss diseases such as osteoporosis.This article has an associated First Person interview with the first author of the paper.


Assuntos
Adenilil Ciclases/metabolismo , Cílios/metabolismo , AMP Cíclico/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Cílios/enzimologia , Mecanotransdução Celular , Células-Tronco Mesenquimais/enzimologia , Camundongos , Transdução de Sinais
2.
FASEB J ; 33(3): 4178-4187, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30550359

RESUMO

Macroscale loading of bone generates a complex local mechanical microenvironment that drives osteogenesis and bone mechanoadaptation. One such mechanical stimulus generated is hydrostatic pressure (HP); however, the effect of HP on mesenchymal stem cells (MSCs) and the mechanotransduction mechanisms utilized by these cells to sense this stimulus are yet to be fully elucidated. In this study, we demonstrate that cyclic HP is a potent mediator of cytoskeletal reorganization and increases in osteogenic responses in MSCs. In particular, we demonstrate that the intermediate filament (IF) network undergoes breakdown and reorganization with centripetal translocation of IF bundles toward the perinuclear region. Furthermore, we show for the first time that this IF remodeling is required for loading-induced MSC osteogenesis, revealing a novel mechanism of MSC mechanotransduction. In addition, we demonstrate that chemical disruption of IFs with withaferin A induces a similar mechanism of IF breakdown and remodeling as well as a subsequent increase in osteogenic gene expression in MSCs, exhibiting a potential mechanotherapeutic effect to enhance MSC osteogenesis. This study therefore highlights a novel mechanotransduction mechanism of pressure-induced MSC osteogenesis involving the understudied cytoskeletal structure, the IF, and demonstrates a potential new therapy to enhance bone formation in bone-loss diseases such as osteoporosis.-Stavenschi, E., Hoey, D. A. Pressure-induced mesenchymal stem cell osteogenesis is dependent on intermediate filament remodeling.


Assuntos
Filamentos Intermediários/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Osteogênese/fisiologia , Animais , Osso e Ossos/metabolismo , Osso e Ossos/fisiologia , Linhagem Celular , Citoesqueleto/metabolismo , Citoesqueleto/fisiologia , Expressão Gênica/fisiologia , Mecanotransdução Celular/fisiologia , Camundongos
3.
Stem Cell Res Ther ; 9(1): 276, 2018 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-30359324

RESUMO

BACKGROUND: Physical loading is necessary to maintain bone tissue integrity. Loading-induced fluid shear is recognised as one of the most potent bone micromechanical cues and has been shown to direct stem cell osteogenesis. However, the effect of pressure transients, which drive fluid flow, on human bone marrow stem cell (hBMSC) osteogenesis is undetermined. Therefore, the objective of the study is to employ a systematic analysis of cyclic hydrostatic pressure (CHP) parameters predicted to occur in vivo on early hBMSC osteogenic responses and late-stage osteogenic lineage commitment. METHODS: hBMSC were exposed to CHP of 10 kPa, 100 kPa and 300 kPa magnitudes at frequencies of 0.5 Hz, 1 Hz and 2 Hz for 1 h, 2 h and 4 h of stimulation, and the effect on early osteogenic gene expression of COX2, RUNX2 and OPN was determined. Moreover, to decipher whether CHP can induce stem cell lineage commitment, hBMSCs were stimulated for 4 days for 2 h/day using 10 kPa, 100 kPa and 300 kPa pressures at 2 Hz frequency and cultured statically for an additional 1-2 weeks. Pressure-induced osteogenesis was quantified based on ATP release, collagen synthesis and mineral deposition. RESULTS: CHP elicited a positive, but variable, early osteogenic response in hBMSCs in a magnitude- and frequency-dependent manner, that is gene specific. COX2 expression elicited magnitude-dependent effects which were not present for RUNX2 or OPN mRNA expression. However, the most robust pro-osteogenic response was found at the highest magnitude (300 kPa) and frequency regimes (2 Hz). Interestingly, long-term mechanical stimulation utilising 2 Hz frequency elicited a magnitude-dependent release of ATP; however, all magnitudes promoted similar levels of collagen synthesis and significant mineral deposition, demonstrating that lineage commitment is magnitude independent. This therefore demonstrates that physiological levels of pressures, as low as 10 kPa, within the bone can drive hBMSC osteogenic lineage commitment. CONCLUSION: Overall, these findings demonstrate an important role for cyclic hydrostatic pressure in hBMSCs and bone mechanobiology, which should be considered when studying pressure-driven fluid shear effects in hBMSCs mechanobiology. Moreover, these findings may have clinical implication in terms of bioreactor-based bone tissue engineering strategies.


Assuntos
Células da Medula Óssea/metabolismo , Regulação da Expressão Gênica , Mecanotransdução Celular , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Osteogênese/genética , Trifosfato de Adenosina/biossíntese , Fenômenos Biomecânicos , Reatores Biológicos , Células da Medula Óssea/citologia , Diferenciação Celular , Linhagem da Célula/genética , Colágeno/genética , Colágeno/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Humanos , Pressão Hidrostática , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Osteopontina/genética , Osteopontina/metabolismo , Cultura Primária de Células
4.
Sci Rep ; 8(1): 3824, 2018 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-29491434

RESUMO

Skeletal homeostasis requires the continued replenishment of the bone forming osteoblast from a mesenchymal stem cell (MSC) population, a process that has been shown to be mechanically regulated. However, the mechanisms by which a biophysical stimulus can induce a change in biochemical signaling, mechanotransduction, is poorly understood. As a precursor to loading-induced bone formation, deciphering the molecular mechanisms of MSC osteogenesis is a critical step in developing novel anabolic therapies. Therefore, in this study we characterize the expression of the mechanosensitive calcium channel Transient Receptor Potential subfamily V member 4 (TRPV4) in MSCs and demonstrate that TRPV4 localizes to areas of high strain, specifically the primary cilium. We demonstrate that TRPV4 is required for MSC mechanotransduction, mediating oscillatory fluid shear induced calcium signaling and early osteogenic gene expression. Furthermore, we demonstrate that TRPV4 can be activated pharmacologically eliciting a response that mirrors that seen with mechanical stimulation. Lastly, we show that TRPV4 localization to the primary cilium is functionally significant, with MSCs with defective primary cilia exhibiting an inhibited osteogenic response to TRPV4 activation. Collectively, this data demonstrates a novel mechanism of stem cell mechanotransduction, which can be targeted therapeutically, and further highlights the critical role of the primary cilium in MSC biology.


Assuntos
Cílios/metabolismo , Mecanotransdução Celular , Células-Tronco Mesenquimais/citologia , Resistência ao Cisalhamento , Canais de Cátion TRPV/metabolismo , Animais , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Sinalização do Cálcio , Linhagem Celular , Membrana Celular/metabolismo , Citosol/metabolismo , Regulação da Expressão Gênica , Homeostase , Camundongos , Osteogênese
5.
J Biomech ; 55: 99-106, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28256244

RESUMO

A potent regulator of bone anabolism is physical loading. However, it is currently unclear whether physical stimuli such as fluid shear within the marrow cavity is sufficient to directly drive the osteogenic lineage commitment of resident mesenchymal stem cells (MSC). Therefore, the objective of the study is to employ a systematic analysis of oscillatory fluid flow (OFF) parameters predicted to occur in vivo on early MSC osteogenic responses and late stage lineage commitment. MSCs were exposed to OFF of 1Pa, 2Pa and 5Pa magnitudes at frequencies of 0.5Hz, 1Hz and 2Hz for 1h, 2h and 4h of stimulation. Our findings demonstrate that OFF elicits a positive osteogenic response in MSCs in a shear stress magnitude, frequency, and duration dependent manner that is gene specific. Based on the mRNA expression of osteogenic markers Cox2, Runx2 and Opn after short-term fluid flow stimulation, we identified that a regime of 2Pa shear magnitude and 2Hz frequency induces the most robust and reliable upregulation in osteogenic gene expression. Furthermore, long-term mechanical stimulation utilising this regime, elicits a significant increase in collagen and mineral deposition when compared to static control demonstrating that mechanical stimuli predicted within the marrow is sufficient to directly drive osteogenesis.


Assuntos
Linhagem da Célula , Hidrodinâmica , Células-Tronco Mesenquimais/citologia , Osteogênese , Resistência ao Cisalhamento , Estresse Mecânico , Animais , Biomarcadores/metabolismo , Fenômenos Biomecânicos , Diferenciação Celular/fisiologia , Linhagem Celular , Regulação da Expressão Gênica , Camundongos , Fatores de Tempo
6.
Mol Biol Cell ; 28(6): 736-745, 2017 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-28100636

RESUMO

Duplication of the centrosomes is a tightly regulated process. Abnormal centrosome numbers can impair cell division and cause changes in how cells migrate. Duplicated centrosomes are held together by a proteinaceous linker made up of rootletin filaments anchored to the centrioles by C-NAP1. This linker is removed in a NEK2A kinase-dependent manner as mitosis begins. To explore C-NAP1 activities in regulating centrosome activities, we used genome editing to ablate it. C-NAP1-null cells were viable and had an increased frequency of premature centriole separation, accompanied by reduced density of the centriolar satellites, with reexpression of C-NAP1 rescuing both phenotypes. We found that the primary cilium, a signaling structure that arises from the mother centriole docked to the cell membrane, was intact in the absence of C-NAP1, although components of the ciliary rootlet were aberrantly localized away from the base of the cilium. C-NAP1-deficient cells were capable of signaling through the cilium, as determined by gene expression analysis after fluid flow-induced shear stress and the relocalization of components of the Hedgehog pathway. Centrosome amplification induced by DNA damage or by PLK4 or CDK2 overexpression was markedly reduced in the absence of C-NAP1. We conclude that centriole splitting reduces the local density of key centriolar precursors to impede overduplication.


Assuntos
Autoantígenos/metabolismo , Autoantígenos/fisiologia , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Centríolos/metabolismo , Autoantígenos/genética , Ciclo Celular , Proteínas de Ciclo Celular/genética , Linhagem Celular , Centríolos/fisiologia , Centrossomo/metabolismo , Cílios/metabolismo , Humanos , Mitose , Transdução de Sinais , Fuso Acromático/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA