Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Physiol ; 14: 1233920, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37916223

RESUMEN

Bones constantly change and adapt to physical stress throughout a person's life. Mechanical signals are important regulators of bone remodeling and repair by activating skeletal stem and progenitor cells (SSPCs) to proliferate and differentiate into bone-forming osteoblasts using molecular signaling mechanisms not yet fully understood. SSPCs reside in a dynamic specialized microenvironment called the niche, where external signals integrate to influence cell maintenance, behavior and fate determination. The nature of the niche in bone, including its cellular and extracellular makeup and regulatory molecular signals, is not completely understood. The mechanisms by which the niche, with all of its components and complexity, is modulated by mechanical signals during homeostasis and repair are virtually unknown. This review summarizes the current view of the cells and signals involved in mechanical adaptation of bone during homeostasis and repair, with an emphasis on identifying novel targets for the prevention and treatment of age-related bone loss and hard-to-heal fractures.

2.
Bone Res ; 11(1): 50, 2023 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-37752132

RESUMEN

Skeletal stem and progenitor cells (SSPCs) perform bone maintenance and repair. With age, they produce fewer osteoblasts and more adipocytes leading to a loss of skeletal integrity. The molecular mechanisms that underlie this detrimental transformation are largely unknown. Single-cell RNA sequencing revealed that Notch signaling becomes elevated in SSPCs during aging. To examine the role of increased Notch activity, we deleted Nicastrin, an essential Notch pathway component, in SSPCs in vivo. Middle-aged conditional knockout mice displayed elevated SSPC osteo-lineage gene expression, increased trabecular bone mass, reduced bone marrow adiposity, and enhanced bone repair. Thus, Notch regulates SSPC cell fate decisions, and moderating Notch signaling ameliorates the skeletal aging phenotype, increasing bone mass even beyond that of young mice. Finally, we identified the transcription factor Ebf3 as a downstream mediator of Notch signaling in SSPCs that is dysregulated with aging, highlighting it as a promising therapeutic target to rejuvenate the aged skeleton.


Asunto(s)
Adipocitos , Osteogénesis , Animales , Ratones , Osteogénesis/genética , Adiposidad , Envejecimiento/genética , Artrodesis , Ratones Noqueados , Agitación Psicomotora
3.
Exp Biol Med (Maywood) ; 247(21): 1885-1897, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35666091

RESUMEN

Delivering the parathyroid hormone (PTH) gene has been attempted preclinically in a handful of studies, but delivering full-length PTH (1-84) using adeno-associated viral (AAV) vectors has not. Given the difficulty in achieving therapeutic levels of secreted proteins using gene therapy, this study seeks to determine the feasibility of doing so with PTH. An AAV vector was used to deliver human PTH driven by a strong promoter. We demonstrate the ability to secrete full-length PTH from various cell types in vitro. PTH secretion from hepatocytes was measured over time and a fluorescent marker was used to compare the secretion rate of PTH in various cell types. Potency was measured by the ability of PTH to act on the PTH receptors of osteosarcoma cells and induced proliferation. PTH showed potency in vitro by inducing proliferation in two osteosarcoma cell lines. In vivo, AAV was administered systemically in immunocompromised mice which received xenografts of osteosarcoma cells. Animals that received the highest dose of AAV-PTH had higher liver and plasma concentrations of PTH. All dosing groups achieved measurable plasma concentrations of human PTH that were above the normal range. The high-dose group also had significantly larger tumors compared to control groups on the final day of the study. The tumors also showed dose-dependent differences in morphology. When looking at endocrine signaling and endogenous bone turnover, we observed a significant difference in tibial growth plate width in animals that received the high-dose AAV as well as dose-dependent changes in blood biomarkers related to PTH. This proof-of-concept study shows promise for further exploration of an AAV gene therapy to deliver full-length PTH for hypoparathyroidism. Additional investigation will determine efficacy in a disease model, but data shown establish bioactivity in well-established models of osteosarcoma.


Asunto(s)
Hormona Paratiroidea , Humanos , Animales , Ratones , Hormona Paratiroidea/genética
4.
Bone ; 154: 116223, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34624558

RESUMEN

Fracture healing is a complex, multistep process that is highly sensitive to mechanical signaling. To optimize repair, surgeons prescribe immediate weight-bearing as-tolerated within 24 hours after surgical fixation; however, this recommendation is based on anecdotal evidence and assessment of bulk healing outcomes (e.g., callus size, bone volume, etc.). Given challenges in accurately characterizing the mechanical environment and the ever-changing properties of the regenerate, the principles governing mechanical regulation of repair, including their cell and molecular basis, are not yet well defined. However, the use of mechanobiological rodent models, and their relatively large genetic toolbox, combined with recent advances in imaging approaches and single-cell analyses is improving our understanding of the bone microenvironment in response to loading. This review describes the identification and characterization of distinct cell populations involved in bone healing and highlights the most recent findings on mechanical regulation of bone homeostasis and repair with an emphasis on osteo-angio coupling. A discussion on aging and its impact on bone mechanoresponsiveness emphasizes the need for novel mechanotherapeutics that can re-sensitize skeletal stem and progenitor cells to physical rehabilitation protocols.


Asunto(s)
Callo Óseo , Curación de Fractura , Curación de Fractura/fisiología , Mecanotransducción Celular , Células Madre , Soporte de Peso
5.
JBMR Plus ; 3(9): e10199, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31667455

RESUMEN

Aging is associated with significant bone loss and increased fracture risk, which has been attributed to a diminished response to anabolic mechanical loading. In adults, skeletal progenitors proliferate and differentiate into bone-forming osteoblasts in response to increasing mechanical stimuli, though the effects of aging on this response are not well-understood. Here we show that both adult and aged mice exhibit load-induced periosteal bone formation, though the response is significantly attenuated with age. We also show that the acute response of adult bone to loading involves expansion of Sca-1+Prrx1+ and Sca-1-Prrx1+ cells in the periosteum. On the endosteal surface, loading enhances proliferation of both these cell populations, though the response is delayed by 2 days relative to the periosteal surface. In contrast to the periosteum and endosteum, the marrow does not exhibit increased proliferation of Sca-1+Prrx1+ cells, but only of Sca-1-Prrx1+ cells, underscoring fundamental differences in how the stem cell niche in distinct bone envelopes respond to mechanical stimuli. Notably, the proliferative response to loading is absent in aged bone even though there are similar baseline numbers of Prrx1 + cells in the periosteum and endosteum, suggesting that the proliferative capacity of progenitors is attenuated with age, and proliferation of the Sca-1+Prrx1+ population is critical for load-induced periosteal bone formation. These findings provide a basis for the development of novel therapeutics targeting these cell populations to enhance osteogenesis for overcoming age-related bone loss. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.

6.
J Bone Miner Res ; 34(5): 896-910, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30645780

RESUMEN

Elucidating the effects of mechanical stimulation on bone repair is crucial for optimization of the healing process. Specifically, the regulatory role that mechanical loading exerts on the osteogenic stem cell pool and vascular morphology during healing is incompletely understood. Because dynamic loading has been shown to enhance osteogenesis and repair, we hypothesized that loading induces the expansion of the osteoprogenitor cell population within a healing bone defect, leading to an increased presence of osteogenic cells. We further hypothesized that loading during the repair process regulates vascular and collagen matrix morphology and spatial interactions between vessels and osteogenic cells. To address these hypotheses, we used a mechanobiological bone repair model, which produces a consistent and reproducible intramembranous repair response confined in time and space. Bilateral tibial defects were created in adult C57BL/6 mice, which were subjected to axial compressive dynamic loading either during the early cellular invasion phase on postsurgical days (PSDs) 2 to 5 or during the matrix deposition phase on PSD 5 to 8. Confocal and two-photon microscopy was used to generate high-resolution three-dimensional (3D) renderings of longitudinal thick sections of the defect on PSD 10. Endomucin (EMCN)-positive vessels, Paired related homeobox 1 (Prrx1+) stem cell antigen-1 positive (Sca-1+) primitive osteoprogenitors (OPCs), and osterix positive (Osx+) preosteoblasts were visualized and quantified using deep tissue immunohistochemistry. New bone matrix was visualized with second harmonic generation autofluorescence of collagen fibers. We found that mechanical loading during the matrix deposition phase (PSD 5 to 8) increased vessel volume and number, and aligned vessels and collagen fibers to the load-bearing direction of bone. Furthermore, loading led to a significant increase in the proliferation and number of Prrx1+ Sca-1+ primitive OPCs, but not Osx+ preosteoblasts within the defect. Together, these data illustrate the adaptation of both collagen matrix and vascular morphology to better withstand mechanical load during bone repair, and that the mechanoresponsive cell population consists of the primitive osteogenic progenitors. © 2019 American Society for Bone and Mineral Research.


Asunto(s)
Matriz Ósea/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Células Madre/metabolismo , Tibia/metabolismo , Animales , Matriz Ósea/patología , Femenino , Proteínas de Homeodominio/metabolismo , Ratones , Osteoblastos/patología , Sialoglicoproteínas/metabolismo , Factor de Transcripción Sp7/metabolismo , Células Madre/patología , Tibia/patología , Soporte de Peso
8.
Bone ; 108: 145-155, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29305998

RESUMEN

Mechanical loading is an important aspect of post-surgical fracture care. The timing of load application relative to the injury event may differentially regulate repair depending on the stage of healing. Here, we used a novel mechanobiological model of cortical defect repair that offers several advantages including its technical simplicity and spatially confined repair program, making effects of both physical and biological interventions more easily assessed. Using this model, we showed that daily loading (5N peak load, 2Hz, 60 cycles, 4 consecutive days) during hematoma consolidation and inflammation disrupted the injury site and activated cartilage formation on the periosteal surface adjacent to the defect. We also showed that daily loading during the matrix deposition phase enhanced both bone and cartilage formation at the defect site, while loading during the remodeling phase resulted in an enlarged woven bone regenerate. All loading regimens resulted in abundant cellular proliferation throughout the regenerate and fibrous tissue formation directly above the defect demonstrating that all phases of cortical defect healing are sensitive to physical stimulation. Stress was concentrated at the edges of the defect during exogenous loading, and finite element (FE)-modeled longitudinal strain (εzz) values along the anterior and posterior borders of the defect (~2200µÎµ) was an order of magnitude larger than strain values on the proximal and distal borders (~50-100µÎµ). It is concluded that loading during the early stages of repair may impede stabilization of the injury site important for early bone matrix deposition, whereas loading while matrix deposition and remodeling are ongoing may enhance stabilization through the formation of additional cartilage and bone.


Asunto(s)
Hueso Cortical/patología , Hueso Cortical/fisiopatología , Modelos Biológicos , Cicatrización de Heridas , Animales , Fenómenos Biomecánicos , Matriz Ósea/metabolismo , Proliferación Celular , Condrogénesis , Fuerza Compresiva , Femenino , Hematoma/patología , Inflamación/patología , Ratones Endogámicos C57BL , Tamaño de los Órganos , Osteoclastos/metabolismo , Reproducibilidad de los Resultados , Estrés Mecánico , Tibia/patología , Soporte de Peso/fisiología
9.
ACS Appl Mater Interfaces ; 9(48): 41794-41806, 2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-29116745

RESUMEN

Mesenchymal stem cell (MSC) differentiation can be manipulated by nanotopographic interface providing a unique strategy to engineering stem cell therapy and circumventing complex cellular reprogramming. However, our understanding of the nanotopographic-mechanosensitive properties of MSCs and the underlying biophysical linkage of the nanotopography-engineered stem cell to directed commitment remains elusive. Here, we show that osteogenic differentiation of human MSCs (hMSCs) can be largely promoted using our nanoengineered topographic glass substrates in the absence of dexamethasone, a key exogenous factor for osteogenesis induction. We demonstrate that hMSCs sense and respond to surface nanotopography, through modulation of adhesion, cytoskeleton tension, and nuclear activation of TAZ (transcriptional coactivator with PDZ-binding motif), a transcriptional modulator of hMSCs. Our findings demonstrate the potential of nanotopographic surfaces as noninvasive tools to advance cell-based therapies for bone engineering and highlight the origin of biophysical response of hMSC to nanotopography.


Asunto(s)
Células Madre Mesenquimatosas , Diferenciación Celular , Células Cultivadas , Humanos , Nanoestructuras , Osteogénesis , Transducción de Señal , Factores de Transcripción
10.
J Bone Miner Res ; 32(4): 821-833, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27869327

RESUMEN

Chronic use of selective serotonin reuptake inhibitors (SSRIs) for the treatment of depression has been linked to osteoporosis. In this study, we investigated the effect of chronic SSRI use on fracture healing in two murine models of bone regeneration. First, we performed a comprehensive analysis of endochondral bone healing in a femur fracture model. C57/BL6 mice treated with fluoxetine, the most commonly prescribed SSRI, developed a normal cartilaginous soft-callus at 14 days after fracture and demonstrated a significantly smaller and biomechanically weaker bony hard-callus at 28 days. In order to further dissect the mechanism that resulted in a smaller bony regenerate, we used an intramembranous model of bone healing and revealed that fluoxetine treatment resulted in a significantly smaller bony callus at 7 and 14 days postinjury. In order to test whether the smaller bony regenerate following fluoxetine treatment was caused by an inhibition of osteogenic differentiation and/or mineralization, we employed in vitro experiments, which established that fluoxetine treatment decreases osteogenic differentiation and mineralization and that this effect is serotonin-independent. Finally, in a translational approach, we tested whether cessation of the medication would result in restoration of the regenerative potential. However, histologic and µCT analysis revealed non-union formation in these animals with fibrous tissue interposition within the callus. In conclusion, fluoxetine exerts a direct, inhibitory effect on osteoblast differentiation and mineralization, shown in two disparate murine models of bone repair. Discontinuation of the drug did not result in restoration of the healing potential, but rather led to complete arrest of the repair process. Besides the well-established effect of SSRIs on bone homeostasis, our study provides strong evidence that fluoxetine use negatively impacts fracture healing. © 2017 American Society for Bone and Mineral Research.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Fracturas del Fémur , Curación de Fractura/efectos de los fármacos , Osteoblastos , Inhibidores Selectivos de la Recaptación de Serotonina/efectos adversos , Animales , Modelos Animales de Enfermedad , Fracturas del Fémur/metabolismo , Fracturas del Fémur/patología , Fluoxetina/farmacología , Masculino , Ratones , Osteoblastos/metabolismo , Osteoblastos/patología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
11.
Integr Biol (Camb) ; 8(7): 785-94, 2016 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-27332785

RESUMEN

Angiogenesis is a process by which new blood vessels emerge from existing vessels through endothelial cell sprouting, migration, proliferation, and tubule formation. Angiogenesis during skeletal growth, homeostasis and repair is a complex and incompletely understood process. As the skeleton adapts to mechanical loading, we hypothesized that mechanical stimulation regulates "osteo-angio" crosstalk in the context of angiogenesis. We showed that conditioned media (CM) from osteoblasts exposed to fluid shear stress enhanced endothelial cell proliferation and migration, but not tubule formation, relative to CM from static cultures. Endothelial cell sprouting was studied using a dual-channel collagen gel-based microfluidic device that mimics vessel geometry. Static CM enhanced endothelial cell sprouting frequency, whereas loaded CM significantly enhanced both frequency and length. Both sprouting frequency and length were significantly enhanced in response to factors released from osteoblasts exposed to fluid shear stress in an adjacent channel. Osteoblasts released angiogenic factors, of which osteopontin, PDGF-AA, IGBP-2, MCP-1, and Pentraxin-3 were upregulated in response to mechanical loading. These data suggest that in vivo mechanical forces regulate angiogenesis in bone by modulating "osteo-angio" crosstalk through release of paracrine factors, which we term "osteokines".


Asunto(s)
Proteínas Angiogénicas/metabolismo , Mecanotransducción Celular/fisiología , Neovascularización Fisiológica/fisiología , Osteoblastos/metabolismo , Comunicación Paracrina/fisiología , Células 3T3 , Animales , Ratones , Resistencia al Corte/fisiología , Estrés Mecánico
12.
Curr Rheumatol Rep ; 17(9): 58, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26233599

RESUMEN

Mechanical loading is a potent anabolic regulator of bone mass, and the first line of defense for bone loss is weight-bearing exercise. Likewise, protected weight bearing is the first prescribed physical therapy following orthopedic reconstructive surgery. In both cases, enhancement of new bone formation is the goal. Our understanding of the physical cues, mechanisms of force sensation, and the subsequent cellular response will help identify novel physical and therapeutic treatments for age- and disuse-related bone loss, delayed- and nonunion fractures, and significant bony defects. This review highlights important new insights into the principles and mechanisms governing mechanical adaptation of the skeleton during homeostasis and repair and ends with a summary of clinical implications stemming from our current understanding of how bone adapts to biophysical force.


Asunto(s)
Regeneración Ósea/fisiología , Remodelación Ósea/fisiología , Homeostasis/fisiología , Adaptación Fisiológica , Humanos , Mecanotransducción Celular/fisiología , Células Madre Mesenquimatosas/fisiología , Osteoblastos/fisiología , Osteocitos/fisiología , Estrés Mecánico , Soporte de Peso/fisiología
13.
Bone ; 81: 260-269, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26191778

RESUMEN

During spaceflight, astronauts will be exposed to a complex mixture of ionizing radiation that poses a risk to their health. Exposure of rodents to ionizing radiation on Earth causes bone loss and increases osteoclasts in cancellous tissue, but also may cause persistent damage to stem cells and osteoprogenitors. We hypothesized that ionizing radiation damages skeletal tissue despite a prolonged recovery period, and depletes the ability of cells in the osteoblast lineage to respond at a later time. The goal of the current study was to test if irradiation prevents bone accrual and bone formation induced by an anabolic mechanical stimulus. Tibial axial compression was used as an anabolic stimulus after irradiation with heavy ions. Mice (male, C57BL/6J, 16 weeks) were exposed to high atomic number, high energy (HZE) iron ions ((56)Fe, 2 Gy, 600 MeV/ion) (IR, n=5) or sham-irradiated (Sham, n=5). In vivo axial loading was initiated 5 months post-irradiation; right tibiae in anesthetized mice were subjected to an established protocol known to stimulate bone formation (cyclic 9N compressive pulse, 60 cycles/day, 3 day/wk for 4 weeks). In vivo data showed no difference due to irradiation in the apparent stiffness of the lower limb at the initiation of the axial loading regimen. Axial loading increased cancellous bone volume by microcomputed tomography and bone formation rate by histomorphometry in both sham and irradiated animals, with a main effect of axial loading determined by two-factor ANOVA with repeated measure. There were no effects of radiation in cancellous bone microarchitecture and indices of bone formation. At the tibia diaphysis, results also revealed a main effect of axial loading on structure. Furthermore, irradiation prevented axial loading-induced stimulation of bone formation rate at the periosteal surface of cortical tissue. In summary, axial loading stimulated the net accrual of cancellous and cortical mass and increased cancellous bone formation rate despite prior exposure to ionizing radiation, in this case, HZE particles. Our findings suggest that mechanical stimuli may prove an effective treatment to improve skeletal structure following exposure to ionizing radiation.


Asunto(s)
Metabolismo/fisiología , Metabolismo/efectos de la radiación , Osteogénesis/fisiología , Osteogénesis/efectos de la radiación , Radiación Ionizante , Soporte de Peso/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Tibia/metabolismo , Tibia/efectos de la radiación
15.
Genes Dev ; 29(8): 817-31, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25846796

RESUMEN

The bone microenvironment is composed of niches that house cells across variable oxygen tensions. However, the contribution of oxygen gradients in regulating bone and blood homeostasis remains unknown. Here, we generated mice with either single or combined genetic inactivation of the critical oxygen-sensing prolyl hydroxylase (PHD) enzymes (PHD1-3) in osteoprogenitors. Hypoxia-inducible factor (HIF) activation associated with Phd2 and Phd3 inactivation drove bone accumulation by modulating osteoblastic/osteoclastic cross-talk through the direct regulation of osteoprotegerin (OPG). In contrast, combined inactivation of Phd1, Phd2, and Phd3 resulted in extreme HIF signaling, leading to polycythemia and excessive bone accumulation by overstimulating angiogenic-osteogenic coupling. We also demonstrate that genetic ablation of Phd2 and Phd3 was sufficient to protect ovariectomized mice against bone loss without disrupting hematopoietic homeostasis. Importantly, we identify OPG as a HIF target gene capable of directing osteoblast-mediated osteoclastogenesis to regulate bone homeostasis. Here, we show that coordinated activation of specific PHD isoforms fine-tunes the osteoblastic response to hypoxia, thereby directing two important aspects of bone physiology: cross-talk between osteoblasts and osteoclasts and angiogenic-osteogenic coupling.


Asunto(s)
Huesos/enzimología , Homeostasis , Osteoprotegerina/metabolismo , Oxígeno/metabolismo , Prolil Hidroxilasas/genética , Prolil Hidroxilasas/metabolismo , Células 3T3 , Animales , Resorción Ósea/genética , Huesos/citología , Comunicación Celular , Hipoxia de la Célula/fisiología , Células Cultivadas , Activación Enzimática , Femenino , Silenciador del Gen , Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Transducción de Señal/genética , Células Madre/enzimología
16.
J Biomech ; 48(1): 53-8, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25443882

RESUMEN

Axial compression of mouse limbs is commonly used to induce bone formation in a controlled, non-invasive manner. Determination of peak strains caused by loading is central to interpreting results. Load-strain calibration is typically performed using uniaxial strain gauges attached to the diaphyseal, periosteal surface of a small number of sacrificed animals. Strain is measured as the limb is loaded to a range of physiological loads known to be anabolic to bone. The load-strain relationship determined by this subgroup is then extrapolated to a larger group of experimental mice. This method of strain calculation requires the challenging process of strain gauging very small bones which is subject to variability in placement of the strain gauge. We previously developed a method to estimate animal-specific periosteal strain during axial ulnar loading using an image-based computational approach that does not require strain gauges. The purpose of this study was to compare the relationship between load-induced bone formation rates and periosteal strain at ulnar midshaft using three different methods to estimate strain: (A) Nominal strain values based solely on load-strain calibration; (B) Strains calculated from load-strain calibration, but scaled for differences in mid-shaft cross-sectional geometry among animals; and (C) An alternative image-based computational method for calculating strains based on beam theory and animal-specific bone geometry. Our results show that the alternative method (C) provides comparable correlation between strain and bone formation rates in the mouse ulna relative to the strain gauge-dependent methods (A and B), while avoiding the need to use strain gauges.


Asunto(s)
Matemática/métodos , Osteogénesis/fisiología , Estrés Mecánico , Cúbito/fisiología , Adaptación Fisiológica/fisiología , Animales , Calibración , Femenino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Periostio/fisiología , Soporte de Peso/fisiología
17.
Am J Physiol Endocrinol Metab ; 306(8): E937-44, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24619882

RESUMEN

Mechanical loading is an important regulator in skeletal growth, maintenance, and aging. Estrogen receptors have a regulatory role in mechanically induced bone adaptation. Estrogen receptor-α (ERα) is known to enhance load-induced bone formation, whereas ERß negatively regulates this process. We hypothesized that ERß regulates mechanical signaling in osteoblasts. We tested this hypothesis by subjecting primary calvarial cells isolated from wild-type and ERß-knockout mice (BERKO) to oscillatory fluid flow in the absence or presence of estradiol (E2). We found that the known responses to fluid shear stress, i.e., phosphorylation of the mitogen-activated protein kinase ERK and upregulation of COX-2 expression, were inhibited in BERKO cells in the absence of E2. Flow-induced increase in prostaglandin E2 (PGE2) release was not altered in BERKO cells in the absence of E2, but was increased when E2 was present. Additionally, immunofluorescence analysis and estrogen response element luciferase assays revealed increased ERα expression and flow- and ligand-induced nuclear translocation as well as transcriptional activity in BERKO cells in both the presence and absence of E2. Taken together, these data suggest that ERß plays both ligand-dependent and ligand-independent roles in mechanical signaling in osteoblasts. Furthermore, our data suggest that one mechanism by which ERß regulates mechanotransduction in osteoblasts may result from its inhibitory effect on ERα expression and function. Targeting estrogen receptors (e.g., inhibiting ERß) may represent an effective approach for prevention and treatment of age-related bone loss.


Asunto(s)
Receptor beta de Estrógeno/fisiología , Mecanotransducción Celular/genética , Osteoblastos/metabolismo , Animales , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Estradiol/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mecanotransducción Celular/efectos de los fármacos , Ratones , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Cráneo/citología
18.
Plast Reconstr Surg ; 133(1): 79-89, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24374670

RESUMEN

BACKGROUND: Restoration of biomechanical strength following surgical reconstruction of tendon or ligament insertion tears is challenging because these injuries typically heal as fibrous scars. The authors hypothesize that injuries at the tendon-bone interface would benefit from reconstruction with decellularized composite tendon-bone grafts. METHODS: Tendon-bone grafts were harvested from Sprague-Dawley rats. Grafts subjected to decellularization were compared histologically and biomechanically with untreated grafts ex vivo and in a new in vivo model. Wistar rats underwent Sprague-Dawley allograft reconstruction using a pair-matched design. The rats were killed at 2 or 4 weeks. B-cell and macrophage infiltration was determined using immunohistochemistry, and explants were tested biomechanically. RESULTS: Decellularization resulted in a decrease in cells from 164 ± 61 (untreated graft) to 13 ± 7 cells per high-power field cells (p < 0.005) and a corresponding significant decrease in DNA content, and preserved scaffold architecture of the tendon-bone interface. Biomechanical comparison revealed no difference in failure load (p = 0.32), ultimate tensile stress (p = 0.76), or stiffness (p = 0.22) between decellularized grafts and untreated controls. Following in vivo reconstruction with tendon-bone interface grafts, decellularized grafts were stronger than untreated grafts at 2 weeks (p = 0.047) and at 4 weeks (p < 0.005). A persistent increase in B-cell and macrophage infiltration was observed in both the capsule surrounding the tendon-bone interface and the tendon substance in untreated controls. CONCLUSION: Decellularized tendon-bone grafts display better biomechanical properties at early healing time points and a decreased immune response compared with untreated grafts in vivo.


Asunto(s)
Tendón Calcáneo/trasplante , Trasplante Óseo/métodos , Traumatismos de los Tendones/cirugía , Andamios del Tejido , Alotrasplante Compuesto Vascularizado/métodos , Tendón Calcáneo/fisiología , Animales , Fenómenos Biomecánicos/fisiología , Calcáneo/cirugía , Modelos Animales de Enfermedad , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Procedimientos de Cirugía Plástica/métodos , Recuperación de la Función/fisiología , Traumatismos de los Tendones/fisiopatología , Ingeniería de Tejidos/métodos , Recolección de Tejidos y Órganos/métodos , Cicatrización de Heridas/fisiología
19.
J Orthop Res ; 31(11): 1828-38, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23881789

RESUMEN

Mechanical loading is a key anabolic regulator of bone mass. Stromal cell-derived factor-1 (SDF-1) is a stem cell homing factor that is important in hematopoiesis, angiogenesis, and fracture healing, though its involvement in skeletal mechanoadaptation is virtually unknown. The objective of this study was to characterize skeletal expression patterns of SDF-1 and CXCR4, the receptor for SDF-1, and to determine the role of SDF-1 signaling in load-induced periosteal bone formation. Sixteen-week-old C57BL/6 mice were treated with PBS or AMD3100, an antagonist against CXCR4, and exposed to in vivo ulnar loading (2.8 N peak-to-peak, 2 Hz, 120 cycles). SDF-1 was expressed in cortical and trabecular osteocytes and marrow cells, and CXCR4 was primarily expressed in marrow cells. SDF-1 and CXCR4 expression was enhanced in response to mechanical stimulation. The CXCR4 receptor antagonist AMD3100 significantly attenuated load-induced bone formation and led to smaller adaptive changes in cortical geometric properties as determined by histomorphometric analysis. Our data suggest that SDF-1/CXCR4 signaling plays a critical role in skeletal mechanoadaptation, and may represent a unique therapeutic target for prevention and treatment of age-related and disuse bone loss.


Asunto(s)
Quimiocina CXCL12/fisiología , Osteogénesis , Receptores CXCR4/fisiología , Animales , Bencilaminas , Diferenciación Celular , Quimiocina CXCL12/genética , Ciclamas , Compuestos Heterocíclicos/farmacología , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/análisis , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/genética , Transducción de Señal
20.
J Biomech ; 46(13): 2271-6, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23880439

RESUMEN

Large variations in axial ulnar load strain calibration results suggest that animal-specific calibrations may be necessary. However, the optimal set of geometric measures for performing an animal-specific calibration are not known, potentially as a result of confounding effects associated with experimentally introduced variation. The purpose of this study was to characterize the inherent variability of ulnar geometric measures known to influence periosteal midshaft strain during axial ulnar exogenous loading, and to further quantify the relationship between the variance of those geometric measures and periosteal strain during axial loading. Thirty-nine right mouse forelimbs were scanned with microCT. Seven geometric measures that influence periosteal strain resulting from a combined axial and bending loading were computed and used to estimate animal-specific strains on the periosteal midshaft. Animal specific strains were estimated using a theoretical model based on the generalized flexure formula. The predicted mean and standard deviation of the simulated midshaft strain gauge measurement resulting from the inter-animal geometric differences was -985 ± 148 µÎµ/N. The complete beam bending term associated with bending about the I(min) axis accounted for 89% of the variance and reduced the residual RMSE to 50.4 µÎµ. Eccentricity associated with the axial loading contributed a substantial portion of variation to the computed strain suggesting that calibration procedures to account for animal differences should incorporate that variable. The method developed in this study provides a relatively simple procedure for computing animal-specific strains using microCT scan data, without the need of a load/strain calibration study or computationally intensive finite element models.


Asunto(s)
Cúbito/fisiología , Animales , Calibración , Femenino , Miembro Anterior/diagnóstico por imagen , Miembro Anterior/fisiología , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Estrés Mecánico , Cúbito/diagnóstico por imagen , Microtomografía por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA