Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur Radiol Exp ; 7(1): 38, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37532922

RESUMO

BACKGROUND: Heterotopic ossification (HO) is a frequent and debilitating complication of traumatic musculoskeletal injuries and orthopedic procedures. Prophylactic dosing of botulinum toxin type A (BTxA) holds potential as a novel treatment option if accurately distributed throughout soft-tissue volumes where protection is clinically desired. We developed a high-resolution, microcomputed tomography (microCT)-based imaging strategy to assess drug distribution and validated this platform by quantifying distribution achieved via a prototype delivery system versus a single-bolus injection. METHODS: We injected an iodine-containing contrast agent (iodixanol 320 mg I/mL) into dissected rabbit musculature followed by microCT imaging and analysis. To contrast the performance of distributed versus bolus injections, a three-dimensional (3D) 64-cm3-printed soft-tissue holder was developed. A centered 2-cm3 volume of interest (VOI) was targeted with a single-bolus injection or an equal volume distributed injection delivered via a 3D-printed prototype. VOI drug coverage was quantified as a percentage of the VOI volume that was < 1.0 mm from the injected fluid. RESULTS: The microCT-based approach enabled high-resolution quantification of injection distribution within soft tissue. The distributed dosing prototype provided significantly greater tissue coverage of the targeted VOI (72 ± 3%, mean ± standard deviation) when compared to an equal volume bolus dose (43 ± 5%, p = 0.031) while also enhancing the precision of injection targeting. CONCLUSIONS: A microCT-based imaging technique precisely quantifies drug distribution within a soft-tissue VOI, providing a path to overcome a barrier for clinical translation of prophylactic inhibition of HO by BTxA. RELEVANCE STATEMENT: This platform will facilitate rapid optimization of injection parameters for clinical devices used to effectively and safely inhibit the formation of heterotopic ossification. KEY POINTS: • MicroCT provides high-resolution quantification of soft-tissue drug distribution. • Distributed dosing is required to maximize soft-tissue drug coverage. • Imaging platform will enable rapid screening of 3D-printed drug distribution prototypes.


Assuntos
Iodo , Ossificação Heterotópica , Animais , Coelhos , Microtomografia por Raio-X/métodos , Sistemas de Liberação de Medicamentos
2.
Front Bioeng Biotechnol ; 11: 1206008, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37383524

RESUMO

Voluntary wheel running (VWR) is widely used to study how exercise impacts a variety of physiologies and pathologies in rodents. The primary activity readout of VWR is aggregated wheel turns over a given time interval (most often, days). Given the typical running frequency of mice (∼4 Hz) and the intermittency of voluntary running, aggregate wheel turn counts, therefore, provide minimal insight into the heterogeneity of voluntary activity. To overcome this limitation, we developed a six-layer convolutional neural network (CNN) to determine the hindlimb foot strike frequency of mice exposed to VWR. Aged female C57BL/6 mice (22 months, n = 6) were first exposed to wireless angled running wheels for 2 h/d, 5 days/wk for 3 weeks with all VWR activities recorded at 30 frames/s. To validate the CNN, we manually classified foot strikes within 4800 1-s videos (800 randomly chosen for each mouse) and converted those values to frequency. Upon iterative optimization of model architecture and training on a subset of classified videos (4400), the CNN model achieved an overall training set accuracy of 94%. Once trained, the CNN was validated on the remaining 400 videos (accuracy: 81%). We then applied transfer learning to the CNN to predict the foot strike frequency of young adult female C57BL6 mice (4 months, n = 6) whose activity and gait differed from old mice during VWR (accuracy: 68%). In summary, we have developed a novel quantitative tool that non-invasively characterizes VWR activity at a much greater resolution than was previously accessible. This enhanced resolution holds potential to overcome a primary barrier to relating intermittent and heterogeneous VWR activity to induced physiological responses.

3.
JBMR Plus ; 7(2): e10712, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36751418

RESUMO

The anabolic response of aged bone to skeletal loading is typically poor. Efforts to improve mechanotransduction in aged bone have met with limited success. This study investigated whether the bone response to direct skeletal loading is improved by reducing sympathetic suppression of osteoblastic bone formation via ß2AR. To test this possibility, we treated aged wild-type C57BL/6 mice with a selective ß2AR antagonist, butaxamine (Butax), before each of nine bouts of cantilever bending of the right tibia. Midshaft periosteal bone formation was assessed by dynamic histomorphometry of loaded and contralateral tibias. Butax treatment did not alter osteoblast activity of contralateral tibias. Loading alone induced a modest but significant osteogenic response. However, when loading was combined with Butax pretreatment, the anabolic response was significantly elevated compared with loading preceded by saline injection. Subsequent studies in osteoblastic cultures revealed complex negative interactions between adrenergic and mechanically induced intracellular signaling. Activation of ß2AR by treatment with the ß1, ß2-agonist isoproterenol (ISO) before fluid flow exposure diminished mechanically stimulated ERK1/2 phosphorylation in primary bone cell outgrowth cultures and AKT phosphorylation in MC3T3-E1 pre-osteoblast cultures. Expression of mechanosensitive Fos and Ptgs2 genes was enhanced with ISO treatment and reduced with flow in both MC3T3-E1 and primary cultures. Finally, co-treatment of MC3T3-E1 cells with Butax reversed these ISO effects, confirming a critical role for ß2AR in these responses. In combination, these results demonstrate that selective inhibition of ß2AR is sufficient to enhance the anabolic response of the aged skeleton to loading, potentially via direct effects upon osteoblasts. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

4.
JBMR Plus ; 3(5): e10087, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31131340

RESUMO

Nearly all exogenous loading models of bone adaptation apply dynamic loading superimposed upon a time invariant static preload (SPL) in order to ensure stable, reproducible loading of bone. Given that SPL may alter aspects of bone mechanotransduction (eg, interstitial fluid flow), we hypothesized that SPL inhibits bone formation induced by dynamic loading. As a first test of this hypothesis, we utilized a newly developed device that enables stable dynamic loading of the murine tibia with SPLs ≥ -0.01 N. We subjected the right tibias of BALB/c mice (4-month-old females) to dynamic loading (-3.8 N, 1 Hz, 50 cycles/day, 10 s rest) superimposed upon one of three SPLs: -1.5 N, -0.5 N, or -0.03 N. Mice underwent exogenous loading 3 days/week for 3 weeks. Metaphyseal trabecular bone adaptation (µCT) and midshaft cortical bone formation (dynamic histomorphometry) were assessed following euthanasia (day 22). Ipsilateral tibias of mice loaded with a -1.5-N SPL demonstrated significantly less trabecular bone volume/total volume (BV/TV) than contralateral tibias (-12.9%). In contrast, the same dynamic loading superimposed on a -0.03-N SPL significantly elevated BV/TV versus contralateral tibias (12.3%) and versus the ipsilateral tibias of the other SPL groups (-0.5 N: 46.3%, -1.5 N: 37.2%). At the midshaft, the periosteal bone formation rate (p.BFR) induced when dynamic loading was superimposed on -1.5-N and -0.5-N SPLs was significantly amplified in the -0.03-N SPL group (>200%). These data demonstrate that bone anabolism induced by dynamic loading is markedly inhibited by SPL magnitudes commonly implemented in the literature (ie, -0.5 N, -1.5 N). The inhibitory impact of SPL has not been recognized in bone adaptation models and, as such, SPLs have been neither universally reported nor standardized. Our study therefore identifies a previously unrecognized, potent inhibitor of mechanoresponsiveness that has potentially confounded studies of bone adaptation and translation of insights from our field. © 2018 The Authors. JBMR Plus Published by Wiley Periodicals, Inc. on behalf of the American Society for Bone and Mineral Research.

5.
J Musculoskelet Neuronal Interact ; 19(1): 79-93, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30839306

RESUMO

OBJECTIVES: To clarify the effects of neuromuscular dysfunction on hindlimb loading, muscle atrophy, and bone homeostasis. METHODS: We quantified changes to hindlimb loading, muscle atrophy, and bone morphology following either Botulinum toxin A (BTxA) induced muscle paralysis or peripheral nerve injury (PNI) in mice; two in vivo models that we anticipated would differently alter gait and mechanical loading patterns due to their distinct effects on neuromuscular signaling. To confirm the expected behavioral effects of PNI, we assessed mechanical allodynia of the ipsilateral hindlimb using von Frey testing and activity (distance traveled and speed) was monitored in both groups using open field testing. Peak vertical ground reaction forces (GRF) and ankle and knee kinematics during normal locomotion were quantified and used to estimate peak mid-diaphyseal normal strains. Muscle atrophy and trabecular and cortical bone morphology were assessed via high-resolution microCT imaging. RESULTS: BTxA-induced calf paralysis caused severe muscle atrophy and altered gait kinetics and kinematics and reduced gait-induced normal strains. PNI increased mechanical allodynia but did not alter gait, nor did it cause muscle atrophy. We observed that muscle paralysis and PNI both led to severe trabecular bone loss but only BTxA-induced paralysis increased cortical bone resorption. CONCLUSIONS: While mechanical stimuli clearly have essential functions in bone development and adaptation, these data emphasize that neuromuscular signaling, independent of load-induced mechanical strains, may modulate trabecular bone homeostasis in normal and disease states.


Assuntos
Osso e Ossos/fisiologia , Doenças Neuromusculares/fisiopatologia , Paralisia/fisiopatologia , Traumatismos dos Nervos Periféricos/fisiopatologia , Animais , Toxinas Botulínicas Tipo A/farmacologia , Transtornos Neurológicos da Marcha/etiologia , Homeostase/fisiologia , Camundongos , Atrofia Muscular/fisiopatologia , Fármacos Neuromusculares/farmacologia , Paralisia/induzido quimicamente
6.
PLoS One ; 13(11): e0207354, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30427927

RESUMO

At sufficient dose, intramuscular injection of Botulinum toxin A causes muscle wasting that is physiologically consistent with surgical denervation and other types of neuromuscular dysfunction. The aim of this study was to clarify early molecular and micro-RNA alterations in skeletal muscle following Botulinum toxin A-induced muscle paralysis. Quadriceps were analyzed for changes in expression of micro- and messenger RNA and protein levels after a single injection of 0.4, 2 or 4U Botulinum toxin A (/100g body weight). After injection with 2.0U Botulinum toxin A, quadriceps exhibited significant reduction in muscle weight and increased levels of ubiquitin ligase proteins at 7, 14 and 28 days. Muscle miR-1 and miR-133a/b levels were decreased at these time points, whereas a dose-responsive increase in miR-206 expression at day 14 was observed. Expression of the miR-133a/b target genes RhoA, Tgfb1 and Ctfg, and the miR-1/206 target genes Igf-1 and Hdac4, were upregulated at 28 days after Botulinum toxin A injection. Increased levels of Hdac4 protein were observed after injection, consistent with anticipated expression changes in direct and indirect Hdac4 target genes, such as Myog. Our results suggest Botulinum toxin A-induced denervation of muscle shares molecular characteristics with surgical denervation and other types of neuromuscular dysfunction, and implicates miR-133/Tgf-ß1/Ctfg and miR-1/Hdac4/Myog signaling during the resultant muscle atrophy.


Assuntos
Toxinas Botulínicas Tipo A/farmacologia , Histona Desacetilases/genética , MicroRNAs/genética , Músculo Esquelético/efeitos dos fármacos , Fármacos Neuromusculares/farmacologia , Paralisia/induzido quimicamente , Paralisia/genética , Animais , Toxinas Botulínicas Tipo A/administração & dosagem , Feminino , Histona Desacetilases/análise , Injeções Intramusculares , Camundongos Endogâmicos C57BL , MicroRNAs/análise , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Fármacos Neuromusculares/administração & dosagem , Paralisia/fisiopatologia , Transcriptoma/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
7.
Am J Physiol Cell Physiol ; 313(5): C533-C540, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28855162

RESUMO

Transient muscle paralysis engendered by a single injection of botulinum toxin A (BTxA) rapidly induces profound focal bone resorption within the medullary cavity of adjacent bones. While initially conceived as a model of mechanical disuse, osteoclastic resorption in this model is disproportionately severe compared with the modest gait defect that is created. Preliminary studies of bone marrow following muscle paralysis suggested acute upregulation of inflammatory cytokines, including TNF-α and IL-1. We therefore hypothesized that BTxA-induced muscle paralysis would rapidly alter the inflammatory microenvironment and the osteoclastic potential of bone marrow. We tested this hypothesis by defining the time course of inflammatory cell infiltration, osteoinflammatory cytokine expression, and alteration in osteoclastogenic potential in the tibia bone marrow following transient muscle paralysis of the calf muscles. Our findings identified inflammatory cell infiltration within 24 h of muscle paralysis. By 72 h, osteoclast fusion and pro-osteoclastic inflammatory gene expression were upregulated in tibia bone marrow. These alterations coincided with bone marrow becoming permissive to the formation of osteoclasts of greater size and greater nuclei numbers. Taken together, our data are consistent with the thesis that transient calf muscle paralysis induces acute inflammation within the marrow of the adjacent tibia and that these alterations are temporally consistent with a role in mediating muscle paralysis-induced bone resorption.


Assuntos
Reabsorção Óssea/fisiopatologia , Inflamação/etiologia , Músculo Esquelético/efeitos dos fármacos , Osteoclastos/patologia , Paralisia/fisiopatologia , Animais , Medula Óssea/patologia , Reabsorção Óssea/etiologia , Toxinas Botulínicas Tipo A/toxicidade , Feminino , Inflamação/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuromusculares/toxicidade , Paralisia/induzido quimicamente , Paralisia/imunologia , Linfócitos T/imunologia
8.
Bone ; 90: 69-79, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27262776

RESUMO

Myeloma facilitates destruction of bone and marrow. Since physical activity encourages musculoskeletal preservation we evaluated whether low-intensity vibration (LIV), a means to deliver mechanical signals, could protect bone and marrow during myeloma progression. Immunocompromised-mice (n=25) were injected with human-myeloma cells, while 8 (AC) were saline-injected. Myeloma-injected mice (LIV; n=13) were subjected to daily-mechanical loading (15min/d; 0.3g @ 90Hz) while 12 (MM) were sham-handled. At 8w, femurs had 86% less trabecular bone volume fraction (BV/TV) in MM than in AC, yet only a 21% decrease in LIV was observed in comparison to AC, reflecting a 76% increase versus MM. Cortical BV was 21% and 15% lower in MM and LIV, respectively, than in AC; LIV showing 30% improvement over MM. Similar outcomes were observed in the axial skeleton, showing a 35% loss in MM with a 27% improved retention of bone in the L5 of LIV-treated mice as compared to MM. Transcortical-perforations in the femur from myeloma-induced osteolysis were 9× higher in MM versus AC, reduced by 57% in LIV. Serum-TRACP5b, 61% greater in MM versus AC, rose by 33% in LIV compared to AC, a 45% reduction in activity when compared to MM. Histomorphometric analyses of femoral trabecular bone demonstrated a 70% elevation in eroded surfaces of MM versus AC, while measures in LIV were 58% below those in MM. 72% of marrow in the femur of MM mice contained tumor, contrasted by a 31% lower burden in LIV. MM mice (42%) presented advanced-stage necrosis of tibial marrow while present in just 8% of LIV. Myeloma infiltration inversely correlated to measures of bone quality, while LIV slowed the systemic, myeloma-associated decline in bone quality and inhibited tumor progression through the hindlimbs.


Assuntos
Osso e Ossos/patologia , Progressão da Doença , Mieloma Múltiplo/patologia , Vibração , Animais , Medula Óssea/patologia , Linhagem Celular Tumoral , Osso Cortical/patologia , Modelos Animais de Doenças , Fêmur/diagnóstico por imagem , Fêmur/patologia , Citometria de Fluxo , Hematopoese , Humanos , Mecanotransdução Celular , Mieloma Múltiplo/diagnóstico por imagem , Mieloma Múltiplo/imunologia , Osteoclastos/patologia , Fosfatase Ácida Resistente a Tartarato/metabolismo , Microtomografia por Raio-X
9.
Clin Orthop Relat Res ; 473(9): 2825-30, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25804882

RESUMO

BACKGROUND: Short-term muscle atrophy induced by botulinum toxin A (BTxA) has been observed to impair osteogenesis in a rat closed femur fracture model. However, it is unclear whether the underlying mechanism is a direct effect of BTxA on muscle-bone interactions or an indirect effect that is driven by skeletal unloading. Because skeletal trauma in the closed fracture model also leads to disuse atrophy, we sought to mitigate this confounding variable by examining BTxA effects on muscle-bone interactions in two complementary in vivo models in which osteogenesis is induced in the absence of skeletal unloading. The overall aim of this study was to identify a potential strategy to inhibit pathological bone formation and heterotopic ossification (HO). QUESTIONS/PURPOSES: (1) Does muscle paralysis inhibit periosteal osteogenesis induced by a transcortical defect? (2) Does muscle paralysis inhibit heterotopic bone formation stimulated by intramuscular bone morphogenetic protein (BMP) injection? METHODS: Focal osteogenesis was induced in the right hindlimb of mice through surgical initiation of a small transcortical defect in the tibia (fracture callus; n = 7/group) or intramuscular injection of BMP-2 (HO lesion; n = 6/group), both in the presence/absence of adjacent calf paralysis. High-resolution micro-CT images were obtained in all experimental groups 21 days postinduction and total volume (ie, perimeter of periosteal callus or HO lesion) and bone volume (calcified tissue within the total volume) were quantified as primary outcome measures. Finally, these outcome measures were compared to determine the effect of muscle paralysis on inhibition of local osteogenesis in both studies. RESULTS: After a transcortical defect, BTxA-treated mice showed profound inhibition of osteogenesis in the periosteal fracture callus 21 days postsurgery compared with saline-treated mice (total volume: 0.08 ± 0.06 versus 0.42 ± 0.11 mm(3), p < 0.001; bone volume: 0.07 ± 0.05 versus 0.32 ± 0.07 mm(3), p < 0.001). Similarly, BMP-2-induced HO formation was inhibited by adjacent muscle paralysis at the same time point (total volume: 1.42 ± 0.31 versus 3.42 ± 2.11 mm(3), p = 0.034; bone volume: 0.68 ± 0.18 versus 1.36 ± 0.79 mm(3), p = 0.045). CONCLUSIONS: Our data indicate that BTxA-induced neuromuscular inhibition mitigated osteogenesis associated with both a transcortical defect and BMP-2-induced HO. CLINICAL RELEVANCE: Focal neuromuscular inhibition represents a promising new approach that may lead to a new clinical intervention to mitigate trauma-induced HO, a healthcare challenge that is severely debilitating for civilian and war-wounded populations, is costly to both the patient and the healthcare system, and currently lacks effective treatments.


Assuntos
Toxinas Botulínicas Tipo A/farmacologia , Fármacos Neuromusculares/farmacologia , Ossificação Heterotópica/prevenção & controle , Osteogênese/efeitos dos fármacos , Paralisia , Animais , Proteína Morfogenética Óssea 2 , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Ossificação Heterotópica/induzido quimicamente , Ossificação Heterotópica/diagnóstico por imagem , Ossificação Heterotópica/fisiopatologia , Microtomografia por Raio-X
10.
Med Sci Sports Exerc ; 47(5): 1095-103, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25207932

RESUMO

PURPOSE: As our society becomes increasingly sedentary, compliance with exercise regimens that require numerous high-energy activities each week become less likely. Alternatively, given an osteogenic exercise intervention that required minimal effort, it is reasonable to presume that participation would be enhanced. Insertion of brief rest intervals between each cycle of mechanical loading holds potential to achieve this result because substantial osteoblast function is activated by many fewer loading repetitions within each loading bout. Here, we examined the complementary hypothesis that the number of bouts per week of rest-inserted loading could be reduced from three bouts per week without loss of osteogenic efficacy. METHODS: We conducted a series of 3-wk in vivo experiments that noninvasively exposed the right tibiae of mice to either cyclic (1 Hz) or rest-inserted loading interventions and quantified osteoblast function via dynamic histomorphometry. RESULTS: Although reducing loading bouts from three bouts per week (i.e., nine total bouts) to one bout per week (i.e., three total bouts) effectively mitigated the osteogenic benefit of cyclic loading, the same reduction did not significantly reduce periosteal bone formation parameters induced by rest-inserted loading. The osteogenic response was robust to the timing of the rest-inserted loading bouts (three bouts in the first week vs one bout per week for 3 wk). However, elimination of any single bout of the three one-bout-per-week bouts mitigated the osteogenic response to rest-inserted loading. Finally, periosteal osteoblast function assessed after the 3-wk intervention was not sensitive to the timing or number of rest-inserted loading bouts. CONCLUSIONS: We conclude that rest-inserted loading holds potential to retain the osteogenic benefits of mechanical loading with significantly reduced frequency of bouts of activity while also enabling greater flexibility in the timing of the activity.


Assuntos
Osteogênese/fisiologia , Condicionamento Físico Animal/métodos , Suporte de Carga/fisiologia , Animais , Feminino , Camundongos Endogâmicos C57BL , Descanso , Estresse Mecânico , Tíbia/fisiologia , Fatores de Tempo
11.
J Bone Miner Res ; 29(11): 2346-56, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24806738

RESUMO

Intramuscular administration of Botulinum toxin (BTx) has been associated with impaired osteogenesis in diverse conditions of bone formation (eg, development, growth, and healing), yet the mechanisms of neuromuscular-bone crosstalk underlying these deficits have yet to be identified. Motivated by the emerging utility of zebrafish (Danio rerio) as a rapid, genetically tractable, and optically transparent model for human pathologies (as well as the potential to interrogate neuromuscular-mediated bone disorders in a simple model that bridges in vitro and more complex in vivo model systems), in this study, we developed a model of BTx-induced muscle paralysis in adult zebrafish, and we examined its effects on intramembranous ossification during tail fin regeneration. BTx administration induced rapid muscle paralysis in adult zebrafish in a manner that was dose-dependent, transient, and focal, mirroring the paralytic phenotype observed in animal and human studies. During fin regeneration, BTx impaired continued bone ray outgrowth, morphology, and patterning, indicating defects in early osteogenesis. Further, BTx significantly decreased mineralizing activity and crystalline mineral accumulation, suggesting delayed late-stage osteoblast differentiation and/or altered secondary bone apposition. Bone ray transection proximal to the amputation site focally inhibited bone outgrowth in the affected ray, implicating intra- and/or inter-ray nerves in this process. Taken together, these studies demonstrate the potential to interrogate pathological features of BTx-induced osteoanabolic dysfunction in the regenerating zebrafish fin, define the technological toolbox for detecting bone growth and mineralization deficits in this process, and suggest that pathways mediating neuromuscular regulation of osteogenesis may be conserved beyond established mammalian models of bone anabolic disorders.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Toxinas Botulínicas/toxicidade , Calcificação Fisiológica/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Paralisia/metabolismo , Peixe-Zebra/metabolismo , Adulto , Animais , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Osteoblastos/metabolismo , Osteoblastos/patologia , Paralisia/induzido quimicamente , Paralisia/patologia
12.
PLoS One ; 9(1): e84868, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24404194

RESUMO

Age-related decline in periosteal adaptation negatively impacts the ability to utilize exercise to enhance bone mass and strength in the elderly. We recently observed that in senescent animals subject to cyclically applied loading, supplementation with Cyclosporin A (CsA) substantially enhanced the periosteal bone formation rates to levels observed in young animals. We therefore speculated that if the CsA supplement could enhance bone response to a variety of types of mechanical stimuli, this approach could readily provide the means to expand the range of mild stimuli that are robustly osteogenic at senescence. Here, we specifically hypothesized that a given CsA supplement would enhance bone formation induced in the senescent skeleton by both cyclic (1-Hz) and rest-inserted loading (wherein a 10-s unloaded rest interval is inserted between each load cycle). To examine this hypothesis, the right tibiae of senescent female C57BL/6 mice (22 Mo) were subjected to cyclic or rest-inserted loading supplemented with CsA at 3.0 mg/kg. As previously, we initially found that while the periosteal bone formation rate (p.BFR) induced by cyclic loading was enhanced when supplemented with 3.0 mg/kg CsA (by 140%), the response to rest-inserted loading was not augmented at this CsA dosage. In follow-up experiments, we observed that while a 30-fold lower CsA dosage (0.1 mg/kg) significantly enhanced p.BFR induced by rest-inserted loading (by 102%), it was ineffective as a supplement with cyclic loading. Additional experiments and statistical analysis confirmed that the dose-response relations were significantly different for cyclic versus rest-inserted loading, only because the two stimuli required distinct CsA dosages for efficacy. While not anticipated a priori, clarifying the complexity underlying the observed interaction between CsA dosage and loading type holds potential for insight into how bone response to a broad range of mechanical stimuli may be substantially enhanced in the senescent skeleton.


Assuntos
Osso e Ossos/efeitos dos fármacos , Ciclosporina/administração & dosagem , Osteogênese/efeitos dos fármacos , Fatores Etários , Envelhecimento/fisiologia , Animais , Relação Dose-Resposta a Droga , Feminino , Camundongos
13.
Bone ; 57(2): 413-22, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24063948

RESUMO

When the skeleton is catabolically challenged, there is great variability in the timing and extent of bone resorption observed at cancellous and cortical bone sites. It remains unclear whether this resorptive heterogeneity, which is often evident within a single bone, arises from increased permissiveness of specific sites to bone resorption or localized resorptive events of varied robustness. To explore this question, we used the mouse model of calf paralysis induced bone loss, which results in metaphyseal and diaphyseal bone resorption of different timing and magnitude. Given this phenotypic pattern of resorption, we hypothesized that bone loss in the proximal tibia metaphysis and diaphysis occurs through resorption events that are spatially and temporally distinct. To test this hypothesis, we undertook three complimentary in vivo/µCT imaging studies. Specifically, we defined spatiotemporal variations in endocortical bone resorption during the 3weeks following calf paralysis, applied a novel image registration approach to determine the location where bone resorption initiates within the proximal tibia metaphysis, and explored the role of varied basal osteoclast activity on the magnitude of bone loss initiation in the metaphysis using µCT based bone resorption parameters. A differential response of metaphyseal and diaphyseal bone resorption was observed throughout each study. Acute endocortical bone loss following muscle paralysis occurred almost exclusively within the metaphyseal compartment (96.5% of total endocortical bone loss within 6days). Using our trabecular image registration approach, we further resolved the initiation of metaphyseal bone loss to a focused region of significant basal osteoclast function (0.03mm(3)) adjacent to the growth plate. This correlative observation of paralysis induced bone loss mediated by basal growth plate cell dynamics was supported by the acute metaphyseal osteoclastic response of 5-week vs. 13-month-old mice. Specifically, µCT based bone resorption rates normalized to initial trabecular surface (BRRBS) were 3.7-fold greater in young vs. aged mice (2.27±0.27µm(3)/µm(2)/day vs. 0.60±0.44µm(3)/µm(2)/day). In contrast to the focused bone loss initiation in the metaphysis, diaphyseal bone loss initiated homogeneously throughout the long axis of the tibia predominantly in the second week following paralysis (81.3% of diaphyseal endocortical expansion between days 6 and 13). The timing and homogenous nature are consistent with de novo osteoclastogenesis mediating the diaphyseal resorption. Taken together, our data suggests that tibial metaphyseal and diaphyseal bone loss induced by transient calf paralysis are spatially and temporally discrete events. In a broader context, these findings are an essential first step toward clarifying the timing and origins of multiple resorptive events that would require targeting to fully inhibit bone loss following neuromuscular trauma.


Assuntos
Reabsorção Óssea/complicações , Reabsorção Óssea/fisiopatologia , Diáfises/fisiopatologia , Músculo Esquelético/fisiopatologia , Paralisia/complicações , Paralisia/fisiopatologia , Tíbia/fisiopatologia , Animais , Reabsorção Óssea/diagnóstico por imagem , Diáfises/diagnóstico por imagem , Camundongos , Músculo Esquelético/diagnóstico por imagem , Paralisia/diagnóstico por imagem , Tíbia/diagnóstico por imagem , Fatores de Tempo , Microtomografia por Raio-X
14.
PLoS One ; 8(9): e74205, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24040202

RESUMO

Bone has long been established to be a highly mechanosensitive tissue. When subjected to mechanical loading, bone exhibits profoundly different anabolic responses depending on the temporal pattern in which the stimulus is applied. This phenomenon has been termed temporal processing, and involves complex signal amplification mechanisms that are largely unidentified. In this study, our goal was to characterize transcriptomic perturbations arising from the insertion of intermittent rest periods (a temporal variation with profound effects on bone anabolism) in osteoblastic cells subjected to fluid flow, and assess the utility of these perturbations to identify signaling pathways that are differentially activated by this temporal variation. At the level of the genome, we found that the common and differential alterations in gene expression arising from the two flow conditions were distributionally distinct, with the differential alterations characterized by many small changes in a large number of genes. Using bioinformatics analysis, we identified distinct up- and down-regulation transcriptomic signatures associated with the insertion of rest intervals, and found that the up-regulation signature was significantly associated with MAPK signaling. Confirming the involvement of the MAPK pathway, we found that the insertion of rest intervals significantly elevated flow-induced p-ERK1/2 levels by enabling a second spike in activity that was not observed in response to continuous flow. Collectively, these studies are the first to characterize distinct transcriptomic perturbations in bone cells subjected to continuous and intermittent stimulation, and directly demonstrate the utility of systems-based transcriptomic analysis to identify novel acute signaling pathways underlying temporal processing in bone cells.


Assuntos
Osso e Ossos/metabolismo , Regulação da Expressão Gênica , Sistema de Sinalização das MAP Quinases , Mecanotransdução Celular , Osteoblastos/metabolismo , Transcriptoma , Animais , Osso e Ossos/citologia , Linhagem Celular , Perfilação da Expressão Gênica , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Osteoblastos/citologia , Reologia , Estresse Mecânico
15.
Dev Biol ; 380(1): 99-110, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23652029

RESUMO

The ESET (also called SETDB1) protein contains an N-terminal tudor domain that mediates protein-protein interactions and a C-terminal SET domain that catalyzes methylation of histone H3 at lysine 9. We report here that ESET protein is transiently upregulated in prehypertrophic chondrocytes in newborn mice. To investigate the in vivo effects of ESET on chondrocyte differentiation, we generated conditional knockout mice to specifically eliminate the catalytic SET domain of ESET protein only in mesenchymal cells. Such deletion of the ESET gene caused acceleration of chondrocyte hypertrophy in both embryos and young animals, depleting chondrocytes that are otherwise available to form epiphyseal plates for endochondral bone growth. ESET-deficient mice are thus characterized by defective long bone growth and trabecular bone formation. To understand the underlying mechanism for ESET regulation of chondrocytes, we carried out co-expression experiments and found that ESET associates with histone deacetylase 4 to bind and inhibit the activity of Runx2, a hypertrophy-promoting transcription factor. Repression of Runx2-mediated gene transactivation by ESET is dependent on its H3-K9 methyltransferase activity as well as its associated histone deacetylase activity. In addition, knockout of ESET is associated with repression of Indian hedgehog gene in pre- and early hypertrophic chondrocytes. Together, these results provide clear evidence that ESET controls hypertrophic differentiation of growth plate chondrocytes and endochondral ossification during embryogenesis and postnatal development.


Assuntos
Condrócitos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Lâmina de Crescimento/metabolismo , Histona-Lisina N-Metiltransferase/fisiologia , Alelos , Animais , Osso e Ossos/embriologia , Osso e Ossos/metabolismo , Cartilagem/embriologia , Diferenciação Celular , Epigênese Genética , Proteínas Hedgehog/metabolismo , Histona Desacetilases/metabolismo , Histona-Lisina N-Metiltransferase/genética , Mesoderma/citologia , Camundongos , Camundongos Knockout , Estrutura Terciária de Proteína
16.
Ageing Res Rev ; 11(3): 353-60, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22240208

RESUMO

Physical exercise is thought to hold promise as a non-invasive countermeasure against skeletal fragility arising from post-menopausal and age-related osteoporosis. Importantly, mechanical loading and exercise are capable of increasing bone size via periosteal expansion, which by far, is the most effective means of strengthening the structure of a given bone. The focus of this review was to therefore explore whether exercise has the potential to increase periosteal modeling and bone size in the senescent skeleton. A survey of exercise trials in humans suggests that exercise interventions that enhance periosteal modeling in the young skeleton fail to do the same in the elderly skeleton. Underlying this ineffectiveness, in vitro studies indicate that aging lowers basal levels of cell function and degrades bone mechanotransduction at a variety of levels from altered second messenger signaling to gene expression driving proliferation and/or differentiation. Given these age-related alterations, the ultimate efficacy of an exercise intervention may depend upon concurrent supplementation that directly address deficits in signaling and/or cell function. In this context, in vivo animal models of mechanical loading that simulate the muted periosteal adaptation in the elderly hold potential to examine the efficacy of countermeasures. Preliminary in vivo experiments suggest that pharmacologically counteracting age-related deficits in cellular function can restore exercise induced periosteal modeling in the senescent skeleton to levels observed in young animals. If the safety and efficacy of this strategy were to be confirmed for human use, it would enable the utilization of exercise as a viable countermeasure against skeletal fragility at senescence.


Assuntos
Envelhecimento/fisiologia , Remodelação Óssea/fisiologia , Exercício Físico/fisiologia , Mecanotransdução Celular/fisiologia , Osteogênese/fisiologia , Animais , Senescência Celular/fisiologia , Humanos , Osteoporose/fisiopatologia , Osteoporose/prevenção & controle , Ensaios Clínicos Controlados Aleatórios como Assunto/métodos
17.
Bone ; 50(1): 14-22, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21920486

RESUMO

Mechanical loading of the skeleton, as induced by muscle function during activity, plays a critical role in maintaining bone homeostasis. It is not understood, however, whether diminished loading (and thus diminished mechanical stimuli) directly mediates the bone resorption that is associated with disuse. Our group has recently developed a murine model in which we have observed rapid and profound bone loss in the tibia following transient paralysis of the calf muscles. As cortical bone loss is achieved via rapid endocortical expansion without alterations in periosteal morphology, we believe this model holds unique potential to explore the spatial relation between altered mechanical stimuli and subsequent bone resorption. Given the available literature, we hypothesized that endocortical resorption following transient muscle paralysis would be spatially homogeneous. To test this hypothesis, we first validated an image registration algorithm that quantified site-specific cortical bone alterations with high precision and accuracy. We then quantified endocortical expansion in the tibial diaphysis within 21 days following transient muscle paralysis and found that, within the analyzed mid-diaphyseal region (3.15 mm), site-specific bone loss was focused on the anterior surface in the proximal region but shifted to the posterior surface at the distal end of the analyzed volume. This site-specific, and highly repeatable biologic response suggests active osteoclast chemotaxis or focal activation of osteoclastic resorption underlies the spatially consistent endocortical resorption induced by transient muscle paralysis. Clarifying this relation holds potential to yield unique insight into how the removal of factors critical for bone homeostasis acutely precipitates local modulation of cellular responses within bone.


Assuntos
Reabsorção Óssea/patologia , Músculo Esquelético/fisiopatologia , Paralisia/fisiopatologia , Tíbia/anatomia & histologia , Tíbia/patologia , Animais , Feminino , Humanos , Processamento de Imagem Assistida por Computador/métodos , Camundongos , Camundongos Endogâmicos C57BL , Microtomografia por Raio-X/métodos
18.
FASEB J ; 26(3): 1110-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22125315

RESUMO

A unilateral injection of botulinum toxin A (BTxA) in the calf induces paralysis and profound loss of ipsalateral trabecular bone within days. However, the cellular mechanism underlying acute muscle paralysis-induced bone loss (MPIBL) is poorly understood. We hypothesized that MPIBL arises via rapid and extensive osteoclastogenesis. We performed a series of in vivo experiments to explore this thesis. First, we observed elevated levels of the proosteoclastogenic cytokine receptor activator for nuclear factor-κB ligand (RANKL) within the proximal tibia metaphysis at 7 d after muscle paralysis (+113%, P<0.02). Accordingly, osteoclast numbers were increased 122% compared with the contralateral limb at 5 d after paralysis (P=0.04) and MPIBL was completely blocked by treatment with human recombinant osteoprotegerin (hrOPG). Further, conditional deletion of nuclear factor of activated T-cells c1 (NFATc1), the master regulator of osteoclastogenesis, completely inhibited trabecular bone loss (-2.2±11.9%, P<0.01). All experiments included negative control assessments of contralateral limbs and/or within-animal pre- and postintervention imaging. In summary, transient muscle paralysis induced acute RANKL-mediated osteoclastogenesis resulting in profound local bone resorption. Elucidation of the pathways that initiate osteoclastogenesis after paralysis may identify novel targets to inhibit bone loss and prevent fractures.


Assuntos
Osso e Ossos/metabolismo , Músculo Esquelético/metabolismo , Osteoclastos/metabolismo , Paralisia/metabolismo , Ligante RANK/metabolismo , Animais , Reabsorção Óssea/metabolismo , Reabsorção Óssea/prevenção & controle , Toxinas Botulínicas Tipo A/toxicidade , Contagem de Células , Feminino , Deleção de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/patologia , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/patologia , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Osteoprotegerina/farmacologia , Paralisia/induzido quimicamente , Músculo Quadríceps/metabolismo , Músculo Quadríceps/patologia , Proteínas Recombinantes/farmacologia , Tíbia/metabolismo , Fatores de Tempo
19.
PLoS Comput Biol ; 6(9)2010 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-20838577

RESUMO

The increasing incidence of osteoporosis worldwide requires anabolic treatments that are safe, effective, and, critically, inexpensive given the prevailing overburdened health care systems. While vigorous skeletal loading is anabolic and holds promise, deficits in mechanotransduction accrued with age markedly diminish the efficacy of readily complied, exercise-based strategies to combat osteoporosis in the elderly. Our approach to explore and counteract these age-related deficits was guided by cellular signaling patterns across hierarchical scales and by the insight that cell responses initiated during transient, rare events hold potential to exert high-fidelity control over temporally and spatially distant tissue adaptation. Here, we present an agent-based model of real-time Ca(2+)/NFAT signaling amongst bone cells that fully described periosteal bone formation induced by a wide variety of loading stimuli in young and aged animals. The model predicted age-related pathway alterations underlying the diminished bone formation at senescence, and hence identified critical deficits that were promising targets for therapy. Based upon model predictions, we implemented an in vivo intervention and show for the first time that supplementing mechanical stimuli with low-dose Cyclosporin A can completely rescue loading induced bone formation in the senescent skeleton. These pre-clinical data provide the rationale to consider this approved pharmaceutical alongside mild physical exercise as an inexpensive, yet potent therapy to augment bone mass in the elderly. Our analyses suggested that real-time cellular signaling strongly influences downstream bone adaptation to mechanical stimuli, and quantification of these otherwise inaccessible, transient events in silico yielded a novel intervention with clinical potential.


Assuntos
Osso e Ossos/fisiologia , Senescência Celular/fisiologia , Biologia Computacional/métodos , Osteogênese/fisiologia , Suporte de Carga/fisiologia , Envelhecimento/efeitos dos fármacos , Envelhecimento/fisiologia , Análise de Variância , Animais , Fenômenos Biomecânicos/fisiologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Cálcio/metabolismo , Senescência Celular/efeitos dos fármacos , Ciclosporina/farmacologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fatores de Transcrição NFATC/metabolismo , Osteócitos/fisiologia , Osteogênese/efeitos dos fármacos , Osteoporose/patologia , Reprodutibilidade dos Testes , Tíbia/citologia
20.
J Biomech ; 43(14): 2765-70, 2010 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-20674920

RESUMO

The critical role that mechanical stimuli serve in mediating bone repair is recognized but incompletely understood. Further, previous attempts to understand this role have utilized application of externally applied mechanical loads to study the tissue's response. In this project, we have therefore endeavored to capitalize on bone's own consistently diverse loading environment to develop a novel model that would enable assessment of the influence of physiologically engendered mechanical stimuli on cortical defect repair. We used an inverse dynamics approach with finite element analysis (FEA) to first quantify normal strain distributions generated in mouse tibia during locomotion. The strain environment of the tibia, as previously reported for other long bones, was found to arise primarily due to bending and was consistent in orientation through the stance phase of gait. Based on these data, we identified three regions within a transverse cross-section of the mid-diaphysis as uniform locations of either peak tension, peak compression, or the neutral axis of bending (i.e. minimal strain magnitude). We then used FEA to quantify the altered strain environment that would be produced by a 0.6mm diameter cylindrical cortical bone defect at each diaphyseal site and, in an in situ study confirmed our ability to accurately place defects at the desired diaphyseal locations. The resulting model will enable the exploration of cortical bone healing within the context of physiologically engendered mechanical strain.


Assuntos
Locomoção/fisiologia , Tíbia/lesões , Tíbia/fisiopatologia , Fraturas da Tíbia/fisiopatologia , Animais , Fenômenos Biomecânicos , Feminino , Análise de Elementos Finitos , Consolidação da Fratura/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Estresse Mecânico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...