Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 134(10)2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38512413

RESUMO

Elevated bone resorption and diminished bone formation have been recognized as the primary features of glucocorticoid-associated skeletal disorders. However, the direct effects of excess glucocorticoids on bone turnover remain unclear. Here, we explored the outcomes of exogenous glucocorticoid treatment on bone loss and delayed fracture healing in mice and found that reduced bone turnover was a dominant feature, resulting in a net loss of bone mass. The primary effect of glucocorticoids on osteogenic differentiation was not inhibitory; instead, they cooperated with macrophages to facilitate osteogenesis. Impaired local nutrient status - notably, obstructed fatty acid transportation - was a key factor contributing to glucocorticoid-induced impairment of bone turnover in vivo. Furthermore, fatty acid oxidation in macrophages fueled the ability of glucocorticoid-liganded receptors to enter the nucleus and then promoted the expression of BMP2, a key cytokine that facilitates osteogenesis. Metabolic reprogramming by localized fatty acid delivery partly rescued glucocorticoid-induced pathology by restoring a healthier immune-metabolic milieu. These data provide insights into the multifactorial metabolic mechanisms by which glucocorticoids generate skeletal disorders, thus suggesting possible therapeutic avenues.


Assuntos
Remodelação Óssea , Glucocorticoides , Osteogênese , Animais , Camundongos , Glucocorticoides/farmacologia , Osteogênese/efeitos dos fármacos , Remodelação Óssea/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/imunologia , Macrófagos/efeitos dos fármacos , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 2/genética , Ácidos Graxos/metabolismo , Osso e Ossos/metabolismo , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/imunologia , Microambiente Celular/efeitos dos fármacos
2.
Bioengineering (Basel) ; 9(10)2022 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-36290493

RESUMO

We develop a poly (lactic-co-glycolic acid)/ß-calcium phosphate (PLGA/TCP)-based scaffold through a three-dimensional (3D) printing technique incorporating icaritin (ICT), a unique phytomolecule, and secretome derived from human fetal mesenchymal stem cells (HFS), to provide mechanical support and biological cues for stimulating bone defect healing. With the sustained release of ICT and HFS from the composite scaffold, the cell-free scaffold efficiently facilitates the migration of MSCs and promotes bone regeneration at the femoral defect site in the ovariectomy (OVX)-induced osteoporotic rat model. Furthermore, mechanism study results indicate that the combination of ICT and HFS additively activates the Integrin-FAK (focal adhesion kinase)-ERK1/2 (extracellular signal-regulated kinase 1/2)-Runx2 (Runt-related transcription factor 2) axis, which could be linked to the beneficial recruitment of MSCs to the implant and subsequent osteogenesis enhancement. Collectively, the PLGA/TCP/ICT/HFS (P/T/I/S) bioactive scaffold is a promising biomaterial for repairing osteoporotic bone defects, which may have immense implications for their translation to clinical practice.

3.
ACS Nano ; 16(11): 18071-18089, 2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36108267

RESUMO

Bone grafting is frequently conducted to treat bone defects caused by trauma and tumor removal, yet with significant medical and socioeconomic burdens. Space-occupying bone substitutes remain challenging in the control of osteointegration, and meanwhile activation of endogenous periosteal cells by using non-space-occupying implants to promote new bone formation becomes another therapeutic strategy. Here, we fabricated a magnesium-based artificial bandage with optimal micropatterns for activating periosteum-associated biomineralization. Collagen was self-assembled on the surface of magnesium oxide nanoparticles embedded electrospun fibrous membranes as a hierarchical bandage structure to facilitate the integration with periosteum in situ. After the implantation on the surface of cortical bone in vivo, magnesium ions were released to generate a pro-osteogenic immune microenvironment by activating the endogenous periosteal macrophages into M2 phenotype and, meanwhile, promote blood vessel formation and neurite outgrowth. In a cortical bone defect model, magnesium-based artificial bandage guided the surrounding newly formed bone tissue to cover the defected area. Taken together, our study suggests that the strategy of stimulating bone formation can be achieved with magnesium delivery to periosteum in situ and the proposed periosteal bandages act as a bioactive media for accelerating bone healing.


Assuntos
Nanopartículas , Osteogênese , Óxido de Magnésio/farmacologia , Regeneração Óssea , Magnésio/farmacologia , Periósteo/fisiologia , Periósteo/transplante , Osso Cortical , Bandagens
4.
Biomaterials ; 288: 121751, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36031456

RESUMO

Endowing biomaterials with functional elements enhances their biological properties effectively. However, improving bioactivity and biosafety simultaneously is still highly desirable. Herein, cerium (Ce) and copper (Cu) are incorporated into silicocarnotite (CPS) to modulate the constitution and microstructure for degradability, bioactivity and biosafety regulation. Our results demonstrated that introducing Ce suppressed scaffold degradation, while, co-incorporation of both Ce and Cu accelerated degradability. Osteogenic effect of CPS in vitro was promoted by Ce and optimized by Cu, and Ce-induced angiogenic inhibition could be mitigated by cell coculture method and reversed by Ce-Cu co-incorporation. Ce enhanced osteogenic and angiogenic properties of CPS in a dose-dependent manner in vivo, and Cu-Ce coexistence exhibited optimal bioactivity and satisfactory biosafety. This work demonstrated that coculture in vitro was more appropriately reflecting the behavior of implanted biomaterials in vivo. Interactive effects of multi-metal elements were promising to enhance bioactivity and biosafety concurrently. The present work provided a promising biomaterial for bone repair and regeneration, and offered a comprehensive strategy to design new biomaterials which aimed at adjustable degradation behavior, and enhanced bioactivity and biosafety.


Assuntos
Cério , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Fosfatos de Cálcio , Cério/química , Cério/farmacologia , Contenção de Riscos Biológicos , Cobre/química , Cobre/farmacologia , Osteogênese , Silicatos
5.
Adv Sci (Weinh) ; 9(21): e2202102, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35652188

RESUMO

Peripheral nerve injury is a challenging orthopedic condition that can be treated by autograft transplantation, a gold standard treatment in the current clinical setting. Nevertheless, limited availability of autografts and potential morbidities in donors hampers its widespread application. Bioactive scaffold-based tissue engineering is a promising strategy to promote nerve regeneration. Additionally, magnesium (Mg) ions enhance nerve regeneration; however, an effectively controlled delivery vehicle is necessary to optimize their in vivo therapeutic effects. Herein, a bisphosphonate-based injectable hydrogel exhibiting sustained Mg2+ delivery for peripheral nerve regeneration is developed. It is observed that Mg2+ promoted neurite outgrowth in a concentration-dependent manner by activating the PI3K/Akt signaling pathway and Sema5b. Moreover, implantation of polycaprolactone (PCL) conduits filled with Mg2+ -releasing hydrogel in 10 mm nerve defects in rats significantly enhanced axon regeneration and remyelination at 12 weeks post-operation compared to the controls (blank conduits or conduits filled with Mg2+ -absent hydrogel). Functional recovery analysis reveals enhanced reinnervation in the animals treated with the Mg2+ -releasing hydrogel compared to that in the control groups. In summary, the Mg2+ -releasing hydrogel combined with the 3D-engineered PCL conduit promotes peripheral nerve regeneration and functional recovery. Thus, a new strategy to facilitate the repair of challenging peripheral nerve injuries is proposed.


Assuntos
Hidrogéis , Magnésio , Regeneração Nervosa , Traumatismos dos Nervos Periféricos , Animais , Axônios , Hidrogéis/farmacologia , Magnésio/farmacologia , Regeneração Nervosa/fisiologia , Traumatismos dos Nervos Periféricos/terapia , Fosfatidilinositol 3-Quinases/farmacologia , Poliésteres , Ratos , Engenharia Tecidual/métodos , Alicerces Teciduais
6.
Nat Commun ; 13(1): 427, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-35058428

RESUMO

Epididymal white adipose tissue (eWAT) secretes an array of cytokines to regulate the metabolism of organs and tissues in high-fat diet (HFD)-induced obesity, but its effects on bone metabolism are not well understood. Here, we report that macrophages in eWAT are a main source of osteopontin, which selectively circulates to the bone marrow and promotes the degradation of the bone matrix by activating osteoclasts, as well as modulating bone marrow-derived macrophages (BMDMs) to engulf the lipid droplets released from adipocytes in the bone marrow of mice. However, the lactate accumulation induced by osteopontin regulation blocks both lipolysis and osteoclastogenesis in BMDMs by limiting the energy regeneration by ATP6V0d2 in lysosomes. Both surgical removal of eWAT and local injection of either clodronate liposomes (for depleting macrophages) or osteopontin-neutralizing antibody show comparable amelioration of HFD-induced bone loss in mice. These results provide an avenue for developing therapeutic strategies to mitigate obesity-related bone disorders.


Assuntos
Tecido Adiposo/metabolismo , Osso e Ossos/metabolismo , Epididimo/metabolismo , Homeostase , Macrófagos/metabolismo , Osteopontina/metabolismo , Tecido Adiposo/diagnóstico por imagem , Tecido Adiposo Branco/diagnóstico por imagem , Tecido Adiposo Branco/metabolismo , Animais , Peso Corporal , Reabsorção Óssea/patologia , Osso e Ossos/diagnóstico por imagem , Antígeno CD11b/metabolismo , Osso Esponjoso/diagnóstico por imagem , Osso Esponjoso/metabolismo , Dieta Hiperlipídica , Inflamação/patologia , Metabolismo dos Lipídeos , Lisossomos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Tamanho do Órgão , Subunidades Proteicas/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Microtomografia por Raio-X
7.
Bioact Mater ; 6(11): 4176-4185, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33997501

RESUMO

Displaced fractures of patella often require open reduction surgery and internal fixation to restore the extensor continuity and articular congruity. Fracture fixation with biodegradable magnesium (Mg) pins enhanced fracture healing. We hypothesized that fixation with Mg pins and their degradation over time would enhance healing of patellar fracture radiologically, mechanically, and histologically. Transverse patellar fracture surgery was performed on thirty-two 18-weeks old female New Zealand White Rabbits. The fracture was fixed with a pin made of stainless steel or pure Mg, and a figure-of-eight stainless steel band wire. Samples were harvested at week 8 or 12, and assessed with microCT, tensile testing, microindentation, and histology. Microarchitectural analysis showed that Mg group showed 12% higher in the ratio of bone volume to tissue volume at week 8, and 38.4% higher of bone volume at week 12. Tensile testing showed that the failure load and stiffness of Mg group were 66.9% and 104% higher than the control group at week 8, respectively. At week 12, Mg group was 60.8% higher in ultimate strength than the control group. Microindentation showed that, compared to the Control group, Mg group showed 49.9% higher Vickers hardness and 31% higher elastic modulus at week 8 and 12, respectively. At week 12, the new bone of Mg group remodelled to laminar bone, but those of the control group remained woven bone-like. Fixation of transverse patellar fracture with Mg pins and its degradation enhanced new bone formation and mechanical properties of the repaired patella compared to the Control group.

8.
Bioact Mater ; 6(8): 2511-2522, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33665494

RESUMO

INTRODUCTION: Magnesium (Mg) has a prophylactic potential against the onset of hyperlipidemia. Similar to statin, Mg is recommended as lipid-lowering medication for hypercholesterolemia and concomitantly exhibits an association with increased bone mass. The combination of statin with Mg ions (Mg2+) may be able to alleviate the high-fat diet (HFD)-induced bone loss and reduce the side-effects of statin. This study aimed to explore the feasibility of combined Mg2+ with simvastatin (SIM) for treating HFD-induced bone loss in mice and the involving mechanisms. MATERIALS AND METHODS: C57BL/6 male mice were fed with a HFD or a normal-fat diet (NFD). Mice were intraperitoneally injected SIM and/or orally received water with additional Mg2+ until sacrificed. Enzyme-linked immunosorbent assay was performed to measure cytokines and cholesterol in serum and liver lysates. Bone mineral density (BMD) and microarchitecture were assessed by micro-computed tomography (µCT) in different groups. The adipogenesis in palmitate pre-treated HepG2 cells was performed under various treatments. RESULTS: µCT analysis showed that the trabecular bone mass was significantly lower in the HFD-fed group than that in NFD-fed group since week 8. The cortical thickness in HFD-fed group had a significant decrease at week 24, as compared with NFD-fed group. The combination of Mg2+ and SIM significantly attenuated the trabecular bone loss in HFD-fed mice via arresting the osteoclast formation and bone resorption. Besides, such combination also reduced the hepatocytic synthesis of cholesterol and inhibited matrix metallopeptidase 13 (Mmp13) mRNA expression in pre-osteoclasts. CONCLUSIONS: The combination of Mg2+ and SIM shows a synergistic effect on attenuating the HFD-induced bone loss. Our current formulation may be a cost-effective alternative treatment to be indicated for obesity-related bone loss.

9.
Tissue Eng Part A ; 27(1-2): 87-102, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32375579

RESUMO

Distraction osteogenesis (DO) is a well-established surgical technique for treating bone defect and limb lengthening. The major drawback of DO is the long treatment period as the external fixator has to be kept in place until consolidation is completed. Calcitonin gene-related peptide (CGRP) has been reported to promote angiogenesis by affecting endothelial progenitor cells (EPCs) in limb ischemia and wound healing. Thus, the goal of this study was to evaluate the angiogenic effect of exogenous CGRP on bone regeneration in a rat DO model. Exogenous CGRP was directly injected into the bone defect after each cycle of distraction in vivo. Microcomputed tomography, biomechanical test, and histological analysis were performed to assess the new bone formation. Angiography and immunofluorescence were performed to assess the formation of blood vessels. CD31+CD144+ EPCs in the bone defect were quantified with flow cytometry. In in vitro study, bone marrow stem cells (BMSCs) were used to investigate the effect of CGRP on EPCs production during endothelial differentiation. Our results showed that CGRP significantly promoted bone regeneration and vessel formation after consolidation. CGRP significantly increased the fraction of CD31+CD144+EPCs and the capillary density in the bone defect at the end of distraction phase. CGRP increased EPC population in the endothelial differentiation of BMSCs in vitro by activating PI3K/AKT signaling pathway. Furthermore, differentiated EPCs rapidly assembled into tube-like structures and promoted osteogenic differentiation of BMSCs. In conclusion, CGRP increased EPC population and promoted blood vessel formation and bone regeneration at the defect region in a DO model. Impact statement Distraction osteogenesis (DO) is a well-established surgical technique for limb lengthening and bone defect. The disadvantage of this technique is that external fixator is needed to be kept in place for about 12 months. This may result in increased risk of infection, financial burden, and negative psychological impacts. In this study, we have injected calcitonin gene-related peptide (CGRP) into the defect region after distraction and found that CGRP enhanced vessel formation and bone regeneration in a rat DO model. This suggests that a controlled delivery system for CGRP could be developed and applied clinically for accelerating bone regeneration in patients with DO.


Assuntos
Osteogênese por Distração , Osteogênese , Animais , Regeneração Óssea , Calcitonina , Peptídeo Relacionado com Gene de Calcitonina , Fosfatidilinositol 3-Quinases , Ratos , Microtomografia por Raio-X
10.
Adv Sci (Weinh) ; 7(8): 1902443, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32328412

RESUMO

Biodegradable Mg-based metals may be promising orthopedic implants for treating challenging bone diseases, attributed to their desirable mechanical and osteopromotive properties. This Review summarizes the current status and future research trends for Mg-based orthopedic implants. First, the properties between Mg-based implants and traditional orthopedic implants are compared on the following aspects: in vitro and in vivo degradation mechanisms of Mg-based implants, peri-implant bone responses, the fate of the degradation products, and the cellular and molecular mechanisms underlying the beneficial effects of Mg ions on osteogenesis. Then, the preclinical studies conducted at the low weight bearing sites of animals are introduced. The innovative strategies (for example, via designing Mg-containing hybrid systems) are discussed to address the limitations of Mg-based metals prior to their clinical applications at weight-bearing sites. Finally, the available clinical studies are summarized and the challenges and perspectives of Mg-based orthopedic implants are discussed. Taken together, the progress made on the development of Mg-based implants in basic, translational, and clinical research has laid down a foundation for developing a new era in the treatment of challenging and prevalent bone diseases.

11.
Ann Rheum Dis ; 78(4): 551-561, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30745310

RESUMO

OBJECTIVES: Wnt16 is implicated in bone fracture and bone mass accrual both in animals and humans. However, its functional roles and molecular mechanism in chondrocyte differentiation and osteoarthritis (OA) pathophysiology remain largely undefined. In this study, we analysed its mechanistic association and functional relationship in OA progression in chondrocyte lineage. METHODS: The role of Wnt16 during skeletal development was examined by Col2a1-Wnt16 transgenic mice and Wnt16fl/fl;Col2a1-Cre (Wnt16-cKO) mice. OA progression was assessed by micro-CT analysis and Osteoarthritis Research Society International score after anterior cruciate ligament transection (ACLT) surgery with Wnt16 manipulation by adenovirus intra-articular injection. The molecular mechanism was investigated in vitro using 3D chondrocyte pellet culture and biochemical analyses. Histological analysis was performed in mouse joints and human cartilage specimens. RESULTS: Wnt16 overexpression in chondrocytes in mice significantly inhibited chondrocyte hypertrophy during skeletal development. Wnt16 deficiency exaggerated OA progression, whereas intra-articular injection of Ad-Wnt16 markedly attenuated ACLT-induced OA. Cellular and molecular analyses showed that, instead of ß-catenin and calcium pathways, Wnt16 activated the planar cell polarity (PCP) and JNK pathway by interacting mainly with AP2b1, and to a lesser extend Ror2 and CD146, and subsequently induced PTHrP expression through phosphor-Raptor mTORC1 pathway. CONCLUSIONS: Our findings indicate that Wnt16 activates PCP/JNK and crosstalks with mTORC1-PTHrP pathway to inhibit chondrocyte hypertrophy. Our preclinical study suggests that Wnt16 may be a potential therapeutic target for OA treatment.


Assuntos
Artrite Experimental/patologia , Osteoartrite/patologia , Proteínas Wnt/fisiologia , Animais , Artrite Experimental/metabolismo , Artrite Experimental/fisiopatologia , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Diferenciação Celular/fisiologia , Polaridade Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Condrócitos/patologia , Condrócitos/fisiologia , Progressão da Doença , Humanos , Hipertrofia/prevenção & controle , Sistema de Sinalização das MAP Quinases/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/fisiologia , Camundongos Transgênicos , Osteoartrite/metabolismo , Osteoartrite/fisiopatologia , Proteína Relacionada ao Hormônio Paratireóideo/fisiologia , Proteínas Wnt/deficiência , Proteínas Wnt/metabolismo
13.
Biomaterials ; 182: 58-71, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30107270

RESUMO

Targeting delivery system has been widely used in packaging drugs for medical therapies attributed to its high efficiency and efficacy. A Traditional Chinese Medicine (TCM) formula consisting of Herba Epimedii has previously been shown to effectively treat postmenopausal osteoporosis. We have subsequently found that icaritin, which was a flavonoid isolated from both Herba Epimedii and its serum metabolites after oral administration, inhibited the adipogenic capacity of bone mesenchymal stem cells (BMSCs) while promoted their osteogenesis. However, previous pharmacokinetic analyses have shown that icaritin had a short half-life in blood and only trace amounts of the molecule reach the bone tissue. To overcome this limitation, we developed a bone-targeting liposome containing an oligopeptide of eight aspartate residues (Asp8), which had previously been shown to specifically target the bone, encapsulating icaritin. In vivo, we found that the Asp8-icaritin-liposome enhanced bone formation in ovariectomized mice compared to an icaritin-liposome control lacking the Asp8 moiety. Through in vitro mechanistic studies we further found that icaritin inhibited adipogenesis through an Akt/GSK-3ß/ß-catenin signaling pathway. Taken together, our study shows that Asp8-liposome as a bone-targeting delivery system is effective to carry an osteogenic phytomolecule for facilitating and enhancing its therapeutic effects on the prevention of estrogen depletion-induced osteoporosis.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Medicamentos de Ervas Chinesas/administração & dosagem , Flavonoides/administração & dosagem , Osteoporose/prevenção & controle , Adipogenia/efeitos dos fármacos , Animais , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Medicamentos de Ervas Chinesas/farmacocinética , Medicamentos de Ervas Chinesas/uso terapêutico , Feminino , Flavonoides/farmacocinética , Flavonoides/uso terapêutico , Lipossomos/metabolismo , Camundongos Endogâmicos C57BL , Oligopeptídeos/metabolismo , Osteogênese/efeitos dos fármacos , Osteoporose/metabolismo , Osteoporose/patologia
14.
Biomaterials ; 180: 173-183, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30041069

RESUMO

Magnesium (Mg) is a potential biomaterial suitable for developing biodegradable orthopaedic implants, especially as internal fixators for fracture fixation at non-load bearing skeletal sites. However, Mg alone cannot provide sufficient mechanical support for stable fracture fixation at load bearing sites due to its rapid degradation in the early stage after implantation. In consideration of the strengths and weaknesses of Mg, we developed an innovative magnesium/titanium (Mg/Ti) hybrid fixation system for long bone fracture fixation and investigated the fixation efficacy. The finite element analysis (FEA) results indicated that the Mg/Ti hybrid fixation system provided sufficient mechanical support for fracture fixation at load-bearing skeletal site. As a proof-of-concept, we performed a "Z-shaped" open osteotomy at the mid-shaft of rabbit tibia. For comparison, the animals were divided into two groups: Mg/Ti group (fixated with Mg screws and Ti fixators) and Ti control group (fixated with Ti screws and Ti fixators). The radiographic, four-point bending mechanical test, histological and histomorphometric analysis were postoperatively performed in a temporal manner up to 12 weeks. Both X-ray and micro-CT images of the Mg/Ti group showed a larger callus (14.7% at 3rd week and 24.8% at 6th week, n = 5-7, p < 0.05) in the regions of interest (ROIs) over time, especially at the opposite cortex of the fixation plate. At the 12th week post-operation, the biomechanical test result indicated that the rabbit tibia in the Mg/Ti group healed better and the overall mechanical strength was approximately 3-fold higher (n = 8, p < 0.05) than that at 6th week. Furthermore, the FEA revealed that the Mg/Ti group had a higher mechanical strength (19.5% at week 6 and 31.5% at week 12) at the specified ROI and resulted in an earlier and faster endochondral ossification (68.0% at week 3 and 71.4% at week 6) with a higher expression of osteocalcin (54.0%) and collagen I (34.2%) than the Ti control group (n = 4, p < 0.05). Further evaluation suggested that a higher expression of calcitonin gene-related peptide (CGRP), a known osteogenic neuron peptide, in the fracture callus of the Mg/Ti group might be a major underlying mechanism of enhanced fracture healing attributed to the release of Mg ions during the degradation of Mg screws.


Assuntos
Fixação Interna de Fraturas/métodos , Magnésio/química , Titânio/química , Animais , Placas Ósseas , Parafusos Ósseos , Peptídeo Relacionado com Gene de Calcitonina/química , Análise de Elementos Finitos , Consolidação da Fratura/efeitos dos fármacos , Consolidação da Fratura/fisiologia , Teste de Materiais , Osteocalcina/metabolismo , Coelhos , Suporte de Carga
15.
Nat Med ; 22(10): 1160-1169, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27571347

RESUMO

Orthopedic implants containing biodegradable magnesium have been used for fracture repair with considerable efficacy; however, the underlying mechanisms by which these implants improve fracture healing remain elusive. Here we show the formation of abundant new bone at peripheral cortical sites after intramedullary implantation of a pin containing ultrapure magnesium into the intact distal femur in rats. This response was accompanied by substantial increases of neuronal calcitonin gene-related polypeptide-α (CGRP) in both the peripheral cortex of the femur and the ipsilateral dorsal root ganglia (DRG). Surgical removal of the periosteum, capsaicin denervation of sensory nerves or knockdown in vivo of the CGRP-receptor-encoding genes Calcrl or Ramp1 substantially reversed the magnesium-induced osteogenesis that we observed in this model. Overexpression of these genes, however, enhanced magnesium-induced osteogenesis. We further found that an elevation of extracellular magnesium induces magnesium transporter 1 (MAGT1)-dependent and transient receptor potential cation channel, subfamily M, member 7 (TRPM7)-dependent magnesium entry, as well as an increase in intracellular adenosine triphosphate (ATP) and the accumulation of terminal synaptic vesicles in isolated rat DRG neurons. In isolated rat periosteum-derived stem cells, CGRP induces CALCRL- and RAMP1-dependent activation of cAMP-responsive element binding protein 1 (CREB1) and SP7 (also known as osterix), and thus enhances osteogenic differentiation of these stem cells. Furthermore, we have developed an innovative, magnesium-containing intramedullary nail that facilitates femur fracture repair in rats with ovariectomy-induced osteoporosis. Taken together, these findings reveal a previously undefined role of magnesium in promoting CGRP-mediated osteogenic differentiation, which suggests the therapeutic potential of this ion in orthopedics.


Assuntos
Pinos Ortopédicos , Peptídeo Relacionado com Gene de Calcitonina/efeitos dos fármacos , Fêmur/efeitos dos fármacos , Consolidação da Fratura/efeitos dos fármacos , Magnésio/farmacologia , Neurônios/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Proteína Semelhante a Receptor de Calcitonina/genética , Capsaicina/toxicidade , Proteínas de Transporte de Cátions/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Denervação , Feminino , Fraturas do Fêmur , Fixação Intramedular de Fraturas , Consolidação da Fratura/genética , Gânglios Espinais/citologia , Técnicas de Introdução de Genes , Técnicas de Silenciamento de Genes , Humanos , Neurônios/metabolismo , Osteogênese/genética , Osteoporose Pós-Menopausa , Fraturas por Osteoporose , Ovariectomia , Periósteo/citologia , Ratos , Proteína 1 Modificadora da Atividade de Receptores/genética , Fármacos do Sistema Sensorial/toxicidade , Células-Tronco , Canais de Cátion TRPM/metabolismo , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/metabolismo
16.
Sci Rep ; 5: 15632, 2015 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-26494536

RESUMO

We investigated the systemic effect of sclerostin monoclonal antibody (Scl-Ab) treatment on intact non-operated bones in an open osteotomy male Sprague Dawley (SD) rat model. Six-month-old male SD rats were subjected to transverse osteotomy at the right femur mid-shaft. Rats were injected subcutaneously with vehicle or Scl-Ab (25 mg/kg, 2 times per week) treatment for 9 weeks. Compared with vehicle control, Scl-Ab treatment significantly improved trabecular and cortical bone mass and microarchitecture at L5 vertebrae and left femora by micro-CT at week 6 and 9. Mechanical testing showed that Scl-Ab treatment resulted in significantly higher stiffness, energy to failure and ultimate load at the femora at week 9. Mineral apposition rate, mineralizing surface and bone formation rate on the trabecular bone in the distal femora was significantly increased in Scl-Ab group at week 6 and 9. The administered Scl-Ab was localized in the osteocytes and beta-catenin was strongly expressed in osteoblasts. Scl-Ab treatment significantly increased serum P1NP level and there was no between-group difference in serum level of CTX-1. In conclusion, Scl-Ab treatment could induce rapid and sustained increase in bone formation, bone mass and bone strength in non-operated bones. Sclerostin inhibition might be advantageous to prevent secondary fracture(s).


Assuntos
Anticorpos/administração & dosagem , Desenvolvimento Ósseo/imunologia , Proteínas Morfogenéticas Ósseas/imunologia , Marcadores Genéticos/imunologia , Animais , Biomarcadores/sangue , Remodelação Óssea , Masculino , Ratos , Ratos Sprague-Dawley
17.
J Bone Miner Res ; 30(11): 2044-57, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25917347

RESUMO

Vascular hyperpermeability and highly upregulated bone resorption in the destructive repair progress of steroid-associated osteonecrosis (SAON) are associated with a high expression of VEGF and high Src activity (Src is encoded by the cellular sarcoma [c-src] gene). This study was designed to prove our hypothesis that blocking the VEGF-Src signaling pathway by specific Src siRNA is able to prevent destructive repair in a SAON rabbit model. Destructive repair in SAON was induced in rabbits. At 2, 4, and 6 weeks after SAON induction, VEGF, anti-VEGF, Src siRNA, Src siRNA+VEGF, control siRNA, and saline were introduced via intramedullary injection into proximal femora for each group, respectively. Vascularization and permeability were quantified by dynamic contrast-enhanced (DCE) MRI. At week 6 after SAON induction, proximal femurs were dissected for micro-computed tomography (µCT)-based trabecular architecture with finite element analysis (FEA), µCT-based angiography, and histological analysis. Histological evaluation revealed that VEGF enhanced destructive repair, whereas anti-VEGF prevented destructive repair and Src siRNA and Src siRNA+VEGF prevented destructive repair and enhanced reparative osteogenesis. Findings of angiography and histomorphometry were consistent with those determined by DCE MRI. Src siRNA inhibited VEGF-mediated vascular hyperpermeability but preserved VEGF-induced neovascularization. Bone resorption was enhanced in the VEGF group and inhibited in the anti-VEGF, Src siRNA, Src siRNA+VEGF groups as determined by both 3D µCT and 2D histomorphometry. FEA showed higher estimated failure load in the Src siRNA and Src siRNA+VEGF groups when compared to the vehicle control group. Blockage of VEGF-Src signaling pathway by specific Src siRNA was able to prevent steroid-associated destructive repair while improving reconstructive repair in SAON, which might become a novel therapeutic strategy.


Assuntos
Osteonecrose/induzido quimicamente , Osteonecrose/enzimologia , RNA Interferente Pequeno/metabolismo , Esteroides/efeitos adversos , Cicatrização , Quinases da Família src/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Análise de Elementos Finitos , Técnicas de Silenciamento de Genes , Inativação Gênica , Masculino , Modelos Biológicos , Osteogênese , Osteonecrose/diagnóstico por imagem , Osteonecrose/patologia , Perfusão , Coelhos , Microtomografia por Raio-X , Quinases da Família src/metabolismo
18.
Am J Sports Med ; 43(6): 1413-21, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25825378

RESUMO

BACKGROUND: Surgical repair around the bone-tendon insertion (BTI) may involve bone-to-bone (BB), bone-to-tendon (BT), or tendon-to-tendon (TT) reattachment with varying healing outcome. HYPOTHESIS: The repair of Achilles tendon-calcaneus (ATC) by reattachment of homogeneous tissue (BB or TT) would heal faster, with respect to tensile properties at the healing complex, than those of reattachment of heterogeneous tissues (BT) over time. STUDY DESIGN: Controlled laboratory study. METHODS: Forty-seven adolescent male Chinese goats were divided into BB, BT, and TT groups. Osteotomy of the calcaneus, reattachment of Achilles tendon to the calcaneus after removal of the insertion, and tenotomy of the Achilles tendon were performed to simulate BB, BT, and TT repair, respectively. The ATC healing complexes were harvested at 6, 12, or 24 weeks postoperatively. Mechanical and morphological properties of the healing ATC complexes were assessed by tensile testing and qualitative histology, respectively. The contralateral intact ATC complex was used as the control. RESULTS: Failure load of BT was 33.4% lower than that of TT (P = .0243) at week 12. Ultimate strength of BT was 50.2% and 45.3% lower than that of TT at weeks 12 (P = .0002) and 24 (P = .0001), respectively. Tissue morphological characteristics of the BB and TT groups showed faster remodeling. The BT group showed limited regeneration of fibrocartilage zone and excessive formation of fibrous tissue at the healing interface. CONCLUSION: BTI repair between homogeneous tissues (BB and TT healing) showed better healing quality with respect to mechanical and histological assessments than did healing between heterogeneous tissues (BT healing). CLINICAL RELEVANCE: Anatomic reconstruction of ATC complex injury may be a primary concern when selecting the proper surgical approach. However, it is recommended to select fracture fixation (BB) or tendon repair (TT) instead of bone-tendon reattachment (BT) if possible to ensure better outcome at the healing interface.


Assuntos
Tendão do Calcâneo/fisiologia , Calcâneo/fisiologia , Traumatismos dos Tendões/fisiopatologia , Cicatrização/fisiologia , Tendão do Calcâneo/anatomia & histologia , Tendão do Calcâneo/lesões , Animais , Calcâneo/cirurgia , Fibrocartilagem/fisiologia , Fibrocartilagem/cirurgia , Cabras , Masculino , Modelos Animais , Procedimentos Ortopédicos/métodos , Osteotomia/métodos , Regeneração/fisiologia , Traumatismos dos Tendões/patologia , Traumatismos dos Tendões/cirurgia , Resistência à Tração/fisiologia
19.
J Orthop Res ; 32(8): 997-1005, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24782158

RESUMO

Sclerostin is a negative regulator of bone formation. Sclerostin monoclonal antibody (Scl-Ab) treatment promoted bone healing in various animal models. To further evaluate the healing efficiency of Scl-Ab in osteotomy healing, we investigated the time course effects of systemic administration of Scl-Ab on fracture repair in rat femoral osteotomy model. A total of 120 six-month-old male SD rats were subjected to transverse osteotomy at the right femur mid-shaft. Rats were treated with vehicle or Scl-Ab treatment for 3, 6, or 9 weeks. Fracture healing was evaluated by radiography, micro-CT, micro-CT based angiography, 4-point bending mechanical test and histological assessment. Scl-Ab treatment resulted in significantly higher total mineralized callus volume fraction, BMD and enhanced neovascularization. Histologically, Scl-Ab treatment resulted in a significant reduction in fracture callus cartilage at week 6 and increase in bone volume at week 9, associated with a greater proportion of newly formed bone area at week 6 and 9 by fluorescence microscopy. Mechanical testing showed significantly higher ultimate load in Scl-Ab treatment group at week 6 and 9. This study has demonstrated that Scl-Ab treatment enhanced bone healing in a rat femoral osteotomy model, as reflected in increased bone formation, bone mass and bone strength.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Proteínas Morfogenéticas Ósseas/imunologia , Fraturas do Fêmur/patologia , Consolidação da Fratura/efeitos dos fármacos , Marcadores Genéticos/imunologia , Animais , Calo Ósseo/efeitos dos fármacos , Fraturas do Fêmur/diagnóstico por imagem , Fêmur/efeitos dos fármacos , Fêmur/patologia , Masculino , Neovascularização Fisiológica/efeitos dos fármacos , Osteotomia , Ratos , Microtomografia por Raio-X
20.
J Orthop Res ; 32(4): 507-14, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24375544

RESUMO

Fibrous tissue is often formed in delayed healing of tendon bone insertion (TBI) instead of fibrocartilage. Extracorporeal shockwave (ESW) provides mechanical cues and upregulates expression of fibrocartilage-related makers and cytokines. We hypothesized that ESW would accelerate fibrocartilage regeneration at the healing interface in a delayed TBI healing model. Partial patellectomy with shielding at the TBI interface was performed on 32 female New Zealand White Rabbits for establishing this delayed TBI healing model. The rabbits were separated into the control and ESW group for evaluations at postoperative week 8 and 12. Shielding was removed at week 4 and a single ESW treatment was applied at week 6. Fibrocartilage regeneration was evaluated histomorphologically and immunohistochemically. Vickers hardness of the TBI matrix was measured by micro-indentation. ESW group showed higher fibrocartilage area, thickness, and proteoglycan deposition than the control in week 8 and 12. ESW increased expression of SOX9 and collagen II significantly in week 8 and 12, respectively. ESW group showed a gradual transition of hardness from bone to fibrocartilage to tendon, and had a higher Vickers hardness than the control group at week 12. In conclusion, ESW enhanced fibrocartilage regeneration at the healing interface in a delayed TBI healing model.


Assuntos
Enxerto Osso-Tendão Patelar-Osso , Fibrocartilagem/fisiologia , Regeneração , Som , Animais , Feminino , Coelhos , Distribuição Aleatória
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...