Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
FASEB J ; 37(6): e22950, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37144883

RESUMO

Fracture nonunion and bone defects are challenging for orthopedic surgeons. Milk fat globule-epidermal growth factor 8 (MFG-E8), a glycoprotein possibly secreted by macrophages in a fracture hematoma, participates in bone development. However, the role of MFG-E8 in the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) is unclear. We investigated the osteogenic effect of MFG-E8 in vitro and in vivo. The CCK-8 assay was used to assess the effect of recombinant human MFG-E8 (rhMFG-E8) on the viability of hBMSCs. Osteogenesis was investigated using RT-PCR, Western blotting, and immunofluorescence. Alkaline phosphatase (ALP) and Alizarin red staining were used to evaluate ALP activity and mineralization, respectively. An enzyme-linked immunosorbent assay was conducted to evaluate the secretory MFG-E8 concentration. Knockdown and overexpression of MFG-E8 in hBMSCs were established via siRNA and lentivirus vector transfection, respectively. Exogenous rhMFG-E8 was used to verify the in vivo therapeutic effect in a tibia bone defect model based on radiographic analysis and histological evaluation. Endogenous and secretory MFG-E8 levels increased significantly during the early osteogenic differentiation of hBMSCs. Knockdown of MFG-E8 inhibited the osteogenic differentiation of hBMSCs. Overexpression of MFG-E8 and rhMFG-E8 protein increased the expression of osteogenesis-related genes and proteins and enhanced calcium deposition. The active ß-catenin to total ß-catenin ratio and the p-GSK3ß protein level were increased by MFG-E8. The MFG-E8-induced enhanced osteogenic differentiation of hBMSCs was partially attenuated by a GSK3ß/ß-catenin signaling inhibitor. Recombinant MFG-E8 accelerated bone healing in a rat tibial-defect model. In conclusion, MFG-E8 promotes the osteogenic differentiation of hBMSCs by regulating the GSK3ß/ß-catenin signaling pathway and so, is a potential therapeutic target.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Humanos , Ratos , Animais , Osteogênese/fisiologia , beta Catenina/genética , beta Catenina/metabolismo , Fator VIII/metabolismo , Fator VIII/farmacologia , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Transdução de Sinais/fisiologia , Diferenciação Celular/fisiologia , Glicoproteínas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células Cultivadas , Via de Sinalização Wnt , Células da Medula Óssea/metabolismo
2.
J Orthop Res ; 41(8): 1821-1830, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36691867

RESUMO

The percutaneous sacroiliac (SI) screw is a common fixation option for posterior ring disruption in pelvic fractures. However, SI screw placement is difficult and can injure adjacent neurovascular structures. The sacral-alar-iliac screw (SAI) is a safe, reliable free-hand sacral pelvic fixation technique. To investigate the biomechanical stability of SAI for SI joint dislocation, finite element analysis was performed in unstable Tile-Type B and C pelvic ring injuries. The displacement in S1 (fixation of a unilateral S1 segment with one SI screw), TS1 (fixation of the S1 segment with a transsacra 1 screw), TS2 (fixation of the S2 segment with a transsacra 2 screw), S1AI, and S2AI exceeded the normal SI joint mobility. Sufficient stability after SI joint dislocation was obtained with (TS1 + TS2), (TS2 + S1), (S1AI + S2AI + rod), (S1AI + S2AI), and (S1 + S2AI + S1 pedicle) fixation. The TS1 + TS2 group had the smallest displacement and lowest peak screw stress, followed by (S1 + S2AI + S1 pedicle) placement. Our findings suggest that SAI screws are a valuable option for SI joint dislocation.


Assuntos
Fraturas Ósseas , Luxações Articulares , Fusão Vertebral , Humanos , Análise de Elementos Finitos , Parafusos Ósseos , Fraturas Ósseas/cirurgia , Fixação Interna de Fraturas/métodos , Luxações Articulares/cirurgia , Ílio/cirurgia , Sacro/cirurgia , Sacro/lesões , Articulação Sacroilíaca/cirurgia , Fusão Vertebral/métodos
3.
Bioact Mater ; 19: 429-443, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35574058

RESUMO

The imbalance of bone homeostasis is the root cause of osteoporosis. However current therapeutic approaches mainly focus on either anabolic or catabolic pathways, which often fail to turn the imbalanced bone metabolism around. Herein we reported that a SIRT-1 agonist mediated molecular therapeutic strategy to reverse the imbalance in bone homeostasis by simultaneously regulating osteogenesis and osteoclastogenesis via locally sustained release of SRT2104 from mineral coated acellular matrix microparticles. Immobilization of SRT2104 on mineral coating (MAM/SRT) harnessing their electrostatic interactions resulted in sustained release of SIRT-1 agonist for over 30 days. MAM/SRT not only enhanced osteogenic differentiation and mineralization, but also attenuated the formation and function of excessive osteoclasts via integrating multiple vital upstream signals (ß-catenin, FoxOs, Runx2, NFATc1, etc.) in vitro. Osteoporosis animal model also validated that it accelerated osteoporotic bone healing and improved osseointegration of the surrounding bone. Overall, our work proposes a promising strategy to treat osteoporotic bone defects by reversing the imbalance in bone homeostasis using designated small molecule drug delivery systems.

4.
Biomolecules ; 12(11)2022 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-36358952

RESUMO

Osteoporosis and osteoporotic fractures comprise a substantial health and socioeconomic burden. The leading cause of osteoporosis is an imbalance in bone formation and bone resorption caused by hyperactive osteoclasts. Therefore, a new strategy to suppress osteoclastogenesis is needed. Parkin is likely closely associated with bone metabolism, although its role in osteoclastogenesis is unclear. In this study, the Parkin protein inhibited the receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation, osteoclast-specific gene expression, F-actin ring formation, and bone resorption pit formation in vitro. Moreover, depletion of Parkin enhanced RANKL-induced osteoclast formation, osteoclast-specific gene expression, F-actin ring formation, and bone resorption pit formation. Reactive oxygen species (ROS) activity was suppressed, while autophagy was upregulated with the presence of the Parkin protein. ROS activity was upregulated and autophagy was decreased due to Parkin knockdown. In addition, intravenous administration of Parkin rescued ovariectomy-induced bone loss and reduced osteoclastogenesis in vivo. Collectively, Parkin has therapeutic potential for diseases associated with overactive osteoclasts.


Assuntos
Reabsorção Óssea , Osteoporose , Humanos , Feminino , Animais , Camundongos , Ligante RANK/farmacologia , Osteogênese , Espécies Reativas de Oxigênio/metabolismo , Actinas/metabolismo , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/genética , Ovariectomia/efeitos adversos , Ubiquitina-Proteína Ligases/genética , Osteoporose/tratamento farmacológico , Osteoporose/etiologia , Diferenciação Celular , NF-kappa B/metabolismo , Camundongos Endogâmicos C57BL
5.
Cell Mol Life Sci ; 79(6): 328, 2022 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-35639207

RESUMO

The osteogenic potential of bone marrow mesenchymal stem cells (BMSCs) is critical for bone formation and regeneration. A high non-/delayed-union rate of fracture healing still occurs in specific populations, implying an urgent need to discover novel targets for promoting osteogenesis and bone regeneration. Long non-coding (lnc)RNAs are emerging regulators of multiple physiological processes, including osteogenesis. Based on differential expression analysis of RNA sequencing data, we found that lncRNA AC132217.4, a 3'UTR-overlapping lncRNA of insulin growth factor 2 (IGF2), was highly induced during osteogenic differentiation of BMSCs. Afterward, both gain-of-function and loss-of-function experiments proved that AC132217.4 promotes osteoblast development from BMSCs. As for its molecular mechanism, we found that AC132217.4 binds with IGF2 mRNA to regulate its expression and downstream AKT activation to control osteoblast maturation and function. Furthermore, we identified two splicing factors, splicing component 35 KDa (SC35) and heterogeneous nuclear ribonucleoprotein A1 (HNRNPA1), which regulate the biogenesis of AC132217.4 at the post-transcriptional level. We also identified a transcription factor, ALX1, which regulates AC132217.7 expression at the transcriptional level to promote osteogenesis. Importantly, in-vivo over-expression of AC132217.4 essentially promotes the bone healing process in a murine tibial drill-hole model. Our study demonstrates that lncRNA AC132217.4 is a novel anabolic regulator of BMSC osteogenesis and could be a plausible therapeutic target for improving bone regeneration.


Assuntos
Proteínas de Homeodomínio , Células-Tronco Mesenquimais , Osteogênese , RNA Longo não Codificante , Animais , Diferenciação Celular/genética , Proteínas de Homeodomínio/genética , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Camundongos , Osteogênese/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Transdução de Sinais
6.
Orthop J Sports Med ; 9(12): 23259671211027838, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34938818

RESUMO

BACKGROUND: Arthroscopy-assisted reduction percutaneous internal fixation (ARIF) has emerged recently as an alternative treatment method in treating lower-energy tibial plateau fractures. To date, the comparison of clinical efficacy between ARIF and open reduction internal fixation (ORIF) is limited, with divergent conclusions. PURPOSE: To review studies on the clinical efficacy of ARIF and ORIF in the treatment of tibial plateau fracture. STUDY DESIGN: Systematic review; Level of evidence, 3. METHODS: A search was conducted using the PubMed, Web of Science, Cochrane Library, and EMBASE databases between inception and August 20, 2020, for retrospective and prospective studies evaluating ARIF versus ORIF in the treatment of tibial plateau fracture. We identified 6 clinical studies that met the inclusion criteria, with 231 patients treated with ARIF and 386 patients treated with ORIF. The risk of bias and the quality of evidence of the included studies were assessed. The 2 treatment types were compared in terms of clinical results and complications by using odds ratios (ORs), mean differences (MDs), or standardized mean differences (SMDs), with 95% confidence intervals (CIs). Heterogeneity among studies was quantified using the I 2 statistic. RESULTS: The quality of the studies was high. Compared with ORIF, treatment with ARIF led to better clinical function (SMD = 0.31; 95% CI, 0.14 to 0.48; I 2 = 15%; P = .0005), shorter hospital stay (MD = -2.37; 95% CI, -2.92 to -1.81; I 2 = 0%; P < .001), and more intra-articular lesions found intraoperatively (OR = 3.76; 95% CI, 1.49 to 9.49; I 2 = 66%; P = .005). There were no complications or significant differences between the techniques in the radiological evaluation of reduction. CONCLUSION: Compared with ORIF, the ARIF technique for tibial plateau fractures led to faster postoperative recovery and better clinical function and the ability to find and treat more intra-articular lesions during the operation. However, the radiological evaluation of reduction and complications were not significantly different between the 2 groups.

7.
Stem Cell Res Ther ; 12(1): 268, 2021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33947456

RESUMO

BACKGROUND: Inflammatory microenvironment is significant to the differentiation and function of mesenchymal stem cells (MSCs). It evidentially influences the osteoblastogenesis of MSCs. IL-34, a newly discovered cytokine, playing a key role in metabolism. However, the research on its functional role in the osteogenesis of MSCs was rarely reported. Here, we described the regulatory effects of low-dose IL-34 on both osteoblastogenesis and osteoclastogenesis. METHODS: We performed the osteogenic effects of hBMSCs by exogenous and overexpressed IL-34 in vitro, so were the osteoclastogenesis effects of mBMMs by extracellular IL-34. CCK-8 was used to assess the effect of IL-34 on the viability of hBMSCs and mBMMs. ALP, ARS, and TRAP staining was used to evaluate ALP activity, mineral deposition, and osteoclastogenesis, respectively. qRT-PCR and Western blotting analysis were performed to detect the expression of target genes and proteins. ELISA was used to evaluate the concentrations of IL-34. In vivo, a rat tibial osteotomy model and an OVX model were established. Radiographic analysis and histological evaluation were performed to confirm the therapeutic effects of IL-34 in fracture healing and osteoporosis. Statistical differences were evaluated by two-tailed Student's t test, one-way ANOVA with Bonferroni's post hoc test, and two-way ANOVA with Bonferroni multiple comparisons post hoc test in the comparison of 2 groups, more than 2 groups, and different time points of treated groups, respectively. RESULTS: Promoted osteoblastogenesis of hBMSCs was observed after treated by exogenous or overexpressed IL-34 in vitro, confirmed by increased mineral deposits and ALP activity. Furthermore, exogenous or overexpressed IL-34 enhanced the expression of p-AKT and p-ERK. The specific AKT and ERK signaling pathway inhibitors suppressed the enhancement of osteoblastogenesis induced by IL-34. In a rat tibial osteotomy model, imaging and histological analyses testified the local injection of exogenous IL-34 improved bone healing. However, the additional IL-34 has no influence on both osteoclastogenesis of mBMMs in vitro and osteoporosis of OVX model of rat in vivo. CONCLUSIONS: Collectively, our study demonstrate that low-dose IL-34 regulates osteogenesis of hBMSCs partly via the PIK/AKT and ERK signaling pathway and enhances fracture healing, with neither promoting nor preventing osteoclastogenesis in vitro and osteoporosis in vivo.


Assuntos
Osteogênese , Proteínas Proto-Oncogênicas c-akt , Animais , Diferenciação Celular , Células Cultivadas , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais
8.
Front Cell Dev Biol ; 8: 576104, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33043010

RESUMO

Osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) plays a key role in bone formation. Parkin, an E3 ubiquitin ligase, related to Parkinson's disease and aging. Previous studies have indicated that Parkinson's disease have a higher risk of osteoporotic fracture. To investigate the effects and underlying mechanism of Parkin in the osteogenic differentiation of BMSCs, osteogenic differentiation was analyzed following upregulation or downregulation of Parkin. We found that Parkin was increased during differentiation. Parkin overexpression enhanced osteo-specific markers, and downregulation of Parkin mitigated osteo-specific markers. Moreover, upregulation of Parkin promoted ß-catenin expression and autophagy and vice versa. The upregulation of ß-catenin enhanced autophagy, and the activation of autophagy also increased the expression of ß-catenin in Parkin-downregulated BMSCs. Parkin-overexpressed cell sheets accelerated bone healing in a tibial fracture model. Based on these results, we concluded that Parkin meditates osteoblastic differentiation of BMSCs via ß-catenin and autophagy signaling.

9.
Cell Tissue Res ; 382(3): 457-462, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32803322

RESUMO

Mesenchymal stem cells (MSCs) hold enormous potential for the treatment of immune-related conditions and degenerative diseases, owing to their self-renewal and multilineage differentiation capabilities. Nevertheless, cellular senescence significantly impacts the quantity and quality of MSCs, limiting their clinical use. Mitochondria play essential roles in energy production by oxidative phosphorylation and metabolism of energy sources via the tricarboxylic acid cycle. Therefore, mitochondrial dysfunction is a primary cause of senescence in MSCs. Herein, we summarize the current knowledge regarding the mechanisms underlying mitochondrial dysfunction-associated cellular senescence. We also discuss potential methods to prevent or even reverse MSC senescence.


Assuntos
Senescência Celular/fisiologia , Células-Tronco Mesenquimais/metabolismo , Mitocôndrias/metabolismo , Animais , Diferenciação Celular , Humanos , Camundongos
10.
World J Stem Cells ; 12(5): 339-350, 2020 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-32547682

RESUMO

The postnatal skeleton undergoes growth, modeling, and remodeling. The human skeleton is a composite of diverse tissue types, including bone, cartilage, fat, fibroblasts, nerves, blood vessels, and hematopoietic cells. Fracture nonunion and bone defects are among the most challenging clinical problems in orthopedic trauma. The incidence of nonunion or bone defects following fractures is increasing. Stem and progenitor cells mediate homeostasis and regeneration in postnatal tissue, including bone tissue. As multipotent stem cells, skeletal stem cells (SSCs) have a strong effect on the growth, differentiation, and repair of bone regeneration. In recent years, a number of important studies have characterized the hierarchy, differential potential, and bone formation of SSCs. Here, we describe studies on and applications of SSCs and/or mesenchymal stem cells for bone regeneration.

11.
Cell Prolif ; 53(2): e12752, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31889368

RESUMO

OBJECTIVES: Insulin-like growth factor-binding protein 7 (IGFBP7) is a low-affinity insulin growth factor (IGF) binder that may play an important role in bone metabolism. We previously reported that IGFBP7 enhanced osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) via the Wnt/ß-catenin signalling pathway. In this study, we tried to reveal its function in osteoclast differentiation and osteoporosis. METHODS: We used both in vitro and in vivo studies to investigate the effects of IGFBP7 on RANKL-induced osteoclastogenesis and osteoporosis, together with the underlying molecular mechanisms of these processes. RESULTS: We show that IGFBP7 inhibited receptor activation of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclastogenesis, F-actin ring formation and bone resorption, which was confirmed by using recombinant IGFBP7 protein, lentivirus and siRNA. The NF-κB signalling pathway was inhibited during this process. Moreover, in a mouse ovariectomy-induced osteoporosis model, IGFBP7 treatment attenuated osteoporotic bone loss by inhibiting osteoclast activity. CONCLUSIONS: Taken together, these findings show that IGFBP7 suppressed osteoclastogenesis in vitro and in vivo and suggest that IGFBP7 is a negative regulator of osteoclastogenesis and plays a protective role in osteoporosis. These novel insights into IGFBP7 may facilitate the development of potential treatment strategies for oestrogen deficiency-induced osteoporosis and other osteoclast-related disorders.


Assuntos
Reabsorção Óssea/metabolismo , Estrogênios/metabolismo , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Osteoclastos/metabolismo , Osteogênese/fisiologia , Ligante RANK/metabolismo , Animais , Células da Medula Óssea/metabolismo , Diferenciação Celular/fisiologia , Feminino , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Osteoblastos/metabolismo , Osteoporose/metabolismo
12.
Cell Death Dis ; 10(10): 753, 2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31582734

RESUMO

Interleukin (IL)-37, a pivotal anti-inflammatory cytokine and a fundamental inhibitor of innate immunity, has recently been shown to be abnormally expressed in several autoimmune-related orthopedic diseases, including rheumatoid arthritis, ankylosing spondylitis, and osteoporosis. However, the role of IL-37 during osteogenic differentiation of mesenchymal stem cells (MSCs) remains largely unknown. In this study, extracellular IL-37 significantly increased osteoblast-specific gene expression, the number of mineral deposits, and alkaline phosphatase activity of MSCs. Moreover, a signaling pathway was activated in the presence of IL-37. The enhanced osteogenic differentiation of MSCs due to supplementation of IL-37 was partially rescued by the presence of a PI3K/AKT signaling inhibitor. Using a rat calvarial bone defect model, IL-37 significantly improved bone healing. Collectively, these findings indicate that extracellular IL-37 enhanced osteogenesis of MSCs, at least in part by activation of the PI3K/AKT signaling pathway.


Assuntos
Diferenciação Celular , Espaço Extracelular/metabolismo , Interleucina-1/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Cálcio/metabolismo , Morte Celular/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Imageamento Tridimensional , Masculino , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogênese/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais , Crânio/diagnóstico por imagem , Crânio/patologia , Cicatrização
13.
Stem Cell Res Ther ; 10(1): 189, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31238979

RESUMO

BACKGROUND: Management of fracture healing with a large bone defect remains a tricky subject in orthopedic trauma. Enhancing osteogenesis of human bone marrow-derived mesenchymal stem cells (hBMSCs) is one of the useful therapeutic strategies for fracture healing. Previous studies have revealed that Apelin may play an important role in bone metabolism. However, its function in the osteogenesis of hBMSCs remains unclear. Therefore, in this study, we investigated the effects and mechanism of Apelin on osteogenic differentiation. METHODS: We investigated the osteogenesis effects of hBMSCs by both exogenous Apelin protein and overexpression Apelin in vitro. Cell proliferation assay was used to assess the effect of Apelin on the proliferation of hBMSCs. ALP staining and Alizarin Red staining were used to evaluate ALP activity and mineral deposition respectively. qPCR and Western blotting analysis were used to detect the expression of target genes and proteins. In vivo, a rat tibial osteotomy model was established; radiographic analysis and histological evaluation were used to confirm the therapeutic effects of Apelin in fracture healing. Statistical significance was determined by two-tailed Student's t test when 2 groups were compared. When more than 2 groups were compared, one-way ANOVA followed by Bonferroni's post-hoc test was used. And two-way ANOVA, followed by Bonferroni multiple comparisons post-hoc test, was performed when the treatment groups at different time points were compared. RESULTS: The addition of exogenous Apelin protein or overexpression of Apelin promoted osteoblast differentiation of hBMSCs in vitro. Increased mineral deposits were observed after treatment with extracellular Apelin protein or after the upregulation of Apelin. Moreover, ß-catenin levels were upregulated by Apelin. The enhancement of osteogenic differentiation induced by Apelin was attenuated by specific Wnt/ß-catenin signaling pathway inhibitors. In a rat tibial osteotomy model, local injection of exogenous Apelin protein improved bone healing, as demonstrated by imaging and histological analyses. CONCLUSIONS: Taken together, these findings indicate that Apelin regulates osteogenic differentiation of hMSCs partly via the Wnt/ß-catenin signaling pathway and effectively promotes fracture healing.


Assuntos
Adipócitos/metabolismo , Células da Medula Óssea/citologia , Condrócitos/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Adipócitos/citologia , Animais , Apelina/genética , Apelina/metabolismo , Western Blotting , Cálcio/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células/genética , Proliferação de Células/fisiologia , Condrócitos/citologia , Imunofluorescência , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Osteoblastos/citologia , Osteogênese/genética , Osteogênese/fisiologia , Ratos , Ratos Sprague-Dawley
14.
Int J Nanomedicine ; 13: 5799-5810, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30310282

RESUMO

BACKGROUND: The osteo-immunomodulatory properties of biomaterials play an important role in the outcomes of bone regeneration. Graphene oxide (GO) has been widely applied in many research fields due to its unique properties. However, the immunomodulatory properties of GO as a biomaterial for bone tissue engineering are still unclear. MATERIALS AND METHODS: In this study, we evaluated the Inflammatory response of RAW264.7 cells influenced by GO. Then the osteogenic differentiation of BMSCs, and angiogenic differentiation of human umbilical vein endothelial cells (HUVECs) by stimulation with GO/RAW 264.7-conditioned culture medium were accessed. We also further investi gated the possible mechanisms underlying the osteo- and angio-immunomodulatory effects of GO. RESULTS: Our results showed that GO stimulates the secretion of oncostatin M, tumor necrosis factor alpha and other factors through the nuclear factor-κB pathway. GO/RAW264.7-conditioned medium promoted the osteogenic differentiation of BMSCs, stimulated upregulation of the HUVECs of vascular-related receptors, and promoted their tube formation in vitro. CONCLUSION: In conclusion, our research shows that GO, as a biomaterial, can induce the formation of a beneficial osteo-immunomodulatory environment and is a promising biomaterial for bone tissue engineering.


Assuntos
Grafite/farmacologia , Fatores Imunológicos/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Meios de Cultivo Condicionados/farmacologia , Endocitose/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Imunomodulação/efeitos dos fármacos , Inflamação/genética , Inflamação/patologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/ultraestrutura , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Neovascularização Fisiológica/genética , Células RAW 264.7 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos
15.
Cell Stress Chaperones ; 23(6): 1153-1164, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30187197

RESUMO

Heat shock proteins (HSPs) are a family of proteins produced by cells in response to exposure to stressful conditions. In addition to their role as chaperones, they also play an important role in the cardiovascular, immune, and other systems. Normal bone tissue is maintained by bone metabolism, particularly by the balance between osteoblasts and osteoclasts, which are physiologically regulated by multiple hormones and cytokines. In recent years, studies have reported the vital role of HSPs in bone metabolism. However, the conclusions remain largely controversial, and the exact mechanisms are still unclear, so a review and analyses of previous studies are of importance. This article reviews the current understanding of the roles and effects of HSPs on bone cells (osteoblasts, osteoclasts, and osteocytes), in relation to bone metabolism.


Assuntos
Osso e Ossos/citologia , Osso e Ossos/metabolismo , Proteínas de Choque Térmico/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteócitos/metabolismo , Animais , Citocinese/fisiologia , Hormônios/metabolismo , Humanos , Camundongos , Osteoblastos/citologia , Osteoclastos/citologia , Osteócitos/citologia , Ratos
16.
BMC Musculoskelet Disord ; 19(1): 288, 2018 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-30111311

RESUMO

BACKGROUND: The use of bone graft for the radial head fractures has been previously described and occasionally used by other authors.This is the first paper, to my knowledge, dealing with the relevant issue about the importance that the use of an autologous bone graft can have on the radial head fractures. METHODS: From July 2010 to July 2014, 20 consecutive patients who underwent open reduction and internal fixation for a closed Mason type II radial head fracture were retrospectively reviewed. Patients with Mason type I, III, simple type II, and comminuted type II fractures treated without bone grafting were excluded. A clinical examination and radiographic evaluation were performed. The overall functional result was evaluated using the Mayo Elbow Performance Score (MEPS). The Broberg and Morrey classification was used to evaluate traumatic arthritis. RESULTS: The average follow-up duration was 31 months (range, 24-50 months). Bone union of the radial head fracture was achieved in all patients at an average of 13.5 weeks (range, 12-17 weeks). Postoperative radiographs showed no cases of postsurgical ligamentous instability, necrosis of the radial head, or internal fixation failure. The mean range of motion of the affected elbow was 128° ± 8.4° in flexion, 14.5° ± 11.1° in extension, 68.7° ± 14.1° in pronation, and 65.2° ± 18.2° in supination. The mean MEPS was 92 ± 7.9 points (range, 80-100); the outcome was excellent (90-100 points) in 13 patients and good (75-89 points) in 7 patients. The MEPS tended to be higher in patients with an isolated fracture (p = 0.016). Based on the Broberg and Morrey classification for radiographic assessment of post-traumatic arthritis, 15 elbows had no evidence of degenerative changes (grade 0), and 5 elbows had grade 1 changes. CONCLUSION: Although radial head fractures may not be amenable to internal fixation, our findings suggest that open reduction and internal fixation with an autogenous bone graft from the lateral epicondyle of the humerus provides satisfactory elbow function in patients with comminuted Mason type II radial head fractures.


Assuntos
Transplante Ósseo/métodos , Fixação Interna de Fraturas , Fraturas Cominutivas/cirurgia , Úmero/transplante , Redução Aberta , Fraturas do Rádio/cirurgia , Rádio (Anatomia)/cirurgia , Adulto , Idoso , Transplante Ósseo/efeitos adversos , Feminino , Fixação Interna de Fraturas/efeitos adversos , Consolidação da Fratura , Fraturas Cominutivas/diagnóstico por imagem , Fraturas Cominutivas/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Redução Aberta/efeitos adversos , Rádio (Anatomia)/diagnóstico por imagem , Rádio (Anatomia)/fisiopatologia , Fraturas do Rádio/diagnóstico por imagem , Fraturas do Rádio/fisiopatologia , Recuperação de Função Fisiológica , Estudos Retrospectivos , Fatores de Tempo , Transplante Autólogo , Resultado do Tratamento , Adulto Jovem
17.
Cell Death Dis ; 9(8): 836, 2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-30082727

RESUMO

Forkhead box protein A2 (FOXA2) is a core transcription factor that controls cell differentiation and may have an important role in bone metabolism. However, the role of FOXA2 during osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) remains largely unknown. In this study, decreased expression of FOXA2 was observed during osteogenic differentiation of rat BMSCs (rBMSCs). FOXA2 knockdown significantly increased osteoblast-specific gene expression, the number of mineral deposits and alkaline phosphatase activity, whereas FOXA2 overexpression inhibited osteogenesis-specific activities. Moreover, extracellular signal-regulated protein kinase (ERK) signalling was upregulated following knockdown of FOXA2. The enhanced osteogenesis due to FOXA2 knockdown was partially rescued by an ERK inhibitor. Using a rat tibial defect model, a rBMSC sheet containing knocked down FOXA2 significantly improved bone healing. Collectively, these findings indicated that FOXA2 had an essential role in osteogenic differentiation of BMSCs, partly by activation of the ERK signalling pathway.


Assuntos
Diferenciação Celular , Fator 3-beta Nuclear de Hepatócito/metabolismo , Sistema de Sinalização das MAP Quinases , Animais , Células da Medula Óssea/citologia , Butadienos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fraturas Ósseas/patologia , Fraturas Ósseas/terapia , Fraturas Ósseas/veterinária , Fator 3-beta Nuclear de Hepatócito/antagonistas & inibidores , Fator 3-beta Nuclear de Hepatócito/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Nitrilas/farmacologia , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Alicerces Teciduais/química
18.
Int Immunopharmacol ; 61: 222-230, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29890416

RESUMO

The overproduction of MMPs (matrix metalloproteinases) and members of the ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) protein family plays an important role in the pathogenesis of osteoarthritis (OA). The potential of selective MMPs or ADAMTS inhibitors as chemopreventive agents for OA has been demonstrated in several studies. In this study, we investigated the protective effects of emodin (1,3,8-trihydroxy-6-methylanthaquinone), isolated from the root of Rheum palmatum L., in the inhibition of MMP and ADAMTS expression in both rat chondrocytes and an animal model of OA. The expression of MMP-3, MMP-13, ADAMTS-4, ADAMTS-5, aggrecan, and collagen II mRNA and protein in interleukin-1beta (IL-1ß)-induced rat chondrocytes was followed by quantitative real-time PCR and western blot. The activation of the NF-κB and Wnt/ß-catenin pathways by IL-1ß was assessed by western blot. The in vivo effects of emodin were evaluated by intra-articular injection in rats in an experimental model of OA induced by anterior cruciate ligament transection. Emodin dose-dependently down-regulated the expression of MMP-3, MMP-13, ADAMTS-4 and ADAMTS-5 at both the mRNA and protein level in IL-1ß-stimulated rat chondrocytes. In addition, the IL-1ß-induced activation of NF-κB and Wnt signals was attenuated by emodin, as determined by western blotting. The intra-articular injection of emodin in a rat OA model ameliorated OA progression, as determined in morphological and histological analyses in vivo. Taken together, our findings demonstrate that emodin is a promising therapeutic agent for the prevention and treatment of OA.


Assuntos
Cartilagem Articular/patologia , Condrócitos/efeitos dos fármacos , Emodina/uso terapêutico , Osteoartrite/tratamento farmacológico , Proteína ADAMTS4/genética , Proteína ADAMTS4/metabolismo , Proteína ADAMTS5/genética , Proteína ADAMTS5/metabolismo , Animais , Ligamento Cruzado Anterior/cirurgia , Células Cultivadas , Condrócitos/fisiologia , Modelos Animais de Doenças , Humanos , Masculino , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/genética , Metaloproteinase 3 da Matriz/metabolismo , NF-kappa B/metabolismo , Raízes de Plantas , Ratos , Ratos Sprague-Dawley , Rheum/imunologia , Transdução de Sinais
19.
Acta Biomater ; 73: 377-387, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29678676

RESUMO

The poor healing capacity and typically incomplete regeneration of injured tendons has made tendon repair as a primary clinical concern. Several methods for repairing injured tendons have been developed in the last decade. Tendon regeneration using current tissue engineering techniques requires advanced biomaterials to satisfy both microstructural and mechanical criteria. In this study, a novel chitosan (CS)-based scaffold with asymmetric structure was fabricated using a self-deposition technique. The fabricated scaffolds were assessed with regard to the microstructural and mechanical demands of cell ingrowth and the prevention of peritendinous adhesion. In vitro studies showed that rat tendon stem/progenitor cells (TSPCs) seeded onto the CS scaffold displayed higher levels of tenogenic specific genes expression and protein production. Four and six weeks after the implantation of CS scaffolds on full-site Achilles tendon defects, in vivo tendon repair was evaluated by histology, immunohistochemistry, immunofluorescence, and mechanical measurements. The production of collagen I (COL1) and collagen III (COL3) demonstrated that the CS scaffolds were capable of inducing conspicuous tenogenic differentiation, higher tenomodulin (TNMD) production, and superior phenotypic maturity, compared with the empty defect group. The introduction of TSPCs into the CS scaffold resulted in a synergistic effect on tendon regeneration and yielded better-aligned collagen fibers with elongated, spindle-shaped cells. These findings indicated that the application of TSPC-seeded CS scaffolds would be a feasible approach for tendon repair. STATEMENT OF SIGNIFICANCE: The poor healing capacity of injured tendons and inevitable peritendinous adhesion has made tendon regeneration a clinical priority. In this study, an asymmetric chitosan scaffold was developed to encapsulate rat tendon stem/progenitor cells (TSPCs), which could induce higher levels of tenogenic specific genes and protein expression. Remarkably, the introduction of TSPCs into the asymmetric chitosan scaffold generated a synergistic effect on in vivo tendon regeneration and lead to better-aligned collagen fibers compared with asymmetric chitosan scaffold alone. This work can provide new guidelines for the structure and property design of cell-seeded scaffolds for tendon regeneration.


Assuntos
Quitosana/química , Regeneração , Células-Tronco/citologia , Tendões/patologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Tendão do Calcâneo/citologia , Animais , Materiais Biocompatíveis , Adesão Celular , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Colágeno/química , Hidrogéis/química , Masculino , Microscopia de Fluorescência , Ratos , Ratos Sprague-Dawley , Estresse Mecânico , Resistência à Tração , Cicatrização
20.
FASEB J ; 32(9): 4917-4929, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29630408

RESUMO

Microenvironmental conditions can influence the differentiation and functional roles of mesenchymal stem cells (MSCs). Recent studies have suggested that an inflammatory microenvironment can significantly affect the osteogenic differentiation of MSCs. Here, we show, for the first time, that IL-10 has concentration-dependent, dual roles in the osteogenesis of human bone marrow mesenchymal stem cells (hBMSCs). Low physiologic concentrations of IL-10 (0.01-1.0 ng/ml) activate the p38/MAPK signaling pathway to promote the osteogenesis of hBMSCs, but higher pathologic doses of IL-10 (10-100 ng/ml) inhibit p38/MAPK signaling by activating NF-κB, inhibiting osteogenesis. These results demonstrate that p38/MAPK and NF-κB signaling mediates the double-edged sword effect of IL-10 on hBMSCs. The osteogenic impairment was reversed at higher doses of IL-10 when cells were supplemented with the NF-κB inhibitor BAY11-7082. These data provide important insights into the regulatory effects of IL-10 on the biologic behavior of hBMSCs.-Chen, E., Liu, G., Zhou, X., Zhang, W., Wang, C., Hu, D., Xue, D., Pan, Z. Concentration-dependent, dual roles of IL-10 in the osteogenesis of human BMSCs via P38/MAPK and NF-κB signaling pathways.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Interleucina-10/farmacologia , Osteogênese/efeitos dos fármacos , Células da Medula Óssea/citologia , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA