Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 289
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 38(14): e23810, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39042586

RESUMEN

Osteofibrous dysplasia (OFD) is a rare, benign, fibro-osseous lesion that occurs most commonly in the tibia of children. Tibial involvement leads to bowing and predisposes to the development of a fracture which exhibit significantly delayed healing processes, leading to prolonged morbidity. We previously identified gain-of-function mutations in the MET gene as a cause for OFD. In our present study, we test the hypothesis that gain-of-function MET mutations impair bone repair due to reduced osteoblast differentiation. A heterozygous Met exon 15 skipping (MetΔ15-HET) mouse was created to imitate the human OFD mutation. The mutation results in aberrant and dysregulation of MET-related signaling determined by RNA-seq in the murine osteoblasts extracted from the wide-type and genetic mice. Although no gross skeletal defects were identified in the mice, fracture repair was delayed in MetΔ15-HET mice, with decreased bone formation observed 2-week postfracture. Our data are consistent with a novel role for MET-mediated signaling regulating osteogenesis.


Asunto(s)
Enfermedades del Desarrollo Óseo , Modelos Animales de Enfermedad , Displasia Fibrosa Ósea , Curación de Fractura , Osteogénesis , Proteínas Proto-Oncogénicas c-met , Animales , Ratones , Osteogénesis/genética , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Curación de Fractura/genética , Enfermedades del Desarrollo Óseo/genética , Enfermedades del Desarrollo Óseo/patología , Humanos , Displasia Fibrosa Ósea/genética , Displasia Fibrosa Ósea/patología , Displasia Fibrosa Ósea/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patología , Mutación , Diferenciación Celular , Ratones Endogámicos C57BL , Masculino
2.
J Cell Mol Med ; 28(13): e18522, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38957040

RESUMEN

Bone non-union is a common fracture complication that can severely impact patient outcomes, yet its mechanism is not fully understood. This study used differential analysis and weighted co-expression network analysis (WGCNA) to identify susceptibility modules and hub genes associated with fracture healing. Two datasets, GSE125289 and GSE213891, were downloaded from the GEO website, and differentially expressed miRNAs and genes were analysed and used to construct the WGCNA network. Gene ontology (GO) analysis of the differentially expressed genes showed enrichment in cytokine and inflammatory factor secretion, phagocytosis, and trans-Golgi network regulation pathways. Using bioinformatic site prediction and crossover gene search, miR-29b-3p was identified as a regulator of LIN7A expression that may negatively affect fracture healing. Potential miRNA-mRNA interactions in the bone non-union mechanism were explored, and miRNA-29-3p and LIN7A were identified as biomarkers of skeletal non-union. The expression of miRNA-29b-3p and LIN7A was verified in blood samples from patients with fracture non-union using qRT-PCR and ELISA. Overall, this study identified characteristic modules and key genes associated with fracture non-union and provided insight into its molecular mechanisms. Downregulated miRNA-29b-3p was found to downregulate LIN7A protein expression, which may affect the healing process after fracture in patients with bone non-union. These findings may serve as a prognostic biomarker and potential therapeutic target for bone non-union.


Asunto(s)
Biomarcadores , MicroARNs , Humanos , MicroARNs/genética , MicroARNs/sangre , Biomarcadores/sangre , Redes Reguladoras de Genes , Curación de Fractura/genética , Perfilación de la Expresión Génica , Biología Computacional/métodos , Femenino , Masculino , Ontología de Genes , Regulación de la Expresión Génica , Fracturas no Consolidadas/genética , Persona de Mediana Edad
3.
Cytokine ; 173: 156436, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37979214

RESUMEN

Failure of bone healing after fracture often results in nonunion, but the underlying mechanism of nonunion pathogenesis is poorly understood. Herein, we provide evidence to clarify that the inflammatory microenvironment of atrophic nonunion (AN) mice suppresses the expression levels of DNA methyltransferases 2 (DNMT2) and 3A (DNMT3a), preventing the methylation of CpG islands on the promoters of C-terminal binding protein 1/2 (CtBP1/2) and resulting in their overexpression. Increased CtBP1/2 acts as transcriptional corepressors that, along with histone acetyltransferase p300 and Runt-related transcription factor 2 (Runx2), suppress the expression levels of six genes involved in bone healing: BGLAP (bone gamma-carboxyglutamate protein), ALPL (alkaline phosphatase), SPP1 (secreted phosphoprotein 1), COL1A1 (collagen 1a1), IBSP (integrin binding sialoprotein), and MMP13 (matrix metallopeptidase 13). We also observe a similar phenomenon in osteoblast cells treated with proinflammatory cytokines or treated with a DNMT inhibitor (5-azacytidine). Forced expression of DNMT2/3a or blockage of CtBP1/2 with their inhibitors can reverse the expression levels of BGLAP/ALPL/SPP1/COL1A1/IBSP/MMP13 in the presence of proinflammatory cytokines. Administration of CtBP1/2 inhibitors in fractured mice can prevent the incidence of AN. Thus, we demonstrate that the downregulation of bone healing genes dependent on proinflammatory cytokines/DNMT2/3a/CtBP1/2-p300-Runx2 axis signaling plays a critical role in the pathogenesis of AN. Disruption of this signaling may represent a new therapeutic strategy to prevent AN incidence after bone fracture.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal , Citocinas , ADN (Citosina-5-)-Metiltransferasas , ADN Metiltransferasa 3A , Curación de Fractura , Animales , Ratones , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Citocinas/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Metiltransferasas/metabolismo , Osteoblastos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Curación de Fractura/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A/genética , ADN Metiltransferasa 3A/metabolismo
4.
Tohoku J Exp Med ; 263(1): 17-25, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38267060

RESUMEN

MicroRNAs (miRNAs) are related to the regulation of bone metabolism. Delayed fracture healing (DFH) is a common complication after fracture surgery. The study attempted to examine the role of miR-98-5p and bone morphogenetic protein (BMP)-2 with the onset of DFH. A total of 140 patients with femoral neck fracture were recruited, including 80 cases with normal fracture healing (NFH) and 60 cases with DFH. MC3T3-E1 cells were induced cell differentiation for cell function experiments. Real-time quantitative polymerase chain reaction (RT-qPCR) was carried out to test mRNA levels. Cell proliferation and apoptosis were determined via CCK-8 and flow cytometry assay. Luciferase reporter assay was done to verify the targeted regulatory relationship of miR-98-5p with BMP-2. In comparison with NFH cases, DFH patients owned high levels of serum miR-98-5p and low concentration of BMP-2, and the levels of the two indexes are significantly negatively correlated. Both miR-98-5p and BMP-2 had the ability to predict DFH, while their combined diagnostic value is the highest. BMP-2 was demonstrated to be the target gene of miR-98-5p. Overexpression of BMP-2 reversed the role of miR-98-5p in MC3T3-E1 cell proliferation, apoptosis and differentiation. Increased miR-98-5p and decreased BMP-2 serve as potential biomarkers for the diagnosis of DFH. MiR-98-5p overexpression inhibits osteoblast proliferation and differentiation via targeting BMP-2.


Asunto(s)
Apoptosis , Proteína Morfogenética Ósea 2 , Proliferación Celular , Curación de Fractura , MicroARNs , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Apoptosis/genética , Secuencia de Bases , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 2/genética , Diferenciación Celular/genética , Línea Celular , Fracturas del Cuello Femoral/metabolismo , Fracturas del Cuello Femoral/genética , Curación de Fractura/genética , MicroARNs/genética , MicroARNs/metabolismo
5.
J Cell Mol Med ; 26(3): 601-623, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34984803

RESUMEN

Fracture non-union represents a common complication, seen in 5%-10% of all acute fractures. Despite the enhancement in scientific understanding and treatment methods, rates of fracture non-union remain largely unchanged over the years. This systematic review investigates the biological, molecular and genetic profiles of both (i) non-union tissue and (ii) non-union-related tissues, and the genetic predisposition to fracture non-union. This is crucially important as it could facilitate earlier identification and targeted treatment of high-risk patients, along with improving our understanding on pathophysiology of fracture non-union. Since this is an update on our previous systematic review, we searched the literature indexed in PubMed Medline; Ovid Medline; Embase; Scopus; Google Scholar; and the Cochrane Library using Medical Subject Heading (MeSH) or Title/Abstract words (non-union(s), non-union(s), human, tissue, bone morphogenic protein(s) (BMPs) and MSCs) from August 2014 (date of our previous publication) to 2 October 2021 for non-union tissue studies, whereas no date restrictions imposed on non-union-related tissue studies. Inclusion criteria of this systematic review are human studies investigating the characteristics and properties of non-union tissue and non-union-related tissues, available in full-text English language. Limitations of this systematic review are exclusion of animal studies, the heterogeneity in the definition of non-union and timing of tissue harvest seen in the included studies, and the search term MSC which may result in the exclusion of studies using historical terms such as 'osteoprogenitors' and 'skeletal stem cells'. A total of 24 studies (non-union tissue: n = 10; non-union-related tissues: n = 14) met the inclusion criteria. Soft tissue interposition, bony sclerosis of fracture ends and complete obliteration of medullary canal are commonest macroscopic appearances of non-unions. Non-union tissue colour and surrounding fluid are two important characteristics that could be used clinically to distinguish between septic and aseptic non-unions. Atrophic non-unions had a predominance of endochondral bone formation and lower cellular density, when compared against hypertrophic non-unions. Vascular tissues were present in both atrophic and hypertrophic non-unions, with no difference in vessel density between the two. Studies have found non-union tissue to contain biologically active MSCs with potential for osteoblastic, chondrogenic and adipogenic differentiation. Proliferative capacity of non-union tissue MSCs was comparable to that of bone marrow MSCs. Rates of cell senescence of non-union tissue remain inconclusive and require further investigation. There was a lower BMP expression in non-union site and absent in the extracellular matrix, with no difference observed between atrophic and hypertrophic non-unions. The reduced BMP-7 gene expression and elevated levels of its inhibitors (Chordin, Noggin and Gremlin) could potentially explain impaired bone healing observed in non-union MSCs. Expression of Dkk-1 in osteogenic medium was higher in non-union MSCs. Numerous genetic polymorphisms associated with fracture non-union have been identified, with some involving the BMP and MMP pathways. Further research is required on determining the sensitivity and specificity of molecular and genetic profiling of relevant tissues as a potential screening biomarker for fracture non-unions.


Asunto(s)
Fracturas Óseas , Fracturas no Consolidadas , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Curación de Fractura/genética , Fracturas Óseas/genética , Fracturas no Consolidadas/genética , Predisposición Genética a la Enfermedad , Humanos , Osteogénesis/genética
6.
J Cell Mol Med ; 25(3): 1712-1724, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33471953

RESUMEN

This study explored the therapeutic effect of bone marrow mesenchymal stem cell-derived exosomes on the treatment of obesity-induced fracture healing. Quantitative real-time PCR was used to detect the expression of lncRNA H19, miR-467 and Hoxa10 and combined with WB detection to detect osteogenic markers (RUNX2, OPN, OCN). Determine whether exosomes have entered BMSCs by immunofluorescence staining. Alkaline phosphatase (ALP) and alizarin red staining (ARS) staining were used to detect ALP activity and calcium deposition. We found that high-fat treatment can inhibit the secretion of BMSCs-derived exosomes and affect the expression of H19 carried by them. In vivo and in vitro experiments show that high-fat or obesity factors can inhibit the expression of osteogenic markers and reduce the staining activity of ALP and ARS. The treatment of exosomes from normal sources can reverse the phenomenon of osteogenic differentiation and abnormal fracture healing. Further bioinformatics analysis found that miR-467 as a regulatory molecule of lncRNA H19 and Hoxa10, and we verified the targeting relationship of the three through dual luciferase report experiments. Further, we found similar phenomena in ALP and ARS staining. Bone marrow mesenchymal stem cell-derived exosomes improve fracture healing caused by obesity.


Asunto(s)
Exosomas/metabolismo , Curación de Fractura/genética , Proteínas Homeobox A10/genética , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Osteogénesis/genética , ARN Largo no Codificante/genética , Animales , Diferenciación Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Obesidad/genética , Obesidad/metabolismo
7.
FASEB J ; 34(4): 5208-5222, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32060985

RESUMEN

Emerging evidence highlights the role of the long noncoding RNA (lncRNA) KCNQ1OT1 in fracture healing. Osteoblast proliferation, migration, and survival are pivotal during this process. In this study, we aimed to improve our understanding of the regulatory role of lncRNA KCNQ1OT1 during osteoblast proliferation, migration, and survival. We searched the gene expression omnibus databases and LncBase Experimental V.2 to identify key microRNAs (miRNAs) targets of KCNQ1OT1. MiR-701-3p was selected as a differentially expressed miRNA and RNA immunoprecipitation assays were performed to verify its interaction with KCNQ1OT1. Fibroblast growth factor receptor 3 (FGFR3) was also identified as a target of miR-701-3p. We further identified KCNQ1OT1 as a competing endogenous RNA of miR-701-3p that could influence osteoblast proliferation, migration, and apoptosis in vitro and in vivo. Taken together, our results indicate that the KCNQ1OT1/miR-701-3p/FGFR3 axis is an important regulator of osteoblast proliferation, migration, and apoptosis, and provide a new therapeutic avenue for fracture healing.


Asunto(s)
Modelos Animales de Enfermedad , Fracturas del Fémur/terapia , Curación de Fractura/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , ARN Largo no Codificante/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Apoptosis , Proliferación Celular , Fracturas del Fémur/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal
8.
J Cell Mol Med ; 24(11): 6385-6396, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32307908

RESUMEN

N6-methyladenosine (m6A) modification has been reported in various diseases and implicated in increasing numbers of biological processes. However, previous studies have not focused on the role of m6A modification in fracture healing. Here, we demonstrated that m6A modifications are decreased during fracture healing and that methyltransferase-like 3 (METTL3) is the main factor involved in the abnormal changes in m6A modifications. Down-regulation of METTL3 promotes osteogenic processes both in vitro and in vivo, and this effect is recapitulated by the suppression of miR-7212-5p maturation. Further studies have shown that miR-7212-5p inhibits osteoblast differentiation in MC3T3-E1 cells by targeting FGFR3. The present study demonstrated an important role of the METTL3/miR-7212-5p/FGFR3 axis and provided new insights on m6A modification in fracture healing.


Asunto(s)
Adenosina/análogos & derivados , Diferenciación Celular/genética , Curación de Fractura/genética , Metiltransferasas/metabolismo , MicroARNs/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patología , Adenosina/metabolismo , Animales , Línea Celular , Regulación de la Expresión Génica , Metilación , Metiltransferasas/genética , Ratones Endogámicos C57BL , MicroARNs/genética , Proteínas de Unión al ARN/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo
9.
J Cell Mol Med ; 24(19): 11524-11534, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32862562

RESUMEN

Bone fracture is one of the most common injuries. Despite the high regenerative capacity of bones, failure of healing still occurs to near 10% of the patients. Herein, we aim to investigate the modulatory role of neurofibromatosis type I gene (NF1) to osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) and new bone formation after fracture in a rat model. We studied the NF1 gene expression in normal and non-union bone fracture models. Then, we evaluated how NF1 overexpression modulated osteogenic differentiation of BMSCs, autophagy activity, mTORC1 signalling and osteoclastic bone resorption by qRT-PCR, Western blot and immunostaining assays. Finally, we injected lentivirus-NF1 (Lv-NF1) to rat non-union bone fracture model and analysed the bone formation process. The NF1 gene expression was significantly down-regulated in non-union bone fracture group, indicating NF1 is critical in bone healing process. In the NF1 overexpressing BMSCs, autophagy activity and osteogenic differentiation were significantly enhanced. Meanwhile, the NF1 overexpression inhibited mTORC1 signalling and osteoclastic bone resorption. In rat non-union bone fracture model, the NF1 overexpression significantly promoted bone formation during fracture healing. In summary, we proved the NF1 gene is critical in non-union bone healing, and NF1 overexpression promoted new bone formation after fracture by enhancing autophagy and inhibiting mTORC1 signalling. Our results may provide a novel therapeutic clue of promoting bone fracture healing.


Asunto(s)
Autofagia/genética , Fracturas Óseas/genética , Fracturas Óseas/patología , Genes de Neurofibromatosis 1 , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Osteogénesis/genética , Transducción de Señal , Animales , Resorción Ósea/genética , Resorción Ósea/patología , Diferenciación Celular/genética , Modelos Animales de Enfermedad , Curación de Fractura/genética , Fracturas no Consolidadas/genética , Fracturas no Consolidadas/patología , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patología , Ratas Sprague-Dawley
10.
Development ; 144(2): 221-234, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28096214

RESUMEN

Fractures heal predominantly through the process of endochondral ossification. The classic model of endochondral ossification holds that chondrocytes mature to hypertrophy, undergo apoptosis and new bone forms by invading osteoprogenitors. However, recent data demonstrate that chondrocytes transdifferentiate to osteoblasts in the growth plate and during regeneration, yet the mechanism(s) regulating this process remain unknown. Here, we show a spatially-dependent phenotypic overlap between hypertrophic chondrocytes and osteoblasts at the chondro-osseous border in the fracture callus, in a region we define as the transition zone (TZ). Hypertrophic chondrocytes in the TZ activate expression of the pluripotency factors [Sox2, Oct4 (Pou5f1), Nanog], and conditional knock-out of Sox2 during fracture healing results in reduction of the fracture callus and a delay in conversion of cartilage to bone. The signal(s) triggering expression of the pluripotency genes are unknown, but we demonstrate that endothelial cell conditioned medium upregulates these genes in ex vivo fracture cultures, supporting histological evidence that transdifferentiation occurs adjacent to the vasculature. Elucidating the cellular and molecular mechanisms underlying fracture repair is important for understanding why some fractures fail to heal and for developing novel therapeutic interventions.


Asunto(s)
Transdiferenciación Celular/genética , Condrocitos/fisiología , Neovascularización Fisiológica/fisiología , Osteoblastos/fisiología , Osteogénesis/fisiología , Células Madre Pluripotentes/fisiología , Animales , Huesos/citología , Huesos/fisiología , Callo Óseo/crecimiento & desarrollo , Callo Óseo/metabolismo , Cartílago/citología , Cartílago/fisiología , Células Cultivadas , Condrocitos/citología , Condrogénesis/fisiología , Curación de Fractura/genética , Curación de Fractura/fisiología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Fisiológica/genética , Osteoblastos/citología , Regulación hacia Arriba/genética
11.
Am J Pathol ; 189(1): 147-161, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30339839

RESUMEN

The terminal complement complex (TCC) is formed on activation of the complement system, a crucial arm of innate immunity. TCC formation on cell membranes results in a transmembrane pore leading to cell lysis. In addition, sublytic TCC concentrations can modulate various cellular functions. TCC-induced effects may play a role in the pathomechanisms of inflammatory disorders of the bone, including rheumatoid arthritis and osteoarthritis. In this study, we investigated the effect of the TCC on bone turnover and repair. Mice deficient for complement component 6 (C6), an essential component for TCC assembly, and mice with a knockout of CD59, which is a negative regulator of TCC formation, were used in this study. The bone phenotype was analyzed in vivo, and bone cell behavior was analyzed ex vivo. In addition, the mice were subjected to a femur osteotomy. Under homeostatic conditions, C6-deficient mice displayed a reduced bone mass, mainly because of increased osteoclast activity. After femur fracture, the inflammatory response was altered and bone formation was disturbed, which negatively affected the healing outcome. By contrast, CD59-knockout mice only displayed minor skeletal alterations and uneventful bone healing, although the early inflammatory reaction to femur fracture was marginally enhanced. These results demonstrate that TCC-mediated effects regulate bone turnover and promote an adequate response to fracture, contributing to an uneventful healing outcome.


Asunto(s)
Regeneración Ósea , Complejo de Ataque a Membrana del Sistema Complemento , Fracturas del Fémur , Curación de Fractura , Osteoclastos , Animales , Regeneración Ósea/genética , Regeneración Ósea/inmunología , Antígenos CD59/deficiencia , Técnicas de Cultivo de Célula , Complemento C6/deficiencia , Complejo de Ataque a Membrana del Sistema Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Eritrocitos/inmunología , Eritrocitos/metabolismo , Eritrocitos/patología , Fracturas del Fémur/genética , Fracturas del Fémur/inmunología , Fracturas del Fémur/metabolismo , Fracturas del Fémur/patología , Curación de Fractura/genética , Curación de Fractura/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Noqueados , Osteoclastos/inmunología , Osteoclastos/metabolismo , Osteoclastos/patología , Ovinos
12.
Curr Osteoporos Rep ; 18(3): 312-324, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32394316

RESUMEN

PURPOSE OF REVIEW: The clinical significance, target pathways, recent successes, and challenges that preclude translation of RNAi bone regenerative approaches are overviewed. RECENT FINDINGS: RNA interference (RNAi) is a promising new therapeutic approach for bone regeneration by stimulating or inhibiting critical signaling pathways. However, RNAi suffers from significant delivery challenges. These challenges include avoiding nuclease degradation, achieving bone tissue targeting, and reaching the cytoplasm for mRNA inhibition. Many drug delivery systems have overcome stability and intracellular localization challenges but suffer from protein adsorption that results in clearance of up to 99% of injected dosages, thus severely limiting drug delivery efficacy. While RNAi has myriad promising attributes for use in bone regenerative applications, delivery challenges continue to plague translation. Thus, a focus on drug delivery system development is critical to provide greater delivery efficiency and bone targeting to reap the promise of RNAi.


Asunto(s)
Regeneración Ósea/genética , Curación de Fractura/genética , MicroARNs/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Tratamiento con ARN de Interferencia/métodos , Callo Óseo , Sistemas de Liberación de Medicamentos , Humanos , MicroARNs/uso terapéutico , Nanopartículas , ARN Interferente Pequeño/uso terapéutico
13.
Curr Osteoporos Rep ; 18(5): 526-540, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32712794

RESUMEN

PURPOSE OF REVIEW: The development of the skeleton is controlled by cellular decisions determined by the coordinated activation of multiple transcription factors. Recent evidence suggests that the transcriptional regulator proteins, Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), could have important roles in directing the activity of these transcriptional programs. However, in vitro evidence for the roles of YAP and TAZ in skeletal cells has been hopelessly contradictory. The goals of this review are to provide a cross-sectional view on the state of the field and to synthesize the available data toward a unified perspective. RECENT FINDINGS: YAP and TAZ are regulated by diverse upstream signals and interact downstream with multiple transcription factors involved in skeletal development, positioning YAP and TAZ as important signal integration nodes in an hourglass-shaped signaling pathway. Here, we provide a survey of putative transcriptional co-effectors for YAP and TAZ in skeletal cells. Synthesizing the in vitro data, we conclude that TAZ is consistently pro-osteogenic in function, while YAP can exhibit either pro- or anti-osteogenic activity depending on cell type and context. Synthesizing the in vivo data, we conclude that YAP and TAZ combinatorially promote developmental bone formation, bone matrix homeostasis, and endochondral fracture repair by regulating a variety of transcriptional programs depending on developmental stage. Here, we discuss the current understanding of the roles of the transcriptional regulators YAP and TAZ in skeletal development, and provide recommendations for continued study of molecular mechanisms, mechanotransduction, and therapeutic implications for skeletal disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Desarrollo Óseo/genética , Matriz Ósea/metabolismo , Curación de Fractura/genética , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Factores de Transcripción/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Homeostasis/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Factores de Transcripción/fisiología , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Proteínas Señalizadoras YAP
14.
Int J Mol Sci ; 21(23)2020 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-33266324

RESUMEN

Recent studies on insulin, leptin, osteocalcin (OCN), and bone remodeling have evoked interest in the interdependence of bone formation and energy household. Accordingly, this study attempts to investigate trauma specific hormone changes in a murine trauma model and its influence on fracture healing. Thereunto 120 female wild type (WT) and leptin-deficient mice underwent either long bone fracture (Fx), traumatic brain injury (TBI), combined trauma (Combined), or neither of it and therefore served as controls (C). Blood samples were taken weekly after trauma and analyzed for insulin and OCN concentrations. Here, WT-mice with Fx and, moreover, with combined trauma showed a greater change in posttraumatic insulin and OCN levels than mice with TBI alone. In the case of leptin-deficiency, insulin changes were still increased after bony lesion, but the posttraumatic OCN was no longer trauma specific. Four weeks after trauma, hormone levels recovered to normal/basal line level in both mouse strains. Thus, WT- and leptin-deficient mice show a trauma specific hyperinsulinaemic stress reaction leading to a reduction in OCN synthesis and release. In WT-mice, this causes a disinhibition and acceleration of fracture healing after combined trauma. In leptin-deficiency, posttraumatic OCN changes are no longer specific and fracture healing is impaired regardless of the preceding trauma.


Asunto(s)
Lesiones Traumáticas del Encéfalo/complicaciones , Curación de Fractura , Fracturas Óseas/complicaciones , Fracturas Óseas/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Osteocalcina/metabolismo , Animales , Biomarcadores , Modelos Animales de Enfermedad , Femenino , Curación de Fractura/genética , Fracturas Óseas/genética , Hormonas/sangre , Hormonas/metabolismo , Leptina/deficiencia , Ratones , Ratones Noqueados , Modelos Biológicos , Osteogénesis
15.
Int J Mol Sci ; 21(7)2020 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-32235405

RESUMEN

The process of fracture healing consists of an inflammatory reaction and cartilage and bone tissue reconstruction. The inflammatory cytokine interleukin-1ß (IL-1ß) signal is an important major factor in fracture healing, whereas its relevance to retinoid receptor (an RAR inverse agonist, which promotes endochondral bone formation) remains unclear. Herein, we investigated the expressions of IL-1ß and retinoic acid receptor gamma (RARγ) in a rat fracture model and the effects of IL-1ß in the presence of one of several RAR inverse agonists on chondrocytes. An immunohistochemical analysis revealed that IL-1ß and RARγ were expressed in chondrocytes at the fracture site in the rat ribs on day 7 post-fracture. In chondrogenic ATDC5 cells, IL-1ß decreases the levels of aggrecan and type II collagen but significantly increased the metalloproteinase-13 (Mmp13) mRNA by real-time reverse transcription-polymerase chain reaction (RT-PCR) analysis. An RAR inverse agonist (AGN194310) inhibited IL-1ß-stimulated Mmp13 and Ccn2 mRNA in a dose-dependent manner. Phosphorylated extracellular signal regulated-kinases (pERK1/2) and p-p38 mitogen-activated protein kinase (MAPK) were increased time-dependently by IL-1ß treatment, and the IL-1ß-induced p-p38 MAPK was inhibited by AGN194310. Experimental p38 inhibition led to a drop in the IL-1ß-stimulated expressions of Mmp13 and Ccn2 mRNA. MMP13, CCN2, and p-p38 MAPK were expressed in hypertrophic chondrocytes near the invaded vascular endothelial cells. As a whole, these results point to role of the IL-1ß via p38 MAPK as important signaling in the regulation of the endochondral bone formation in fracture healing, and to the actions of RAR inverse agonists as potentially relevant modulators of this process.


Asunto(s)
Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Curación de Fractura/efectos de los fármacos , Interleucina-1beta/metabolismo , Retinoides/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Biomarcadores , Curación de Fractura/genética , Expresión Génica , Inmunohistoquímica , Masculino , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , Transporte de Proteínas , Ratas , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo
16.
J Cell Mol Med ; 23(6): 3855-3866, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30955246

RESUMEN

As fracture healing is related to gene expression, fracture healing is prospected to be implicated in long non-coding RNAs (lncRNAs). This study focuses on the effects of epigenetic silencing of long non-coding RNA maternally expressed gene 3 (lncRNA MEG3) on fracture healing by regulating the Wnt/ß-catenin signalling pathway. Genes expressed in fracture were screened using bioinformatics and the subcellular location of MEG3 was determined using FISH. Next, we successfully established tibia fracture (TF) models of C57BL/6J and Col2a1-ICAT mice and the effect of silencing lncRNA MEG3 on fracture healing was detected after TF mice were treated with phosphate buffer saline (PBS), MEG3 siRNA and scramble siRNA. X-ray imaging, Safranin-O/fast green and haematoxylin-eosin (HE) staining and histomorphometrical and biomechanical analysis were adopted to observe and to detect the fracture healing conditions. Additionally, the positive expression of collagen II and osteocalcin was examined using immunohistochemistry. At last, in the in vitro experiment, the relationship of MEG3 and the Wnt/ß-catenin signalling pathway in fraction healing was investigated. MEG3 was located in the cell nucleus. In addition, it was found that MEG3 and the Wnt/ß-catenin signalling pathway were associated with fraction healing. Moreover, silencing MEG3 was proved to elevate callus area and maximum bending load and to furthermore enhance the recanalization of bone marrow cavity. Finally, MEG3 knockdown elevated levels of Col10a1, Runx2, Osterix, Osteocalcin, Wnt10b and ß-catenin/ß-catenin whereas it reduced p-GSK-3ß/GSK-3ß levels. Taken together, our data supported that epigenetic silencing of lncRNA MEG3 could promote the tibia fracture healing by activating the Wnt/ß-catenin signalling pathway.


Asunto(s)
Curación de Fractura/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , ARN Largo no Codificante/metabolismo , Fracturas de la Tibia/metabolismo , Vía de Señalización Wnt/genética , Animales , Callo Óseo/metabolismo , Colágeno Tipo II/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Curación de Fractura/fisiología , Silenciador del Gen , Glucógeno Sintasa Quinasa 3 beta/química , Glucógeno Sintasa Quinasa 3 beta/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteocalcina/metabolismo , Interferencia de ARN , ARN Largo no Codificante/genética , Factor de Transcripción Sp7/metabolismo , Fracturas de la Tibia/genética , Análisis de Matrices Tisulares , Regulación hacia Arriba , beta Catenina/metabolismo
17.
J Cell Mol Med ; 23(6): 3843-3854, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31025509

RESUMEN

Bone repair involves bone resorption through osteoclastogenesis and the stimulation of neovascularization and osteogenesis by endothelial progenitor cells (EPCs). However, the role of EPCs in osteoclastogenesis is unclear. In this study, we assess the effects of EPC-derived exosomes on the migration and osteoclastic differentiation of primary mouse bone marrow-derived macrophages (BMMs) in vitro using immunofluorescence, western blotting, RT-PCR and Transwell assays. We also evaluated the effects of EPC-derived exosomes on the homing and osteoclastic differentiation of transplanted BMMs in a mouse bone fracture model in vivo. We found that EPCs cultured with BMMs secreted exosomes into the medium and, compared with EPCs, exosomes had a higher expression level of LncRNA-MALAT1. We confirmed that LncRNA-MALAT1 directly binds to miR-124 to negatively control miR-124 activity. Moreover, overexpression of miR-124 could reverse the migration and osteoclastic differentiation of BMMs induced by EPC-derived exosomes. A dual-luciferase reporter assay indicated that the integrin ITGB1 is the target of miR-124. Mice treated with EPC-derived exosome-BMM co-transplantations exhibited increased neovascularization at the fracture site and enhanced fracture healing compared with those treated with BMMs alone. Overall, our results suggest that EPC-derived exosomes can promote bone repair by enhancing recruitment and differentiation of osteoclast precursors through LncRNA-MALAT1.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , Exosomas/metabolismo , Macrófagos/metabolismo , MicroARNs/metabolismo , Osteoclastos/metabolismo , Osteogénesis/genética , ARN Largo no Codificante/metabolismo , Animales , Movimiento Celular/genética , Exosomas/genética , Exosomas/ultraestructura , Curación de Fractura/genética , Curación de Fractura/fisiología , Células HEK293 , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica , ARN Largo no Codificante/genética
18.
Development ; 143(16): 2946-57, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27387871

RESUMEN

Lizards are amniotes with the remarkable ability to regenerate amputated tails. The early regenerated lizard tail forms a blastema, and the regenerated skeleton consists of a cartilage tube (CT) surrounding the regenerated spinal cord. The proximal, but not distal, CT undergoes hypertrophy and ossifies. We hypothesized that differences in cell sources and signaling account for divergent cartilage development between proximal and distal CT regions. Exogenous spinal cord implants induced ectopic CT formation in lizard (Anolis carolinensis) blastemas. Regenerated spinal cords expressed Shh, and cyclopamine inhibited CT induction. Blastemas containing vertebrae with intact spinal cords formed CTs with proximal hypertrophic regions and distal non-hypertrophic regions, whereas removal of spinal cords resulted in formation of proximal CT areas only. In fate-mapping studies, FITC-labeled vertebra periosteal cells were detected in proximal, but not distal, CT areas. Conversely, FITC-labeled blastema cells were restricted to distal CT regions. Proximal cartilage formation was inhibited by removal of periosteum and could be recapitulated in vitro by periosteal cells treated with Ihh and BMP-2. These findings suggest that proximal CTs are directly derived from vertebra periosteal cells in response to BMP and Ihh signaling, whereas distal CTs form from blastema cells in response to Shh signals from regenerated spinal cords.


Asunto(s)
Lagartos/metabolismo , Cola (estructura animal)/fisiología , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Cartílago/metabolismo , Cartílago/fisiología , Ciclohexilaminas/farmacología , Curación de Fractura/efectos de los fármacos , Curación de Fractura/genética , Curación de Fractura/fisiología , Placa de Crecimiento/metabolismo , Placa de Crecimiento/fisiología , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Regeneración/efectos de los fármacos , Regeneración/genética , Regeneración/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Cola (estructura animal)/metabolismo , Tiofenos/farmacología , Alcaloides de Veratrum/farmacología
19.
Calcif Tissue Int ; 105(6): 681-686, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31489468

RESUMEN

Pycnodysostosis (PYCD) is a rare recessive inherited skeletal disease, characterized by short stature, brittle bones, and recurrent fractures, caused by variants in the Cathepsin K encoding gene that leads to impaired osteoclast-mediated bone resorption. Hypophosphatasia (HPP) is a dominant or recessive inherited condition representing a heterogeneous phenotype with dental symptoms, recurrent fractures, and musculoskeletal problems. The disease results from mutation(s) in the tissue non-specific alkaline phosphate encoding gene with reduced activity of alkaline phosphatase and secondarily defective mineralization of bone and teeth. Here, we present the first report of a patient with the coexistence of PYCD and HPP. This patient presented typical clinical findings of PYCD, including short stature, maxillary hypoplasia, and sleep apnoea. However, the burden of disease was caused by over 30 fractures, whereupon most showed delayed healing and non-union. Biochemical analysis revealed suppressed bone resorption and low bone formation capacity. We suggest that the coexistence of impaired bone resorption and mineralization may explain the severe bone phenotype with poor fracture healing.


Asunto(s)
Fracturas Múltiples/genética , Hipofosfatasia/genética , Mutación/genética , Picnodisostosis/genética , Fosfatasa Alcalina/genética , Huesos/metabolismo , Catepsina K/genética , Femenino , Curación de Fractura/genética , Fracturas Óseas/complicaciones , Fracturas Óseas/genética , Humanos , Hipofosfatasia/complicaciones , Masculino , Picnodisostosis/complicaciones
20.
Mol Cell Biochem ; 460(1-2): 165-173, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31313025

RESUMEN

Nonunion is a serious complication after fracture due to its difficulty of self-healing. MicroRNA-26a (miR-26a) has been known to play a crucial role in bone metabolism. In this study, we established a rat nonunion model by removing periosteum, and found that miR-26a was significantly upregulated. Osteogenic differentiation of mesenchymal stem cells (MSCs) isolated from bone marrow transfected with miR-26a mimics was significantly enhanced, evidenced by increased calcium deposition and expression levels of alkaline phosphatase (ALP) and osteocalcin. Bioinformatics analysis suggested that sclerostin domain-containing 1 (SOSTDC1) may be a target of miR-26a, which was confirmed by dual-luciferase assay and western blot. Besides, miR-26a was used for nonunion rats. Delightfully, radiographs of nonunion rats with miR-26a mimics administration showed obvious new bone formation compared with nonhealing control. Hematoxylin-eosin and Masson staining assays revealed that osteogenesis capacity was greatly enhanced by miR-26a mimics' administration. In addition, miR-26a mimics could promote osteogenic differentiation in nonunion rats, evidenced by increased protein levels of ALP and osteocalcin, while SOSTDC1 was suppressed. The injection of miR-26a mimics also gave rise to phosphorylation of GSK3ß and nuclear accumulation of ß-catenin, which indicated the activation of canonical Wnt/ß-catenin signaling. In conclusion, we demonstrated that miR-26a promoted fracture healing of rats with nonunion in vivo and osteogenic differentiation of MSCs in vitro, possibly by targeting SOSTDC1, and that Wnt/ß-catenin signaling pathway was involved in this process.


Asunto(s)
Curación de Fractura/genética , Fracturas no Consolidadas/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , MicroARNs/metabolismo , Vía de Señalización Wnt , Animales , Secuencia de Bases , Diferenciación Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA