Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 287
Filtrar
1.
J Cell Mol Med ; 28(13): e18522, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38957040

RESUMO

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.


Assuntos
Biomarcadores , MicroRNAs , Humanos , MicroRNAs/genética , MicroRNAs/sangue , Biomarcadores/sangue , Redes Reguladoras de Genes , Consolidação da Fratura/genética , Perfilação da Expressão Gênica , Biologia Computacional/métodos , Feminino , Masculino , Ontologia Genética , Regulação da Expressão Gênica , Fraturas não Consolidadas/genética , Pessoa de Meia-Idade
2.
J Orthop Surg Res ; 19(1): 343, 2024 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-38849896

RESUMO

BACKGROUND: Fragility fracture is common in the elderly. Osteoblast differentiation is essential for bone healing and regeneration. Expression pattern of long non-coding RNA MIAT during fracture healing was examined, and its role in osteoblast differentiation was investigated. METHODS: 90 women with simple osteoporosis and 90 women with fragility fractures were included. Another 90 age-matched women were set as the control group. mRNA levels were tested using RT-qPCR. Cell viability was detected via CCK-8, and osteoblastic biomarkers, including ALP, OCN, Collagen I, and RUNX2 were tested via ELISA. The downstream miRNAs and genes targeted by MIAT were predicted by bioinformatics analysis, whose functions and pathways were annotated via GO and KEGG analysis. RESULTS: Serum MIAT was upregulated in osteoporosis women with high accuracy of diagnostic efficacy. Serum MIAT was even elevated in the fragility fracture group, but decreased in a time manner after operation. MIAT knockdown promoted osteogenic proliferation and differentiation of MC3T3-E1, but the influences were reversed by miR-181a-5p inhibitor. A total of 137 overlapping target genes of miR-181a-5p were predicted based on the miRDB, TargetScan and microT datasets, which were mainly enriched for terms related to signaling pathways regulating pluripotency of stem cells, cellular senescence, and osteoclast differentiation. CONCLUSIONS: LncRNA MIAT serves as a promising biomarker for osteoporosis, and promotes osteogenic differentiation via targeting miR-181a-5p.


Assuntos
Biomarcadores , Diferenciação Celular , Consolidação da Fratura , Osteoblastos , RNA Longo não Codificante , RNA Longo não Codificante/genética , Humanos , Feminino , Biomarcadores/sangue , Biomarcadores/metabolismo , Consolidação da Fratura/genética , Consolidação da Fratura/fisiologia , Idoso , Diferenciação Celular/genética , Osteoblastos/metabolismo , Animais , Camundongos , MicroRNAs/genética , Osteoporose/genética , Osteoporose/metabolismo , Osteogênese/genética , Osteogênese/fisiologia , Pessoa de Meia-Idade , Fraturas por Osteoporose/genética , Proliferação de Células/genética , Regulação para Cima
3.
J Orthop Surg Res ; 19(1): 370, 2024 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-38907263

RESUMO

BACKGROUND: Long non-coding RNAs (LncRNAs) are recognized as a pivotal element in the processes of fracture healing and the osteogenic differentiation of stem cells. This study investigated the molecular mechanism and regulatory significance of lncRNA MAGI2-AS3 (MAGI2-AS3) in fracture healing. METHODS: Serum levels of MAGI2-AS3 in patients with normal and delayed fracture healing were verified by RT-qPCR assays. The predictive efficacy of MAGI2-AS3 for delayed fracture healing was analyzed by ROC curve. Osteogenic markers were quantified by RT-qPCR assays. MC3T3-E1 cell viability was detected using CCK-8 assay, and flow cytometry was utilized to measure cell apoptosis. The dual-luciferase reporter gene assay was used to determine the targeted binding between MAGI2-AS3 and miR-223-3p. RESULTS: Serum MAGI2-AS3 expression was decreased in patients with delayed fracture healing compared with patients with normal healing. Elevated MAGI2-AS3 resulted in an upregulation of the proliferative capacity of MC3T3-E1 cells and a decrease in mortality, along with increased levels of both osteogenic markers. However, after transfection silencing MAGI2-AS3, the trend was reversed. Additionally, miR-223-3p was the downstream target of MAGI2-AS3 and was controlled by MAGI2-AS3. miR-223-3p mimic reversed the promoting effects of MAGI2-AS3 overexpression on osteogenic marker levels and cell growth, and induced cell apoptosis. CONCLUSION: The upregulation of MAGI2-AS3 may expedite the healing of fracture patients by targeting miR-223-3p, offering a novel biomarker for diagnosing patients with delayed healing.


Assuntos
Regulação para Baixo , Consolidação da Fratura , MicroRNAs , RNA Longo não Codificante , MicroRNAs/genética , RNA Longo não Codificante/genética , Consolidação da Fratura/genética , Consolidação da Fratura/fisiologia , Humanos , Camundongos , Animais , Osteogênese/genética , Masculino , Feminino , Apoptose/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Adulto , Proliferação de Células/genética , Diferenciação Celular/genética
4.
PLoS One ; 19(5): e0303035, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38820355

RESUMO

Fracture non-unions affect many patients worldwide, however, known risk factors alone do not predict individual risk. The identification of novel biomarkers is crucial for early diagnosis and timely patient treatment. This study focused on the identification of microRNA (miRNA) related to the process of fracture healing. Serum of fracture patients and healthy volunteers was screened by RNA sequencing to identify differentially expressed miRNA at various times after injury. The results were correlated to miRNA in the conditioned medium of human bone marrow mesenchymal stromal cells (BMSCs) during in vitro osteogenic differentiation. hsa-miR-1246, hsa-miR-335-5p, and miR-193a-5p were identified both in vitro and in fracture patients and their functional role in direct BMSC osteogenic differentiation was assessed. The results showed no influence of the downregulation of the three miRNAs during in vitro osteogenesis. However, miR-1246 may be involved in cell proliferation and recruitment of progenitor cells. Further studies should be performed to assess the role of these miRNA in other processes relevant to fracture healing.


Assuntos
Biomarcadores , Diferenciação Celular , MicroRNA Circulante , Células-Tronco Mesenquimais , MicroRNAs , Osteogênese , Humanos , Osteogênese/genética , MicroRNAs/sangue , MicroRNAs/genética , Células-Tronco Mesenquimais/metabolismo , Biomarcadores/sangue , Masculino , MicroRNA Circulante/sangue , MicroRNA Circulante/genética , Feminino , Consolidação da Fratura/genética , Adulto , Fraturas Ósseas/sangue , Fraturas Ósseas/genética , Pessoa de Meia-Idade , Células Cultivadas , Proliferação de Células
5.
Injury ; 55(4): 111410, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38359711

RESUMO

The term "fracture" pertains to the occurrence of bones being either fully or partially disrupted as a result of external forces. Prolonged fracture healing can present a notable danger to the patient's general health and overall quality of life. The significance of osteoblasts in the process of new bone formation is widely recognized, and optimizing their function could be a desirable strategy. Therefore, the mending of bone fractures is intricately linked to the processes of osteogenic differentiation and mineralization. MicroRNAs (miRNAs) are RNA molecules that do not encode for proteins, but rather modulate the functioning of physiological processes by directly targeting proteins. The participation of microRNAs (miRNAs) in experimental investigations has been extensive, and their control functions have earned them the recognition as primary regulators of the human genome. Earlier studies have shown that modulating the expression of miRNAs, either by increasing or decreasing their levels, can initiate the differentiation of osteoblasts. This implies that miRNAs play a pivotal function in promoting osteogenesis, facilitating bone mineralization and formation, ultimately leading to an efficient healing of fractures. Hence, focusing on miRNAs can be considered a propitious therapeutic approach to accelerate the healing of fractures and forestall nonunion. In this manner, the information supplied by this investigation has the potential to aid in upcoming clinical utilization, including its possible use as biomarkers or as resources for devising innovative therapeutic tactics aimed at promoting fracture healing.


Assuntos
Fraturas Ósseas , MicroRNAs , Humanos , Osteogênese/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Consolidação da Fratura/genética , Qualidade de Vida , Fraturas Ósseas/genética , Fraturas Ósseas/terapia , Fraturas Ósseas/metabolismo , Osteoblastos/metabolismo , Diferenciação Celular
6.
Tohoku J Exp Med ; 263(1): 17-25, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38267060

RESUMO

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.


Assuntos
Apoptose , Proteína Morfogenética Óssea 2 , Proliferação de Células , Consolidação da Fratura , MicroRNAs , Idoso , Animais , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Apoptose/genética , Sequência de Bases , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 2/genética , Diferenciação Celular/genética , Linhagem Celular , Fraturas do Colo Femoral/metabolismo , Fraturas do Colo Femoral/genética , Consolidação da Fratura/genética , MicroRNAs/genética , MicroRNAs/metabolismo
7.
Cytokine ; 173: 156436, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37979214

RESUMO

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.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core , Citocinas , DNA (Citosina-5-)-Metiltransferases , DNA Metiltransferase 3A , Consolidação da Fratura , Animais , Camundongos , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Citocinas/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Metiltransferases/metabolismo , Osteoblastos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Consolidação da Fratura/genética , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A/genética , DNA Metiltransferase 3A/metabolismo
8.
Elife ; 122023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-38079220

RESUMO

Insufficient bone fracture repair represents a major clinical and societal burden and novel strategies are needed to address it. Our data reveal that the transforming growth factor-ß superfamily member Activin A became very abundant during mouse and human bone fracture healing but was minimally detectable in intact bones. Single-cell RNA-sequencing revealed that the Activin A-encoding gene Inhba was highly expressed in a unique, highly proliferative progenitor cell (PPC) population with a myofibroblast character that quickly emerged after fracture and represented the center of a developmental trajectory bifurcation producing cartilage and bone cells within callus. Systemic administration of neutralizing Activin A antibody inhibited bone healing. In contrast, a single recombinant Activin A implantation at fracture site in young and aged mice boosted: PPC numbers; phosphorylated SMAD2 signaling levels; and bone repair and mechanical properties in endochondral and intramembranous healing models. Activin A directly stimulated myofibroblastic differentiation, chondrogenesis and osteogenesis in periosteal mesenchymal progenitor culture. Our data identify a distinct population of Activin A-expressing PPCs central to fracture healing and establish Activin A as a potential new therapeutic tool.


Assuntos
Ativinas , Calo Ósseo , Consolidação da Fratura , Camundongos , Humanos , Animais , Consolidação da Fratura/genética , Osteogênese , Células-Tronco , Diferenciação Celular
9.
Signal Transduct Target Ther ; 8(1): 260, 2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-37402714

RESUMO

Traumatic brain injury (TBI) accelerates fracture healing, but the underlying mechanism remains largely unknown. Accumulating evidence indicates that the central nervous system (CNS) plays a pivotal role in regulating immune system and skeletal homeostasis. However, the impact of CNS injury on hematopoiesis commitment was overlooked. Here, we found that the dramatically elevated sympathetic tone accompanied with TBI-accelerated fracture healing; chemical sympathectomy blocks TBI-induced fracture healing. TBI-induced hypersensitivity of adrenergic signaling promotes the proliferation of bone marrow hematopoietic stem cells (HSCs) and swiftly skews HSCs toward anti-inflammation myeloid cells within 14 days, which favor fracture healing. Knockout of ß3- or ß2-adrenergic receptor (AR) eliminate TBI-mediated anti-inflammation macrophage expansion and TBI-accelerated fracture healing. RNA sequencing of bone marrow cells revealed that Adrb2 and Adrb3 maintain proliferation and commitment of immune cells. Importantly, flow cytometry confirmed that deletion of ß2-AR inhibits M2 polarization of macrophages at 7th day and 14th day; and TBI-induced HSCs proliferation was impaired in ß3-AR knockout mice. Moreover, ß3- and ß2-AR agonists synergistically promote infiltration of M2 macrophages in callus and accelerate bone healing process. Thus, we conclude that TBI accelerates bone formation during early stage of fracture healing process by shaping the anti-inflammation environment in the bone marrow. These results implicate that the adrenergic signals could serve as potential targets for fracture management.


Assuntos
Lesões Encefálicas Traumáticas , Consolidação da Fratura , Camundongos , Animais , Consolidação da Fratura/genética , Medula Óssea , Mielopoese , Camundongos Knockout , Lesões Encefálicas Traumáticas/genética , Lesões Encefálicas Traumáticas/complicações , Adrenérgicos
10.
Gene ; 874: 147481, 2023 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-37182560

RESUMO

Despite the advances in bone fracture treatment, a significant fraction of fracture patients will develop non-union. Most non-unions are treated with surgery since identifying the molecular causes of these defects is exceptionally challenging. In this study, compared with marrow bone, we generated a transcriptional atlas of human osteoprogenitor cells derived from healing callus and non-union fractures. Detailed comparison among the three conditions revealed a substantial similarity of callus and nonunion at the gene expression level. Nevertheless, when assayed functionally, they showed different osteogenic potential. Utilizing longitudinal transcriptional profiling of the osteoprogenitor cells, we identified FOS as a putative master regulator of non-union fractures. We validated FOS activity by profiling a validation cohort of 31 tissue samples. Our work identified new molecular targets for non-union classification and treatment while providing a valuable resource to better understand human bone healing biology.


Assuntos
Calo Ósseo , Consolidação da Fratura , Humanos , Consolidação da Fratura/genética , Calo Ósseo/metabolismo , Osteogênese/genética
11.
Hum Gene Ther ; 34(13-14): 649-661, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37212284

RESUMO

Substantial advances have been made in understanding the role of partial PDZ and LIM domain family's proteins in skeletal-related diseases. Yet, little is known about the effect of PDZ and LIM Domain 1 (Pdlim1) on osteogenesis and fracture repair. This study aimed to investigate whether direct gene delivery using an adenovirus vector carrying Pdlim1 (Ad-oePdlim1) or encoding shRNA-Pdlim1 (Ad-shPdlim1) could affect the osteogenic activity of preosteoblastic MC3T3-E1 cells in vitro, and influence the fracture healing of mice in vivo. We found that Ad-shPdlim1 transfection contributed to the calcified nodule formation in MC3T3-E1 cells. Downregulation of Pdlim1 enhanced the alkaline phosphatase activity and increased the expression of osteogenic markers (Runt-related transcription factor 2 [Runx2], collagen type I alpha 1 chain [Col1A1], osteocalcin [OCN], and osteopontin [OPN]). Further analysis indicated that Pdlim1 knockdown could activate ß-catenin signaling, as evidenced by the accumulation of ß-catenin in the nucleus and the increase levels of downstream regulators such as Lef1/Tcf7, axis inhibition protein 2, cyclin D1, and SRY-box transcription factor 9. By contrast, Pdlim1 overexpression resulted in inhibition of the osteogenic activity of MC3T3-E1 cells. In vivo, at day 3 after fracture,Ad-shPdlim1 adenovirus particles were injected into the fracture site of the femur of mice, and the process of fracture healing was evaluated by X-ray, micro-computed tomography and histological examination. Local injection of Ad-shPdlim1 promoted the early cartilage callus formation, restored bone mineral density, and accelerated cartilaginous ossification, with the upregulation of osteogenic gene (Runx2, Col1A1, OCN, and OPN) expression and activation of ß-catenin signaling. Thus, we concluded that inhibition of Pdlim1 contributed to osteogenesis and fracture healing by activating the ß-catenin signaling pathway.


Assuntos
Osteogênese , beta Catenina , Animais , Camundongos , Adenoviridae/genética , Adenoviridae/metabolismo , beta Catenina/genética , Diferenciação Celular/genética , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Consolidação da Fratura/genética , Osteoblastos , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogênese/genética , Microtomografia por Raio-X
12.
Zhongguo Gu Shang ; 36(4): 393-8, 2023 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-37087632

RESUMO

Pentaxin 3 (PTX3), as a multifunctional glycoprotein, plays an important role in regulating inflammatory response, promoting tissue repair, inducing ectopic calcification and maintaining bone homeostasis. The effect of PTX3 on bone mineral density (BMD) may be affected by many factors. In PTX3 knockout mice and osteoporosis (OP) patients, the deletion of PTX3 will lead to decrease of BMD. In Korean community "Dong-gu study", it was found that plasma PTX3 was negatively correlated with BMD of femoral neck in male elderly patients. In terms of bone related cells, PTX3 plays an important role in maintaining the phenotype and function of osteoblasts (OB) in OP state;for osteoclast (OC), PTX3 in inflammatory state could stimulate nuclear factor κ receptor activator of nuclear factor-κB ligand (RANKL) production and its combination with TNF-stimulated gene 6(TSG-6) could improve activity of osteoclasts and promote bone resorption;for mesenchymal stem cells (MSCs), PTX3 could promote osteogenic differentiation of MSCs through PI3K/Akt signaling pathway. In recent years, the role of PTX3 as a new bone metabolism regulator in OP and fracture healing has been gradually concerned by scholars. In OP patients, PTX3 regulates bone mass mainly by promoting bone regeneration. In the process of fracture healing, PTX3 promotes fracture healing by coordinating bone regeneration and bone resorption to maintain bone homeostasis. In view of the above biological characteristics, PTX3 is expected to become a new target for the diagnosis and treatment of OP and other age-related bone diseases and fracture healing.


Assuntos
Reabsorção Óssea , Consolidação da Fratura , Osteoporose , Animais , Masculino , Camundongos , Reabsorção Óssea/metabolismo , Diferenciação Celular , Consolidação da Fratura/genética , Osteoblastos , Osteoclastos , Osteogênese , Osteoporose/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/farmacologia
13.
Biomed Res Int ; 2023: 9950037, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36726840

RESUMO

The present study was designed to evaluate the dynamic expression of lncRNA NORAD in fracture healing of patients with brittle fractures and explore the function and mechanism of NORAD in regulating osteoblastic proliferation, differentiation, and apoptosis. The expression level of NORAD was detected by quantitative real-time PCR. The proliferation, differentiation, and apoptosis of osteoblasts were analyzed by MTT assay, ELISA, and flow cytometry. Luciferase report analysis was used to confirm the interaction between NORAD and its target ceRNA miR-26a. This study showed no significant differences in serum NORAD expression on the 7th day during fracture healing in patients, but increased expression of NORAD was certified on the 14, 21, and 28 days after fixation. Overexpression of NORAD promoted the proliferation and differentiation of osteoblasts and suppressed the apoptosis of osteoblasts. miR-26a proved to be the target gene of NORAD and was inhibited by overexpression of NORAD in osteoblasts. The enhanced expression of miR-26a was negatively linked to the lessened expression of NORAD. NORAD could accelerate the proliferation and differentiation of osteoblasts and inhibit apoptosis, thereby promoting fracture healing.


Assuntos
MicroRNAs , RNA Longo não Codificante , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Consolidação da Fratura/genética , Diferenciação Celular/genética , Osteoblastos/metabolismo , Proliferação de Células/genética
14.
Exp Mol Med ; 55(2): 443-456, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36797542

RESUMO

Bone fracture remains a common occurrence, with a population-weighted incidence of approximately 3.21 per 1000. In addition, approximately 2% to 50% of patients with skeletal fractures will develop an infection, one of the causes of disordered bone healing. Dysfunction of bone marrow mesenchymal stem cells (BMSCs) plays a key role in disordered bone repair. However, the specific mechanisms underlying BMSC dysfunction caused by bone infection are largely unknown. In this study, we discovered that Fibulin2 expression was upregulated in infected bone tissues and that BMSCs were the source of infection-induced Fibulin2. Importantly, Fibulin2 knockout accelerated mineralized bone formation during skeletal development and inhibited inflammatory bone resorption. We demonstrated that Fibulin2 suppressed BMSC osteogenic differentiation by binding to Notch2 and inactivating the Notch2 signaling pathway. Moreover, Fibulin2 knockdown restored Notch2 pathway activation and promoted BMSC osteogenesis; these outcomes were abolished by DAPT, a Notch inhibitor. Furthermore, transplanted Fibulin2 knockdown BMSCs displayed better bone repair potential in vivo. Altogether, Fibulin2 is a negative regulator of BMSC osteogenic differentiation that inhibits osteogenesis by inactivating the Notch2 signaling pathway in infected bone.


Assuntos
Consolidação da Fratura , Osteogênese , Humanos , Osso e Ossos , Diferenciação Celular/genética , Células Cultivadas , Consolidação da Fratura/genética , Osteogênese/genética , Transdução de Sinais , Células da Medula Óssea/metabolismo , Células-Tronco/metabolismo
15.
Clin Transl Med ; 13(1): e1161, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36629031

RESUMO

BACKGROUND: Approximately 10% of all bone fractures result in delayed fracture healing or non-union; thus, the identification of biomarkers and prognostic factors is of great clinical interest. MicroRNAs (miRNAs) are known to be involved in the regulation of the bone healing process and may serve as functional markers for fracture healing. AIMS AND METHODS: This systematic review aimed to identify common miRNAs involved in fracture healing or non-union fractures using a qualitative approach. A systematic literature search was performed with the keywords 'miRNA and fracture healing' and 'miRNA and non-union fracture'. Any original article investigating miRNAs in fracture healing or non-union fractures was screened. Eventually, 82 studies were included in the qualitative analysis for 'miRNA and fracture healing', while 19 were selected for the 'miRNA and fracture non-union' category. RESULTS AND CONCLUSIONS: Out of 151 miRNAs, miR-21, miR-140 and miR-214 were the most investigated miRNAs in fracture healing in general. miR-31-5p, miR-221 and miR-451-5p were identified to be regulated specifically in non-union fractures. Large heterogeneity was detected between studies investigating the role of miRNAs in fracture healing or non-union in terms of patient population, sample types and models used. Nonetheless, our approach identified some miRNAs with the potential to serve as biomarkers for non-union fractures, including miR-31-5p, miR-221 and miR-451-5p. We provide a discussion of involved pathways and suggest on alignment of future research in the field.


Assuntos
Fraturas Ósseas , MicroRNAs , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Prognóstico , Consolidação da Fratura/genética , Fraturas Ósseas/genética , Fraturas Ósseas/terapia , Biomarcadores
16.
Comb Chem High Throughput Screen ; 26(1): 149-162, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35418283

RESUMO

BACKGROUND: Bone fracture healing is a time-consuming and high-priority orthopedic problem worldwide. OBJECTIVE: Discovering the potential mechanism of bone healing at a time course and transcriptional level may better help manage bone fracture. METHODS: In this study, we analyze a time-course bone fracture healing transcriptional dataset in a rat model (GSE592, GSE594, and GSE1371) of Gene Expression Omnibus (GEO). RNA was obtained from female Sprague-Dawley rats with a femoral fracture at the initial time (day 3) as well as early (week 1), middle (week 2), and late (week 4) time periods, with nonfracture rats used as control. Gene Ontology (GO) functional analysis and pathway examinations were performed for further measurements of GSEA and hub genes. RESULTS: Results indicated that the four stages of bone fracture healing at the initial, early, middle, and late time periods represent the phases of hematoma formation, callus formation, callus molding, and mature lamellar bone formation, respectively. Extracellular organization was positively employed throughout the four stages. At the hematoma formation phase, the muscle contraction process was downregulated. Antibacterial peptide pathway was downregulated at all phases. The upregulation of Fn1 (initial, early, middle, and late time periods), Col3a1 (initial, early, and middle time periods), Col11a1 (initial and early time periods), Mmp9 (middle and late time periods), Mmp13 (early, middle, and late time periods) and the downregulation of RatNP-3b (initial, early, middle, and late time periods) were possible symbols for bone fracture healing and may be used as therapeutic targets. CONCLUSION: These findings suggest some new potential pathways and genes in the process of bone fracture healing and further provide insights that can be used in targeted molecular therapy for bone fracture healing.


Assuntos
Fraturas do Fêmur , Consolidação da Fratura , Ratos , Feminino , Animais , Consolidação da Fratura/genética , Ratos Sprague-Dawley , Calo Ósseo/metabolismo , Fraturas do Fêmur/tratamento farmacológico , Fraturas do Fêmur/metabolismo
17.
Biomater Adv ; 139: 213027, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35882120

RESUMO

The hematoma that forms between broken fragments of bone serves as a natural fibrin scaffold, and its removal from the defect site delays bone healing. The hypothesis of this study is that the microarchitectural and mechanical properties of the initially formed hematoma has a significant effect on the regulation of the biological process, which ultimately determines the outcome of bone healing. To mimic three healing conditions in the rat femur (normal, delayed, and non-healing bone defects), three different defect sizes of 0.5, 1.5, and 5.0 mm, are respectively used. The analysis of 3-day-old hematomas demonstrates clear differences in fibrin clot micro-architecture in terms of fiber diameter, fiber density, and porosity of the formed fibrin network, which result in different mechanical properties (stiffness) of the hematoma in each model. Those differences directly affect the biological processes involved. Specifically, RNA-sequencing reveals almost 700 differentially expressed genes between normally healing and non-healing defects, including significantly up-regulated essential osteogenic genes in normally healing defects, also differences in immune cell populations, activated osteogenic transcriptional regulators as well as potential novel marker genes. Most importantly, this study demonstrates that the healing outcome has already been determined during the hematoma phase of bone healing, three days post-surgery.


Assuntos
Consolidação da Fratura , Fraturas Ósseas , Animais , Fibrina , Consolidação da Fratura/genética , Hematoma/genética , Osteogênese/genética , Ratos
18.
Int J Biol Sci ; 18(10): 3961-3980, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35844802

RESUMO

The biomechanical environment plays a dominant role in fracture healing, and Piezo1 is regarded as a major mechanosensor in bone homeostasis. However, the role of Piezo1 in fracture healing is not yet well characterized. In this study, we first delineated that Piezo1 is highly expressed in periosteal stem cells (PSCs) and their derived osteoblastic lineage cells and chondrocytes. Furthermore, downregulation of Piezo1 in callus leads to impaired fracture healing, while activation by its specific agonist promotes fracture healing through stimulation of PSC-modulated chondrogenesis and osteogenesis, along with accelerated cartilage-to-bone transformation. Interestingly, vascular endothelial growth factor A is upregulated after Yoda1 treatment of PSCs, indicating an indirect role of Piezo1 in angiogenesis. Mechanistically, activation of Piezo1 promotes expression of Yes-associated protein (YAP) and its nuclear localization in PSCs, which in turn increases the expression and nuclear localization of ß-catenin. In detail, YAP directly interacts with ß-catenin in the nucleus and forms a transcriptional YAP/ß-catenin complex, which upregulates osteogenic, chondrogenic and angiogenic factors. Lastly, Yoda1 treatment significantly improves fracture healing in a delayed union mouse model generated by tail suspension. These findings indicate that Piezo1 is a potential therapeutic target for fracture delayed union or nonunion.


Assuntos
Consolidação da Fratura , beta Catenina , Animais , Calo Ósseo/metabolismo , Consolidação da Fratura/genética , Canais Iônicos/genética , Canais Iônicos/metabolismo , Camundongos , Osteogênese/genética , Células-Tronco/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
19.
Bioengineered ; 13(5): 13689-13702, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35707851

RESUMO

Diabetes is characterized by increased fracture risk. Evidence from in vivo studies is lacking for anti-fracture strategies in diabetes. Our microarray analyses predicted association of Toll-like receptor 9 (TLR9) with both diabetes and osteoporosis, which was the focus of this work in a murine model of type II diabetic osteoporosis (T2DOP). A T2DOP model with fracture was established in TLR9 knockout (TLR9-/-) mice, which were then treated with the NF-κB signaling pathway inhibitor (PDTC) and activator (TNF-α). The obtained data suggested that TLR9 knockout augmented regeneration of bone tissues and cartilage area in the callus, and diminished fibrous tissues in T2DOP mice. Moreover, TLR9 depletion significantly affected bone mineral density (BMD), bone volume/tissue volume (BV/TV), connectivity density, trabecular number, trabecular separation and trabecular thickness, thus promoting fracture recovery. Bone morphology and structure were also improved in response to TLR9 depletion in T2DOP mice. TLR9 depletion inactivated NF-κB signaling in T2DOP mice. PDTC was found to enhance fracture healing in T2DOP mice, while TNF-α negated this effect. Collectively, these data indicate that TLR9 depletion may hold anti-fracture properties, making it a potential therapeutic target for T2DOP.Abbreviations: Diabetic osteoporosis (DOP); bone mineral density (BMD); Toll-like receptors (TLRs); type 2 diabetes (T2D); Toll-like receptor 9 (TLR9); nuclear factor-kappaB (NF-κB); streptozotocin (STZ); type 2 diabetic osteoporosis (T2DOP); Gene Expression Omnibus (GEO); Kyoto encyclopedia of genes and genomes (KEGG); pyrrolidine dithiocarbamate (PDTC); computed tomography (CT); Hematoxylin-eosin (HE); bone morphogenetic protein 7 (BMP7); analysis of variance (ANOVA).


Assuntos
Diabetes Mellitus Tipo 2 , Osteoporose , Receptor Toll-Like 9 , Animais , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Consolidação da Fratura/genética , Deleção de Genes , Camundongos , NF-kappa B/genética , NF-kappa B/metabolismo , Osteoporose/complicações , Osteoporose/genética , Transdução de Sinais/genética , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
20.
PLoS One ; 17(2): e0263839, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35213543

RESUMO

The ubiquitin/proteasome system controls the stability of Runx2 and JunB, proteins essential for differentiation of mesenchymal progenitor/stem cells (MPCs) to osteoblasts. Local administration of proteasome inhibitor enhances bone fracture healing by accelerating endochondral ossification. However, if a short-term administration of proteasome inhibitor enhances fracture repair and potential mechanisms involved have yet to be exploited. We hypothesize that injury activates the ubiquitin/proteasome system in callus, leading to elevated protein ubiquitination and degradation, decreased MPCs, and impaired fracture healing, which can be prevented by a short-term of proteasome inhibition. We used a tibial fracture model in Nestin-GFP reporter mice, in which a subgroup of MPCs are labeled by Nestin-GFP, to test our hypothesis. We found increased expression of ubiquitin E3 ligases and ubiquitinated proteins in callus tissues at the early phase of fracture repair. Proteasome inhibitor Bortezomib, given soon after fracture, enhanced fracture repair, which is accompanied by increased callus Nestin-GFP+ cells and their proliferation, and the expression of osteoblast-associated genes and Runx2 and JunB proteins. Thus, early treatment of fractures with Bortezomib could enhance the fracture repair by increasing the number and proliferation of MPCs.


Assuntos
Bortezomib/farmacologia , Proliferação de Células/efeitos dos fármacos , Consolidação da Fratura/efeitos dos fármacos , Células-Tronco Mesenquimais/enzimologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Fraturas da Tíbia/enzimologia , Animais , Proliferação de Células/genética , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Modelos Animais de Doenças , Consolidação da Fratura/genética , Masculino , Camundongos , Camundongos Transgênicos , Osteoblastos/enzimologia , Complexo de Endopeptidases do Proteassoma/genética , Fraturas da Tíbia/tratamento farmacológico , Fraturas da Tíbia/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/biossíntese , Ubiquitina-Proteína Ligases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...