Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Adv Healthc Mater ; 13(16): e2303510, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38545904

RESUMEN

Targeted drug delivery and the reduction of off-target effects are crucial for the promising clinical application of nucleic acid drugs. To address this challenge, a new approach for treating osteoarthritis (OA) that accurately delivers antisense oligonucleotides (ASO) targeting matrix metalloproteinase-13 (ASO-MMP13) to chondrocytes, is developed. Small extracellular vesicles (exos) are ligated with chondrocyte affinity peptide (CAP) using Sortase A and subsequently incubated with cholesterol-modified ASO-MMP13 to construct a chondrocyte-targeted drug delivery exo (CAP-exoASO). Compared with exos without CAP (ExoASO), CAP-exoASOs attenuate IL-1ß-induced chondrocyte damage and prolong the retention time of ASO-MMP13 in the joint without distribution in major organs following intra-articular injection. Notably, CAP-exoASOs decrease MMP13 expression (P < 0.001) and upregulate COL2A1 expression (P = 0.006), resulting in reorganization of the cartilage matrix and alleviation of progression in the OA model. Furthermore, the Osteoarthritis Research Society International (OARSI) score of articular cartilage tissues treated with CAP-exoASO is comparable with that of healthy rats (P = 0.148). A mechanistic study demonstrates that CAP-exoASO may reduce inflammation by suppressing the IL-17 and TNF signaling pathways. Based on the targeted delivery effect, CAP-exoASOs successfully accomplish cartilage repair and have considerable potential for development as a promising therapeutic modality for satisfactory OA therapy.


Asunto(s)
Aminoaciltransferasas , Proteínas Bacterianas , Condrocitos , Cisteína Endopeptidasas , Vesículas Extracelulares , Metaloproteinasa 13 de la Matriz , Osteoartritis , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 13 de la Matriz/genética , Animales , Osteoartritis/terapia , Osteoartritis/metabolismo , Osteoartritis/patología , Osteoartritis/genética , Condrocitos/metabolismo , Ratas , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/química , Aminoaciltransferasas/metabolismo , Aminoaciltransferasas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/genética , Masculino , Sistemas de Liberación de Medicamentos/métodos , Ratas Sprague-Dawley , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/administración & dosificación , Cartílago Articular/metabolismo , Cartílago Articular/patología
2.
Adv Sci (Weinh) ; 11(2): e2302037, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38015024

RESUMEN

Mono-methylation of histone H3 on Lys 4 (H3K4me1), which is catalyzed by histone-lysine N-methyltransferase 2D (KMT2D), serves as an important epigenetic regulator in transcriptional control. In this study, the authors identify early B-cell factor 2 (EBF2) as a binding protein of H3K4me1. Combining analyses of RNA-seq and ChIP-seq data, the authors further identify killin (KLLN) as a transcriptional target of KMT2D and EBF2 in pancreatic ductal adenocarcinoma (PDAC) cells. KMT2D-dependent H3K4me1 and EBF2 are predominantly over-lapped proximal to the transcription start site (TSS) of KLLN gene. Comprehensive functional assays show that KMT2D and EBF2 cooperatively inhibit PDAC cells proliferation, migration, and invasion through upregulating KLLN. Such inhibition on PDAC progression is also achieved through increasing H3K4me1 level by GSK-LSD1, a selective inhibitor of lysine-specific demethylase 1 (LSD1). Taken together, these findings reveal a new mechanism underlying PDAC progression and provide potential therapeutic targets for PDAC treatment.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Carcinoma Ductal Pancreático/genética , Regulación de la Expresión Génica , Histona Demetilasas/genética , Histonas/genética , Neoplasias Pancreáticas/genética
3.
Cytotechnology ; 75(6): 505-516, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37841957

RESUMEN

Osteoporosis is a complicated multifactorial disorder characterized by low bone mass and deteriorated bone microarchitecture with an elevated fracture risk. MicroRNAs play important roles in osteoblastic differentiation. In the present study, we found that miR-224-5p was markedly downregulated during the osteogenic differentiation of C2C12 cells. Overexpression of miR-224-5p in C2C12 cells inhibited osteoblast activity, as indicated by reduced ALP activity, matrix mineralization and the expression of osteogenic marker genes. Moreover, we demonstrated that Runx2 and Sp7 were direct targets of miR-224-5p. Furthermore, the specific inhibition of miR-224-5p by femoral bone marrow cavity injection with miR-224-5p antagomir prevented ovariectomy-induced bone loss. Finally, we found that the levels of miR-224-5p were markedly elevated in the sera of patients with osteoporosis. Collectively, this study revealed that miR-224-5p negatively regulates osteogenic differentiation by targeting Runx2 and Sp7. It also highlights the potential use of miR-224-5p as a therapeutic target and diagnostic biomarker for osteoporosis. Supplementary information: The online version contains supplementary material available at 10.1007/s10616-023-00593-z.

4.
Ann Anat ; 250: 152156, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37678499

RESUMEN

BACKGROUND: The aim of this study was to investigate the effect of isopsoralen on osteogenic differentiation of human jawbone marrow mesenchymal cells and its possible mechanism. METHOD: The cytotoxicity and proliferation of cells were measured by a cell counting kit 8. Alkaline phosphatase activity analysis was then used to determine the optimal concentration of isopsoralen to promote the differentiation. Western blot, qRT-PCR and Alizarin Red S staining were used to evaluate the role of Notch signaling pathway in isopsoralen-induced osteogenic differentiation. This study also investigated the anti-osteoporotic effects of ISO using in vivo osteoporosis models. RESULTS: Our results showed that 1 × 10-6 mol / L isopsoralen can effectively promote the proliferation and osteogenic differentiation of cells. Moreover, we found that activation of notch signaling pathway inhibited isopsoralen-induced osteogenesis and inhibition of Notch signal promoted the differentiation of osteoblasts induced by isopsoralen. In vivo experiments revealed that ISO significantly inhibited OVX-induced bone mineral density loss and restored the impaired bone structural properties in osteoporosis model mice. CONCLUSION: Our findings demonstrated that isopsoralen induced osteogenic differentiation by inhibiting Notch signaling and it might be a potential therapeutic agent for treating or preventing osteoporosis.


Asunto(s)
Células Madre Mesenquimatosas , Osteoporosis , Humanos , Ratones , Animales , Osteogénesis , Médula Ósea/metabolismo , Células Cultivadas , Diferenciación Celular , Transducción de Señal , Osteoporosis/tratamiento farmacológico , Células de la Médula Ósea/metabolismo
5.
iScience ; 26(10): 107838, 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37752950

RESUMEN

HS3ST3B1-IT1 was identified as a downregulated long noncoding RNA in osteoarthritic cartilage. However, its roles and mechanisms in the pathogenesis of osteoarthritis (OA) are unclear. In this study, we demonstrated that the expressions of HS3ST3B1-IT1 and its maternal gene HS3ST3B1 were downregulated and positively correlated in osteoarthritic cartilage. Overexpression of HS3ST3B1-IT1 significantly increased chondrocyte viability, inhibited chondrocyte apoptosis, and upregulated extracellular matrix (ECM) proteins, whereas HS3ST3B1-IT1 knockdown had the opposite effects. In addition, HS3ST3B1-IT1 significantly ameliorated monosodium-iodoacetate-induced OA in vivo. Mechanistically, HS3ST3B1-IT1 upregulated HS3ST3B1 expression by blocking its ubiquitination-mediated degradation. Knockdown of HS3ST3B1 reversed the effects of HS3ST3B1-IT1 on chondrocyte viability, apoptosis, and ECM metabolism. AlkB homolog 5 (ALKBH5)-mediated N6-methyladenosine (m6A) demethylation stabilized HS3ST3B1-IT1 RNA. Together, our data revealed that ALKBH5-mediated upregulation of HS3ST3B1-IT1 suppressed OA progression by elevating HS3ST3B1 expression, suggesting that HS3ST3B1-IT1/HS3ST3B1 may serve as potential therapeutic targets for OA treatment.

6.
Cell Rep ; 42(6): 112589, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37270777

RESUMEN

Osteoarthritis (OA) is the most common degenerative disorder, affecting approximately half of the elderly population. In this study, we find that the expressions of long noncoding RNA (lncRNA) IGFBP7-OT and its maternal gene, IGFBP7, are upregulated and positively correlated in osteoarthritic cartilage. Overexpression of IGFBP7-OT significantly inhibits chondrocyte viability, promotes chondrocyte apoptosis, and reduces extracellular matrix components, whereas IGFBP7-OT knockdown has the opposite effects. IGFBP7-OT overexpression promotes cartilage degeneration and markedly aggravates the monosodium iodoacetate-induced OA phenotype in vivo. Further mechanistic research reveals that IGFBP7-OT promotes OA progression by upregulating IGFBP7 expression. Specifically, IGFBP7-OT suppresses the occupancy of DNMT1 and DNMT3a on the IGFBP7 promoter, thereby inhibiting methylation of the IGFBP7 promoter. The upregulation of IGFBP7-OT in OA is partially controlled by METTL3-mediated N6-methyladenosine (m6A) modification. Collectively, our findings reveal that m6A modification of IGFBP7-OT promotes OA progression by regulating the DNMT1/DNMT3a-IGFBP7 axis and provide a potential therapeutical target for OA treatment.


Asunto(s)
ADN Metiltransferasa 3A , Metilasas de Modificación del ADN , Osteoartritis , ARN Largo no Codificante , Anciano , Humanos , Apoptosis , Cartílago/metabolismo , Condrocitos , Metilasas de Modificación del ADN/metabolismo , Metiltransferasas/metabolismo , Osteoartritis/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Regulación hacia Arriba/genética , ADN Metiltransferasa 3A/metabolismo , Animales , Ratones
7.
Cell Biol Int ; 47(8): 1427-1440, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37186433

RESUMEN

Dysregulation of the deubiquitinating protease, UBP43, has been implicated in many human diseases, including cancer. Here, we evaluated the functional significance and mechanism of action of UBP43 in epithelial ovarian cancer. We found that UBP43 was significantly upregulated in the tumor tissues of patients with epithelial ovarian cancer. Similar results were observed in OVCAR-3, Caov-3, TOV-112D, A2780, and SK-OV-3 cells. Furthermore, in vitro functional assays of A2780 and TOV-112D cells demonstrated that UBP43 overexpression promoted cell proliferation, migration, and invasion. Upregulation of UBP43 might result in epithelial-mesenchymal transition by inducing the nuclear transport of ß-catenin, which was accompanied by enhanced N-cadherin but decreased E-cadherin expression. These malignant phenotypes were reversed by UBP43 silencing. Further investigation revealed that the knockdown of UBP43 inhibited cell proliferation by inducing a cell cycle arrest at the G2/M phase. The oncogenic characteristics of UBP43 were validated in a subcutaneous xenograft mouse model. In vivo, tumor growth was delayed in the UBP43-silenced group but accelerated after UBP43 overexpression. Finally, we demonstrated that ß-catenin is a key protein in the UBP43-mediated malignant development of epithelial ovarian cancer. Specifically, overexpression of UBP43 decreased the ubiquitination degradation of ß-catenin and enhanced its protein stability. Also, we observed that the downstream genes of beta-catenin such as cyclin D1, MMP2, and MMP9 were upregulated due to UBP43 overexpression. Thus, we concluded that UBP43 promoted epithelial ovarian cancer tumorigenesis and metastasis through activation of the ß-catenin pathway, suggesting that UBP43 may be a potential therapeutic target for this intractable disease.


Asunto(s)
Neoplasias Ováricas , beta Catenina , Animales , Femenino , Humanos , Ratones , Apoptosis , beta Catenina/metabolismo , Carcinogénesis/genética , Carcinoma Epitelial de Ovario/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/genética , Vía de Señalización Wnt
8.
Front Aging Neurosci ; 15: 1091919, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36845659

RESUMEN

Objective: Variants in the glucocerebrosidase (GBA) gene are the most common and significant risk factor for Parkinson's disease (PD). However, the impact of GBA variants on PD disease progression in the Chinese population remains unclear. This study aimed to explore the significance of GBA status on motor and cognitive impairment in a longitudinal cohort of Chinese patients with PD. Methods: The entire GBA gene was screened by long-range polymerase chain reaction (LR-PCR) and next generation sequencing (NGS). A total of 43 GBA-related PD (GBA-PD) and 246 non-GBA-mutated PD (NM-PD) patients with complete clinical data at baseline and at least one follow-up were recruited for this study. The associations of GBA genotype with rate of motor and cognitive decline, as measured by Unified PD Rating Scale (UPDRS) motor and Montreal Cognitive Assessment (MoCA), were assessed by linear mixed-effect models. Results: The estimated (standard error, SE) UPDRS motor [2.25 (0.38) points/year] and MoCA [-0.53 (0.11) points/year] progression rates in the GBA-PD group were significantly faster than those in the NM-PD group [1.35 (0.19); -0.29 (0.04) points/year; respectively]. In addition, the GBA-PD group showed significantly faster estimated (SE) bradykinesia [1.04 (0.18) points/year], axial impairment [0.38 (0.07) points/year], and visuospatial/executive [-0.15 (0.03) points/year] progression rates than the NM-PD group [0.62 (0.10); 0.17 (0.04); -0.07 (0.01) points/year; respectively]. Conclusion: GBA-PD is associated with faster motor and cognitive decline, specifically greater disability in terms of bradykinesia, axial impairment, and visuospatial/executive function. Better understanding of GBA-PD progression may help predict prognosis and improve clinical trial design.

9.
Br J Cancer ; 127(12): 2141-2153, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36216883

RESUMEN

BACKGROUND: LncRNA FGF14-AS2 is a critical suppressor in breast cancer (BCa) metastasis. However, whether FGF14-AS2 plays a role in the bone metastasis of BCa remains unknown. METHODS: TRAP assay and intratibial injection were carried out to evaluate the role of FGF14-AS2 in BCa bone metastasis in vitro and in vivo. Polyribosome profiling was done to examine the translation level. RNA pulldown combined with LC/MS was performed to identify the lncRNA-binding partner, RIP, dual-luciferase assay, and Co-IP assays as well to testify these physical interactions. The prognostic value of FGF14-AS2 expression level in BCa patients was analysed using Kaplan-Meier Plotter. RESULTS: We found that FGF14-AS2 suppresses osteoclast differentiation and osteolytic metastasis of BCa. Mechanistically, FGF14-AS2 suppresses the translation of RUNX2 by inhibiting the assembly of eIF4E/eIF4G complex and the phosphorylation of eIF4E, thereby reducing the transcription of RANKL, an essential regulator of osteoclast differentiation. Moreover, FGF14-AS2 is downregulated by YTHDF2-mediated RNA degradation in an m6A-dependent manner. Clinically, patients with high YTHDF2 and low FGF14-AS2 expression levels showed worse distant metastasis-free survival (DMFS). CONCLUSIONS: FGF14-AS2 plays a crucial role in osteolytic metastasis, and may serve as a promising prognostic biomarker and therapeutic target for BCa bone metastasis.


Asunto(s)
Neoplasias de la Mama , ARN Largo no Codificante , Humanos , Femenino , Neoplasias de la Mama/genética , ARN Largo no Codificante/genética , Biosíntesis de Proteínas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Proteínas de Unión al ARN/genética
10.
Genes Genomics ; 44(7): 801-812, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35567715

RESUMEN

BACKGROUND: Tissue factor pathway inhibitor-1 (TFPI) is a serine protease inhibitor, which is responsible for inactivating TF-induced coagulation. Recently, increasing studies revealed that TFPI was lowly expressed in tumor cells and exhibited the antitumor activity. OBJECTIVE: The aim of this study was to explore the role and underlying molecular mechanisms of TFPI in breast cancer. METHODS: The expression and prognostic value of TFPI were analyzed using UALCAN and Kaplan-Meier plotter website. The expression level of TFPI in breast cancer tissues and cells was examined by immunohistochemistry (IHC) and western blot analysis, respectively. Cellular proliferation was evaluated by CCK-8 and colony formation assays. Cell migration and invasion were determined by transwell assay. The methylation level of TFPI promoter was determined by methylation-specific PCR. RESULTS: TFPI expression was significantly lower in breast cancer tissues and cells compared to normal breast tissues and normal breast cells. Patients with low TFPI levels showed worse overall survival (OS). Furthermore, overexpression of TFPI significantly inhibited the proliferation, migration and invasion of breast cancer cells. Conversely, knockdown of TFPI promoted the proliferation, migration and invasion of breast cancer cells. Mechanistically, TFPI inhibited the ERK/p38 MAPK signaling pathway in breast cancer. Moreover, DNA hypermethylation of TFPI promoter was responsible for the downregulation of TFPI in breast cancer cells. CONCLUSION: TFPI inhibited breast cancer cell proliferation, migration and invasion through inhibition of the ERK/p38 MAPK signaling pathway, suggesting that TFPI may serve as a novel prognostic biomarker and therapeutic target for breast cancer.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Mama/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Lipoproteínas/genética , Lipoproteínas/metabolismo , Lipoproteínas/uso terapéutico , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/uso terapéutico
12.
Eur J Neurol ; 29(4): 1017-1024, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34951095

RESUMEN

BACKGROUND AND PURPOSE: Variants in the glucocerebrosidase (GBA) gene are recognized as a common and important genetic risk factor for Parkinson disease (PD). However, the impact of variant severity on the clinical phenotype of PD in the Chinese population remains unclear. Thus, the present study aimed to determine the frequency of GBA-related PD (GBA-PD) and the relationship of GBA variant severity with clinical characteristics in a large Chinese cohort. METHODS: Long-range polymerase chain reaction and next generation sequencing were performed for the entire GBA gene. GBA variant severity was classified into five classes: mild, severe, risk, complex, and unknown. RESULTS: Among the total 737 PD patients, 47 GBA variants were detected in 79 (10.72%) patients, and the most common GBA variants were R163Q, L444P, and R120W. Complete demographic and clinical data were obtained for 673 patients, which revealed that 18.50% of early onset PD patients had GBA variants. Compared with patients without GBA variants, GBA-PD patients experienced PD onset an average of 4 years earlier and had more severe motor and nonmotor symptoms. Patients carrying severe and complex variants had a higher burden of nonmotor symptoms, especially depression, and more mood/cognitive and gastrointestinal symptoms than patients carrying mild variants. CONCLUSIONS: GBA-PD is highly prevalent in the Chinese population. The severity of GBA variants underlies distinct phenotypic spectrums, with PD patients carrying severe and complex variants seeming to have similar phenotypes. PD patient stratification by GBA variant severity should become a prerequisite for selecting specific treatments.


Asunto(s)
Glucosilceramidasa , Enfermedad de Parkinson , China/epidemiología , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Glucosilceramidasa/genética , Humanos , Mutación/genética , Enfermedad de Parkinson/epidemiología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/psicología , Prevalencia
13.
Exp Mol Med ; 53(11): 1723-1734, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34737423

RESUMEN

Long noncoding RNAs (lncRNAs) have emerged as important regulators of osteoarthritis (OA), but the biological roles and clinical significance of most lncRNAs in OA are not fully understood. Microarray analysis was performed to identify differentially expressed lncRNAs, mRNAs, and miRNAs between normal and osteoarthritic cartilage. We found that AC008440.5 (abbreviated AC008), as well as AQP1 and ANKH, were highly expressed in osteoarthritic cartilage, whereas miR-328-3p was expressed at a low level in osteoarthritic cartilage. Functional assays showed that ectopic expression of AC008, AQP1, and ANKH significantly decreased chondrocyte viability and promoted chondrocyte apoptosis and extracellular matrix (ECM) degradation, whereas knockdown of AC008, AQP1, and ANKH resulted in the opposite effects. Moreover, miR-328-3p overexpression increased chondrocyte viability and attenuated chondrocyte apoptosis and ECM degradation, whereas inhibition of miR-328-3p resulted in the opposite effects. Bioinformatics analysis, RNA immunoprecipitation (RIP), and luciferase assays revealed that AC008 functioned as a competing endogenous RNA (ceRNA) to regulate miR-328-3p, which specifically targeted the AQP1 and ANKH genes. In addition, miR-328-3p significantly ameliorated MIA-induced OA, whereas AC008 accelerated OA progression in vivo. Furthermore, fat mass and obesity-associated (FTO)-mediated N6-methyladenosine demethylation downregulated AC008 transcription, while lower FTO expression led to upregulation of AC008 transcription in OA. In conclusion, our data reveal that AC008 plays a critical role in OA pathogenesis via the miR-328-3p‒AQP1/ANKH pathway, suggesting that AC008 may be a potential therapeutic target for OA.


Asunto(s)
Adenosina/análogos & derivados , Acuaporina 1/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Osteoartritis/etiología , Osteoartritis/metabolismo , Proteínas de Transporte de Fosfato/genética , Regiones no Traducidas 3' , Adenosina/metabolismo , Anciano , Anciano de 80 o más Años , Apoptosis/genética , Cartílago Articular/metabolismo , Cartílago Articular/patología , Células Cultivadas , Condrocitos/metabolismo , Progresión de la Enfermedad , Matriz Extracelular/metabolismo , Femenino , Humanos , Masculino , Metilación , Persona de Mediana Edad , Modelos Biológicos , Osteoartritis/patología , Interferencia de ARN , ARN Largo no Codificante/genética
14.
J Cell Mol Med ; 25(11): 5025-5037, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33942497

RESUMEN

Osteoporosis is a metabolic disorder characterized by low bone mass and deteriorated microarchitecture, with an increased risk of fracture. Some miRNAs have been confirmed as potential modulators of osteoblast differentiation to maintain bone mass. Our miRNA sequencing results showed that miR-664-3p was significantly down-regulated during the osteogenic differentiation of the preosteoblast MC3T3-E1 cells. However, whether miR-664-3p has an impact on bone homeostasis remains unknown. In this study, we identified overexpression of miR-664-3p inhibited the osteoblast activity and matrix mineralization in vitro. Osteoblastic miR-664-3p transgenic mice exhibited reduced bone mass due to suppressed osteoblast function. Target prediction analysis and experimental validation confirmed Smad4 and Osterix (Osx) are the direct targets of miR-664-3p. Furthermore, specific inhibition of miR-664-3p by subperiosteal injection with miR-664-3p antagomir protected against ovariectomy-induced bone loss. In addition, miR-664-3p expression was markedly higher in the serum from patients with osteoporosis compared to that from normal subjects. Taken together, this study revealed that miR-664-3p suppressed osteogenesis and bone formation via targeting Smad4 and Osx. It also highlights the potential of miR-664-3p as a novel diagnostic and therapeutic target for osteoporotic patients.


Asunto(s)
Diferenciación Celular , MicroARNs/genética , Osteoblastos/patología , Osteogénesis , Osteoporosis/patología , Proteína Smad4/antagonistas & inhibidores , Factor de Transcripción Sp7/antagonistas & inhibidores , Animales , Densidad Ósea , Proliferación Celular , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Osteoporosis/etiología , Osteoporosis/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Factor de Transcripción Sp7/genética , Factor de Transcripción Sp7/metabolismo
15.
Exp Ther Med ; 21(4): 379, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33680101

RESUMEN

The neural crest is one of the key features of craniofacial development. MicroRNA-1 (miR-1) is a single-stranded noncoding RNA that serves an important role in embryonic development. However, the function of miR-1 in neural crest cells (NCCs) is unknown. Therefore, to evaluate the role of miR-1 in NCC development, a miR-1 mutant zebrafish was generated in the current study. Mouse NCCs were isolated from the first branchial arch of embryos at gestational day E9.5, and miR-1 was silenced using a miR-1 inhibitor. To the best of our knowledge, the present study was the first to report that homozygous zebrafish lacking miR-1 exhibited developmental defects in NCC-derived craniofacial bones, heart, melanocytes and iridophores. These defects may be caused by an increase in apoptosis of NCCs during their migration and differentiation in embryonic development. Moreover, the apoptosis analysis and western blotting results demonstrated that this effect was modulated via the mitochondrial apoptosis pathway, and miR-1 inhibited NCC apoptosis by modulating this pathway. These results collectively suggested that miR-1 in NCCs may be essential for craniofacial development.

16.
Cell Death Discov ; 6: 103, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33083023

RESUMEN

Long noncoding RNAs (lncRNAs) have emerged as important regulators in cancers, including breast cancer. However, the overall biological roles and clinical significance of most lncRNAs are not fully understood. This study aimed to elucidate the potential role of a novel lncRNA FGF14-AS2 and the mechanisms underlying metastasis in breast cancer. The lncRNA FGF14-AS2 was significantly downregulated in breast cancer tissues; patients with lower FGF14-AS2 expression had advanced clinical stage. In vitro and in vivo assays of FGF14-AS2 alterations revealed a complex integrated phenotype affecting breast cancer cell migration, invasion, and tumor metastasis. Mechanistically, FGF14-AS2 functioned as a competing endogenous RNA of miR-370-3p, thereby leading to the activation of its coding counterpart, FGF14. Clinically, we observed increased miR-370-3p expression in breast cancer tissues, whereas FGF14 expression was decreased in breast cancer tissues compared to the adjacent normal breast tissues. FGF14-AS2 expression was significantly negatively correlated with miR-370-3p expression, and correlated positively to FGF14 expression. Collectively, our findings support a model in which the FGF14-AS2/miR-370-3p/FGF14 axis is a critical regulator in breast cancer metastasis, suggesting a new therapeutic direction in breast cancer.

17.
Mol Med Rep ; 22(3): 1717-1726, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32705283

RESUMEN

Osteoporosis is a complex multifactorial disorder characterized by microarchitectural deterioration, low bone mass, and increased risk of fractures or broken bones. Balanced bone remodeling is tightly regulated by the differentiation, activity and apoptosis of bone­forming osteoblasts and bone­resorbing osteoclasts. MicroRNAs (miRs) are dysregulated in osteoporosis, but whether they control osteogenic differentiation and skeletal biology, or could serve as therapeutic targets remains to be elucidated. The present study identified miR­27a­3p as a critical suppressor of osteoblastogenesis. Bioinformatics analysis and luciferase reporter assays demonstrated that miR­27a­3p directly targeted and controlled the expression of osterix (Osx), an early response gene essential for bone formation, through its 3'­untranslated region. miR­27a­3p functionally inhibited the differentiation of preosteoblasts by decreasing Osx expression, which synergistically contributed to bone formation. miR­27a­3p level was significantly decreased during osteogenic differentiation and increased in the serum of patients with osteoporosis. Together, miR­27a­3p contributed to diminished osteogenic function during osteogenic differentiation and might thus serve as a therapeutic target and diagnostic biomarker for osteoporosis.


Asunto(s)
MicroARNs/genética , Osteogénesis , Osteoporosis/genética , Factor de Transcripción Sp7/genética , Regiones no Traducidas 3' , Células 3T3 , Anciano , Anciano de 80 o más Años , Animales , Diferenciación Celular , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Persona de Mediana Edad
18.
Biotechnol Lett ; 42(5): 707-716, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32040675

RESUMEN

OBJECTIVES: Following a specific number of mitotic divisions, primary chondrocytes undergo proliferative senescence, thwarting efforts to expand sufficient populations in vitro suitable to meet the needs of scientific research or medical therapies. Therefore, the human telomerase reverse transcriptase (TERT) was used to immortalize human chondrocyte and establish a cell line that escape from cellular senescence. RESULTS: The human chondrocytes were successfully immortalized by ectopic stable expression of TERT. The established TERT-Chondrocyte cell line showed robust proliferation capacity, even in late passages up to P20, and displayed little cellular senescence. Moreover, TERT-Chondrocyte cells at 20th passage showed similar chondrocyte properties to normal chondrocytes at early passages. CONCLUSIONS: Ectopic stable expression of TERT is an effective way to immortalized human chondrocyte. The immortalized chondrocytes displayed little cellular senescence, showed promise as an in vitro model to investigate osteoarthritis, and may be a promising resource for cell-based therapy for damaged cartilage.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Condrocitos/citología , Osteoartritis/patología , Telomerasa/genética , Línea Celular , Proliferación Celular , Células Cultivadas , Senescencia Celular , Condrocitos/metabolismo , Humanos , Telomerasa/metabolismo , Transfección
19.
J Biomed Res ; 35(3): 189-196, 2020 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-33911052

RESUMEN

Triple-negative breast cancer (TNBC) has a poor prognosis and typically earlier onset of metastasis in comparison with other breast cancer subtypes. It has been reported that insulin receptor (INSR) is downregulated in TNBC, however, its clinical significance and functions in TNBC remain to be elucidated. In this study, we found that INSR expression was significantly downregulated in TNBC, and overexpression of INSR suppressed cell migration and invasion in TNBC. In addition, the survival rate of breast cancer patients with low INSR expression was lower than that of patients with high INSR expression. INSR expression was significantly correlated with lymph node metastasis, clinical tumor stages, ER status, PR status, and the proliferation index Ki-67 expression. In summary, our study suggests that INSR may serve as a biomarker for breast cancer prognosis and it may be a potential target for TNBC treatment.

20.
Connect Tissue Res ; 61(6): 577-585, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-31305177

RESUMEN

Purpose: Human jaw bone marrow mesenchymal stem cells (h-JBMMSCs) are multipotent progenitor cells with osteogenic differentiation potential. MicroRNAs (miRNAs) have emerged as crucial modulators of osteoblast differentiation. In this study, we focus on the role of miR-145 and its target protein in osteoblast differentiation of h-JBMMSCs. Materials and Methods: h-JBMMSCs were isolated and cultured in osteogenic medium. miR-145 mimics and inhibitors were used to elevate and inhibit miR-145 expression, respectively. Osteogenic differentiation was determined by Alkaline phosphatase (ALP) and Alizarin red S (ARS) staining, and osteogenic marker detection using quantitative real-time reverse transcription PCR (qRT-PCR) assay. Bioinformatic analysis and luciferase reporter assay were used to identify the target gene of miR-145. Results: MiR-145 was down-regulated during osteogenesis of h-JBMMSCs. Inhibition of miR-145 promoted osteogenic differentiation of h-JBMMSCs, revealed by enhanced activity of alkaline phosphatase (ALP), greater mineralisation, and increased expression levels of the osteogenic markers, such as Runt-related transcription factor 2 (RUNX2), Osterix (OSX), ALP and COL1A1. MiR-145 could negatively regulate semaphorin3A (SEMA3A), which acts as a positive regulator of osteogenesis. MiR-145 inhibitor induced osteogenesis could be partially attenuated by SEMA3A siRNA treatment in h-JBMMSCs. Conclusions: Our data show that miR-145 directly targets SEMA3A, and also suggest miR-145 as a suppressor, plays an important role in the osteogenic differentiation of h-JBMMSCs.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular/genética , Maxilares/citología , Células Madre Mesenquimatosas/citología , MicroARNs/metabolismo , Osteogénesis/genética , Semaforina-3A/metabolismo , Secuencia de Bases , Regulación hacia Abajo/genética , Células HEK293 , Humanos , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...