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











Intervalo de ano de publicação
1.
Curr Med Chem ; 31(29): 4687-4702, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39081025

RESUMO

BACKGROUND: Long non-coding RNA (lncRNA) is a class of single-stranded RNA biomolecules involving over 200 nucleotides and does not encode proteins. Research on lncRNA has become a hot spot for the past few years. DNM3OS (Dynamin 3 Opposite Strand), which has been clearly identified as a regulatory lncRNA, exerts an integral role in the pathophysiology of multiple human diseases. OBJECTIVE: The current review study summarizes the pathogenic mechanism of DNM3OS in various pathophysiological processes, aiming to reveal its important value as a therapeutic drug target for related human diseases and provide a new way for targeted therapy. METHODS: Through systematic retrieval and in-depth study of relevant articles in PubMed, this article analyzes and summarizes the pathogenic roles and molecular mechanisms in pathophysiological processes of long non-coding RNA DNM3OS. RESULTS: DNM3OS exerts an important regulatory role in the occurrence and development of bone diseases, neoplastic diseases, fibrotic diseases, inflammatory diseases, and many other diseases. CONCLUSION: DNM3OS is a potential new biomarker and therapeutic target for the treatment of a series of diseases, consisting of bone diseases, neoplastic diseases, fibrotic diseases, and inflammatory diseases.


Assuntos
Neoplasias , RNA Longo não Codificante , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Humanos , Neoplasias/genética , Neoplasias/patologia , Neoplasias/metabolismo , Inflamação/metabolismo , Doenças Ósseas/genética , Doenças Ósseas/metabolismo , Fibrose , Animais
2.
JBMR Plus ; 8(5): ziae051, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38686038

RESUMO

Genome wide association study (GWAS)-implicated bone mineral density (BMD) signals have been shown to localize in cis-regulatory regions of distal effector genes using 3D genomic methods. Detailed characterization of such genes can reveal novel causal genes for BMD determination. Here, we elected to characterize the "DNM3" locus on chr1q24, where the long non-coding RNA DNM3OS and the embedded microRNA MIR199A2 (miR-199a-5p) are implicated as effector genes contacted by the region harboring variation in linkage disequilibrium with BMD-associated sentinel single nucleotide polymorphism, rs12041600. During osteoblast differentiation of human mesenchymal stem/progenitor cells (hMSC), miR-199a-5p expression was temporally decreased and correlated with the induction of osteoblastic transcription factors RUNX2 and Osterix. Functional relevance of miR-199a-5p downregulation in osteoblastogenesis was investigated by introducing miR-199a-5p mimic into hMSC. Cells overexpressing miR-199a-5p depicted a cobblestone-like morphological change and failed to produce BMP2-dependent extracellular matrix mineralization. Mechanistically, a miR-199a-5p mimic modified hMSC propagated normal SMAD1/5/9 signaling and expressed osteoblastic transcription factors RUNX2 and Osterix but depicted pronounced upregulation of SOX9 and enhanced expression of essential chondrogenic genes ACAN, COMP, and COL10A1. Mineralization defects, morphological changes, and enhanced chondrogenic gene expression associated with miR-199a-5p mimic over-expression were restored with miR-199a-5p inhibitor suggesting specificity of miR-199a-5p in chondrogenic fate specification. The expression of both the DNM3OS and miR-199a-5p temporally increased and correlated with hMSC chondrogenic differentiation. Although miR-199a-5p overexpression failed to further enhance chondrogenesis, blocking miR-199a-5p activity significantly reduced chondrogenic pellet size, extracellular matrix deposition, and chondrogenic gene expression. Taken together, our results indicate that oscillating miR-199a-5p levels dictate hMSC osteoblast or chondrocyte terminal fate. Our study highlights a functional role of miR-199a-5p as a BMD effector gene at the DNM3 BMD GWAS locus, where patients with cis-regulatory genetic variation which increases miR-199a-5p expression could lead to reduced osteoblast activity.

3.
J Thorac Dis ; 16(2): 1234-1246, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38505042

RESUMO

Background: Idiopathic pulmonary fibrosis (IPF) is an unrepairable disease that results in lung dysfunction and decreased quality of life. Prevention of pulmonary fibrosis is challenging, while its pathogenesis remains largely unknown. Herein, we investigated the effect and mechanism of long non-coding RNA (lncRNA) DNM3OS/Antisense RNA in the pathogenesis of pulmonary fibrosis. Methods: EdU (5-ethynyl-2'-deoxyuridine) and wound healing assays were employed to evaluate the role of DNM3OS on cell proliferation and migration. Western blot detected the proteins expressions of alpha-smooth muscle actin (α-SMA), vimentin, and fibronectin. The interactions among genes were evaluated by RNA pull-down, luciferase reporter, RNA immunoprecipitation (RIP), chromatin immunoprecipitation (ChIP) and chromatin Isolation by RNA purification (ChIRP) assays. Results: DNM3OS was upregulated by transforming growth factor beta 1 (TGF-ß1) in a dose- and time-dependent manner. DNM3OS knockdown repressed the growth and migration of lung fibroblast, and fibrotic gene expression (CoL1α1, CoL3α1, α-SMA, vimentin, and fibronectin), while suppression of TSC2 accelerated the above process. DNM3OS recruited EZH2 to the promoter region of TSC2, increased the occupancy of EZH2 and H3K27me3, and thereby suppressed the expression of TSC2. HOXA5 promoted the transcription of DNM3OS. Conclusions: HOXA5-induced DNM3OS promoted the proliferation, migration, and expression of fibrosis-related genes in human embryo lung fibroblast via recruiting EZH2 to epigenetically suppress the expression of TSC2.

4.
Noncoding RNA Res ; 9(2): 294-306, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38505310

RESUMO

Spinal facet joint osteoarthritis (FJOA) is an OA disease with pathogenesis and progression uncovered. Our present study was performed to elucidate the role of DNM3OS on spinal FJOA. In this study, spine facet joint tissue of patients were collected. In vitro and in vivo models were constructed with SW1353 cells and rats. Hematoxylin and eosin (HE) staining, Safranin O-fast Green, Alcian blue staining, and Tolueine blue O (TBO) staining were employed for histology analyses. Quantitative PCR, western blotting, and Immunofluorescence were performed to evaluate the expression of genes. The levels of inflammatory cytokines were measured by enzyme-linked immunosorbent assay analysis. Cell Counting Kit-8 and flow cytometry were used for cell activity and apoptosis evaluation. The targeting sites between microRNA (miR)-127-5p and cadherin 11 (CDH11) were predicted TargetScan and miRbase database and confirmed by Dual-luciferase reporter assays. CHIP and EMS assay were employed to confirm the binding of LEF1and DNM3OS promoter. Our results showed that DNM3OS was found to upregulated, while miR-127-5p was downregulated in severe FJOA patients and inflammation-induced chondrosarcoma SW1353 cells. DNM3OS reduced cell activity, induced cell apoptosis and extracellular matrix (ECM) degradation by sponging miR-127-5p in vitro. miR-127-5p targeted CDH11 and inhibited wnt3a/ß-catenin pathway to regulate OA in vitro. LEF1 promoted DNM3OS transcription to form a positively feedback in activated wnt3a/ß-catenin pathway. In vivo rat model also confirmed that DNM3OS aggravated FJOA. In summary, DNM3OS/miR-127-5p/CDH11 enhanced Wnt3a/ß-Catenin/LEF-1 pathway to form a positive feedback and aggravate spinal FJOA.

5.
Pathol Res Pract ; 235: 153961, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35653924

RESUMO

There is increasing evidence that show long noncoding RNAs including long intergenic noncoding RNAs (lincRNA) play a pivotal regulatory role in the biological processes. Differential expression of lincRNAs can be utilized for cancer diagnosis, prognosis, and targeted therapy. Little is known about their expressions in urothelial tumors. Concerning the potential role of lincRNAs in cancer development, we aimed to investigate the expression levels of LINC00958 and DNM3OS in bladder cancer. Fifty tumor and 50 adjacent non-tumor tissue samples along with their clinicopathological parameters were obtained from bladder cancer patients. Expressions of LINC00958 and DNM3OS were analyzed by Real-time PCR. ROC curve analysis was used to evaluate the diagnostic power of LINC0095 and DNM3OS for BC. Expression level of LINC00958 was considerably increased in cancerous tissues (P < 0.001) and in correlation with cigarette smoking (P = 0.043). DNM3OS expression was higher in the tumor tissues than normal tissues (P < 0.001) and showed a significant association with age (P = 0.038). By using the ROC curve, the diagnostic power of LINC00958 and DNM3OS transcript levels in bladder cancer were estimated to be 87% and 75%, respectively. Our findings offer some important intuitions into the oncogenic role of LINC00958 and DNM3OS in bladder cancer and suggest that they can be candidate biomarkers and may provide new approaches for the diagnosis and therapy if being validated in a larger sample size of clinical samples as well as functional studies.


Assuntos
RNA Longo não Codificante , Neoplasias da Bexiga Urinária , Carcinogênese/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Oncogenes , Prognóstico , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Neoplasias da Bexiga Urinária/diagnóstico , Neoplasias da Bexiga Urinária/genética
6.
Biochem Biophys Res Commun ; 583: 35-42, 2021 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-34717123

RESUMO

Atherosclerosis in diabetes is a leading cause of cardiovascular complications. Intermedin (IMD) is a calcitonin peptide that is known to inhibit macrophage phagocytosis in atherosclerosis, but the exact mechanism is unclear. We investigate genes that are differentially expressed in response to IMD in hyperglycemic conditions and determine whether they delay the progression of atherosclerosis. An atherosclerotic and diabetic-murine model was generated in 8-week-old male ApoE-/- mice receiving streptozotocin and a high-fat diet. The mouse model was treated with IMD and the expression levels of NF-κB, Dnm3os, miR-27b-3p, and SLAMF7 were detected in plaque tissue and macrophages cultured with high glucose concentrations. Phagocytosis was determined by oxidized-low-density lipoprotein (Ox-LDL) uptake and the interactions among Dnm3os, SLAMF7 and miR-27b-3p were assessed by dual-luciferase reporter assays. The expression of NF-κB, Dnm3os, and SLAMF7 was enhanced in atherosclerotic plaques but decreased by IMD. The suppression of Dnm3os reduced plaque formation in IMD-treated mice even further whereas increased by miR-27b-3p. Dnm3os and SLAMF7 were competitively bind to miR-27b-3p in vivo. In vitro, ox-LDL uptake is elevated in macrophages cultured in hyperglycemic conditions but reduced by IMD. Dual-luciferase assays indicate that Dnm3os positively regulates SLAMF7 through miR-27b-3p expression. In conclusion, Dnm3os is involved in macrophage phagocytosis through the competitive binding of SLAMF7 with miR-27b-3p. IMD induces the suppression of Dnm3os to inhibit macrophage phagocytosis and alleviate atherosclerosis in diabetes.

7.
J Cell Mol Med ; 25(18): 9066-9071, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34369082

RESUMO

Emerging studies have suggested that dysregulated long non-coding RNAs (lncRNAs) are associated with the pathogenesis of neurodegenerative diseases (NDD) including Huntington's disease (HD); however, the pathophysiological mechanism by which lncRNA dysregulation participates in HD remains to be elucidated. Here, we aim to analyse the expression of lncRNA-DNM3OS and identify the possible DNM3OS/miR-196b-5p/GAPDH pathway. PC12 cells induced by rat pheochromocytoma expressing HD gene exon 1 fragment with either 23 or 74 polyglutamine repeats fused to the green fluorescent protein (GFP) were cultured. Our results show that GAPDH and DNM3OS were upregulated in HD PC12 cells, downregulation of which lead to inhibition of aggregate formation accompanied by a decreased apoptosis rate and increased relative ROS levels and cell viability. Moreover, upregulated DNM3OS decreased the expression of miR-196b-5p by sponging, and GAPDH was a direct target of miR-196b-5p, playing an important pathogenic role in the formation of aggregates in the HD cell model. Our study uncovers a novel DNM3OS/miR-196b-5p/GAPDH pathway involved in the molecular pathogenesis of HD, which may offer a potential therapeutic strategy for HD.


Assuntos
Doença de Huntington/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Apoptose , Sobrevivência Celular , Células PC12 , Ratos
8.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 38(3): 574-582, 2021 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-34180204

RESUMO

Long non-coding RNA (lncRNA) Dnm3os plays a critical role in peritendinous fibrosis and pulmonary fibrosis, but its role in the process of cardiac fibrosis is still unclear. Therefore, we carried out study by using the myocardial fibrotic tissues obtained by thoracic aortic constriction (TAC) in an early study of our group, and the in vitro cardiac fibroblast activation model induced by transforming growth factor-ß1 (TGF-ß1). Quantitative real-time polymerase chain reaction (RT-qPCR), Western blot, and collagen gel contraction test were used to identify the changes of activation phenotype and the expression of Dnm3os in cardiac fibroblasts. Small interfering RNA was used to silence Dnm3os to explore its role in the activation of cardiac fibroblasts. The results showed that the expression of Dnm3os was increased significantly in myocardial fibrotic tissues and in the activated cardiac fibroblasts. And the activation of cardiac fibroblasts could be alleviated by Dnm3os silencing. Furthermore, the TGF-ß1/Smad2/3 pathway was activated during the process of cardiac fibroblasts activation, while was inhibited after silencing Dnm3os. The results suggest that Dnm3os silencing may affect the process of cardiac fibroblast activation by inhibiting TGF-ß1/Smad2/3 signal pathway. Therefore, interfering with the expression of lncRNA Dnm3os may be a potential target for the treatment of cardiac fibrosis.


Assuntos
RNA Longo não Codificante , Fibroblastos , Fibrose , Humanos , Miocárdio/patologia , Transdução de Sinais , Fator de Crescimento Transformador beta1
9.
Yonsei Med J ; 62(6): 535-544, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34027641

RESUMO

PURPOSE: Long non-coding RNAs (lncRNAs) are essential regulators in the development of ovarian cancer (OC). Nonetheless, the function of lncRNA DNM3 opposite strand/antisense RNA (DNM3OS) in OC remains unclear. This work aimed to investigate the biological roles and underlying mechanisms of DNM3OS in OC. MATERIALS AND METHODS: Quantitative real-time polymerase chain reaction was conducted to examine DNM3OS, microRNA (miR)-193a-3p, and mitogen-activated protein kinase 3 (MAP3K3) mRNA expression in OC tissues and cell lines. Kaplan-Meier survival analysis was employed to analyze the relationship between DNM3OS expression and the prognosis of OC patients. Cell counting kit-8, 5-ethynyl-2'-deoxyuridine, and transwell experiments were conducted to monitor cell proliferation, migration, and invasion, respectively. Western blot was applied to examine epithelial-mesenchymal transition associated protein (E-cadherin and N-cadherin) expression. Luciferase reporter gene and RNA immunoprecipitation experiments were performed to confirm the relationships among DNM3OS, miR-193a-3p, and MAP3K3. Pearson's correlation analysis was adopted to analyze the correlations among DNM3OS, miR-193a-3p, and MAP3K3 mRNA. RESULTS: DNM3OS expression was remarkably increased in OC tissues and cell lines, which was associated with the unfavorable prognosis of the patients. DNM3OS overexpression enhanced OC cell proliferation, migration, and invasion; suppressed E-cadherin protein expression; and facilitated N-cadherin protein expression, while the transfection of miR-193a-3p mimics had the opposite effects. DNM3OS directly interacted with miR-193a-3p, and miR-193a-3p targeted MAP3K3 by directly binding to 3'UTR. DNM3OS could up-regulate the expression of MAP3K3 via repressing miR-193a-3p expression. CONCLUSION: DNM3OS, as an oncogenic lncRNA, increases the malignancy of OC cells via regulation of an miR-193a-3p/MAP3K3 axis.


Assuntos
MicroRNAs , Neoplasias Ovarianas , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Humanos , MAP Quinase Quinase Quinase 3 , MicroRNAs/genética , Proteína Quinase 3 Ativada por Mitógeno , Neoplasias Ovarianas/genética , RNA Antissenso
10.
Cell Mol Biol Lett ; 26(1): 22, 2021 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-34049478

RESUMO

BACKGROUND: Improved chondrogenic differentiation of mesenchymal stem cells (MSCs) by genetic regulation is a potential method for regenerating articular cartilage. MiR-127-5p has been reported to promote cartilage differentiation of rat bone marrow MSCs (rMSCs); however, the regulatory mechanisms underlying hypoxia-stimulated chondrogenic differentiation remain unknown. METHODS: rMSCs were induced to undergo chondrogenic differentiation under normoxic or hypoxic conditions. Expression of lncRNA DNM3OS, miR-127-5p, and GREM2 was detected by quantitative real-time PCR. Proteoglycans were detected by Alcian blue staining. Western blot assays were performed to examine the relative levels of GREM2 and chondrogenic differentiation related proteins. Luciferase reporter assays were performed to assess the association among DNM3OS, miR-127-5p, and GREM2. RESULTS: MiR-127-5p levels were upregulated, while DNM3OS and GREM2 levels were downregulated in rMSCs induced to undergo chondrogenic differentiation, and those changes were attenuated by hypoxic conditions (1% O2). Further in vitro experiments revealed that downregulation of miR-127-5p reduced the production of proteoglycans and expression of chondrogenic differentiation markers (COL1A1, COL2A1, SOX9, and ACAN) and osteo/chondrogenic markers (BMP-2, p-SMAD1/2). MiR-127-5p overexpression produced the opposite results in rMSCs induced to undergo chondrogenic differentiation under hypoxic conditions. GREM2 was found to be a direct target of miR-127-5p, which was suppressed in rMSCs undergoing chondrogenic differentiation. Moreover, DNM3OS could directly bind to miR-127-5p and inhibit chondrogenic differentiation of rMSCs via regulating GREM2. CONCLUSIONS: Our study revealed a novel molecular pathway (DNM3OS/miR-127-5p/GREM2) that may be involved in hypoxic chondrogenic differentiation.


Assuntos
Condrogênese , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , Proteínas/genética , RNA Longo não Codificante/genética , Animais , Diferenciação Celular , Hipóxia Celular , Células Cultivadas , Regulação da Expressão Gênica , Masculino , Células-Tronco Mesenquimais/metabolismo , Ratos
11.
Cell Biosci ; 11(1): 47, 2021 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-33653390

RESUMO

BACKGROUND: Skeletal development and maintenance are complex processes known to be coordinated by multiple genetic and epigenetic signaling pathways. However, the role of long non-coding RNAs (lncRNAs), a class of crucial epigenetic regulatory molecules, has been under explored in skeletal biology. RESULTS: Here we report a young patient with short stature, hypothalamic dysfunction and mild macrocephaly, who carries a maternally inherited 690 kb deletion at Chr.1q24.2 encompassing a noncoding RNA gene, DNM3OS, embedded on the opposite strand in an intron of the DYNAMIN 3 (DNM3) gene. We show that lncRNA DNM3OS sustains the proliferation of chondrocytes independent of two co-cistronic microRNAs miR-199a and miR-214. We further show that nerve growth factor (NGF), a known factor of chondrocyte growth, is a key target of DNM3OS-mediated control of chondrocyte proliferation. CONCLUSIONS: This work demonstrates that DNM3OS is essential for preventing premature differentiation of chondrocytes required for bone growth through endochondral ossification.

12.
Cancer Lett ; 503: 19-31, 2021 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-33472090

RESUMO

Tumor-associated mesenchymal stem cells (MSCs) play a critical role in the growth and metastasis of hepatocellular carcinoma (HCC). However, the mechanism underlying the crosstalk between MSCs and HCC cells is not completely understood. Here, HCC cells were treated with or without conditioned medium of MSCs (CM-MSC), and examined for differential expression of long non-coding RNAs (lncRNAs). Knockdown and overexpression experiments were conducted to explore the function of the lncRNA DNM3OS in MSC-induced HCC growth and metastasis. CM-MSC treatment led to a concentration-dependent induction of DNM3OS in HCC cells. DNM3OS was significantly upregulated in HCC compared to adjacent liver tissues. High DNM3OS expression was associated with TNM stage, vascular invasion, and poor prognosis of HCC patients. Silencing of DNM3OS inhibited HCC cell proliferation and invasion in vitro and tumorigenesis and metastasis in vivo. Overexpression of DNM3OS enhanced HCC cell proliferation, invasion, and metastasis. Biochemically, DNM3OS was mainly localized in the nucleus and physically interacted with KDM6B. The association of DNM3OS with KDM6B induced the expression of TIAM1 through reduction of H3K27me3 at the TIAM1 promoter. TIAM1 overexpression restored the proliferation and invasion of DNM3OS-depleted HCC cells. Our data delineate a mechanism by which MSCs accelerate HCC growth and metastasis through a DNM3OS/KDM6B/TIAM1 axis.


Assuntos
Carcinoma Hepatocelular/patologia , Histona Desmetilases com o Domínio Jumonji/genética , Neoplasias Hepáticas/patologia , Células-Tronco Neoplásicas/química , RNA Longo não Codificante/genética , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T/genética , Animais , Carcinoma Hepatocelular/genética , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Meios de Cultivo Condicionados/química , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Neoplasias Hepáticas/genética , Masculino , Camundongos , Metástase Neoplásica , Transplante de Neoplasias
13.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-920478

RESUMO

@#[摘 要] 目的:检测lncRNA DNM3OS在喉鳞状细胞癌(laryngeal squamous cell carcinoma,LSCC)组织和LSCC细胞株中的表达及其临床意义,探讨其对LSCC TU177细胞体外增殖、迁移及侵袭的影响,并分析DNM3OS与EMT的关系。方法:从河北医科大学第四医院生物标本库选取2014年3月至2018年12月收治的68例LSCC患者手术切除的癌及癌旁组织标本,应用qPCR法检测DNM3OS在LSCC组织和细胞株中的表达水平。采用siRNA敲低TU177细胞中DNM3OS的表达,应用MTS、克隆形成及Transwell小室等方法分别检测敲低DNM3OS表达对TU177细胞增殖、迁移和侵袭等生物学行为的影响。应用qPCR和WB法检测转染si-DNM3OS后对EMT标志物上皮钙黏素(E-cadherin)、神经钙黏素(N-cadherin)、波形蛋白(vimentin)、扭曲蛋白(twist)、锌指转录因子2(SNAI2)mRNA和蛋白的变化。结果:LSCC组织中DNM3OS表达水平明显高于癌旁组织(P<0.01),并与患者的TNM分期、淋巴结转移及生存期有关联(P<0.05或P<0.01)。DNM3OS在LSCC细胞株(Hep-2、AMC-HN-8、TU177、TU212及TU686)中均呈现不同程度的高表达(P<0.05或P<0.01),转染si-DNM3OS后TU177细胞中DNM3OS的表达显著降低(P<0.01)。与对照组相比,DNM3OS表达敲低可抑制TU177细胞的体外增殖、迁移和侵袭能力(P<0.05或P<0.01),可上调TU177细胞中E-cadherin的表达而下调N-cadherin、vimentin、twist和SNAI2的表达(均P<0.01)。结论: DNM3OS高表达与LSCC的恶性进展有关,其可能为预测LSCC患者预后的潜在指标;DNM3OS可能通过影响EMT进程促进LSCC细胞的侵袭和转移。

14.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-888215

RESUMO

Long non-coding RNA (lncRNA) Dnm3os plays a critical role in peritendinous fibrosis and pulmonary fibrosis, but its role in the process of cardiac fibrosis is still unclear. Therefore, we carried out study by using the myocardial fibrotic tissues obtained by thoracic aortic constriction (TAC) in an early study of our group, and the


Assuntos
Humanos , Fibroblastos , Fibrose , Miocárdio/patologia , RNA Longo não Codificante , Transdução de Sinais , Fator de Crescimento Transformador beta1
15.
Methods Mol Biol ; 2176: 49-56, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32865781

RESUMO

Long noncoding RNAs (lncRNAs) are a class of RNA with 200 nucleotides or longer that are not translated into protein. lncRNAs are highly abundant; a study estimates that at least four times more lncRNAs are typically present than coding RNAs in humans. However, function of more than 95% of human lncRNAs are still unknown. Synthetic antisense oligonucleotides called gapmers are powerful tools for lncRNA loss-of-function studies. Gapmers contain a central DNA part, which activates RNase H-mediated RNA degradation, flanked by modified oligonucleotides, such as 2'-O-methyl RNA (2'OMe), 2'-O-methoxyethyl RNA (2'MOE), constrained ethyl nucleosides (cEt), and locked nucleic acids (LNAs). In contrast to siRNA or RNAi-based methods, antisense oligonucleotide gapmer-based knockdown is often more effective against nuclear-localized lncRNA targets, since RNase H is mainly localized in nuclei. As such, gapmers are also potentially a powerful tool for therapeutics targeting lncRNAs in various diseases, including cancer, cardiovascular diseases, lung fibrosis, and neurological/neuromuscular diseases. This chapter will discuss the development and applications of gapmers for lncRNA loss-of-function studies and tips to design effective antisense oligonucleotides.


Assuntos
Técnicas de Silenciamento de Genes/métodos , Oligonucleotídeos Antissenso , RNA Longo não Codificante/genética , Animais , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/terapia , Técnicas de Silenciamento de Genes/história , Terapia Genética/história , Terapia Genética/métodos , História do Século XX , História do Século XXI , Humanos , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/terapia , Neoplasias/genética , Neoplasias/terapia , Oligonucleotídeos Antissenso/síntese química , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/uso terapêutico , Fibrose Pulmonar/genética , Fibrose Pulmonar/terapia , RNA Longo não Codificante/antagonistas & inibidores
16.
J Oral Pathol Med ; 49(9): 865-875, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32463958

RESUMO

BACKGROUND: Non-coding RNAs play a critical role in the occurrence and development of oral cancer. The present study is aimed to identify long non-coding RNA (lncRNA) that might be novel effective targets for the treatments of oral cancer and the underlying mechanism. METHODS: The microarray profiling and RNA-sequencing analysis were performed to identify lncRNAs related to oral cancer development, and lncRNA DNM3OS was selected. DNM3OS knockdown was generated in cancer cell lines, and the specific effects of DNM3OS knockdown on cell phenotype were examined. DNM3OS targeted miRNA and miRNA targeted downstream mRNA were selected, the predicted bindings were verified, and the specific effects of miRNA on oral cancer cells were examined. Finally, the dynamic effects of DNM3OS and miRNA on target mRNA expression and oral cancer cell phenotype were examined. RESULTS: DNM3OS was upregulated in oral cancer tissues and cells. DNM3OS knockdown in CAL27 and SCC-9 cells inhibited cell viability and migration. DNM3OS targeted miR-204-5p to inhibit miR-204-5p expression. miR-204-5p overexpression suppressed oral cancer cell aggressiveness. miR-204-5p targeted HIP1 to inhibit HIP1 expression. HIP1 knockdown inhibited oral cancer cell viability and migration. The effects of DNM3OS knockdown were significantly reversed by miR-204-5p inhibition. Within oral carcinoma tissue samples, expression of DNM3OS and HIP1 was increased whereas the miR-204-5p expression was downregulated; miR-204-5p had a negative correlation with DNM3OS and HIP1, respectively, while DNM3OS and HIP1 were positively correlated with each other. CONCLUSION: Long non-coding RNA DNM3OS, miR-204-5p, and HIP1 form an axis that modulates oral cancer cell viability and migration.


Assuntos
MicroRNAs , Neoplasias Bucais , RNA Longo não Codificante , Sobrevivência Celular/genética , Humanos , MicroRNAs/genética , Neoplasias Bucais/genética , RNA Longo não Codificante/genética
17.
Cancer Cell Int ; 20: 23, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31992960

RESUMO

BACKGROUND: Retinoblastoma (RB) is acknowledged to be the commonest intraocular malignancy in infants and children and the outcome of RB patients is unfavorable due to limited early diagnosis and effective therapy. SMAD family member 6 (SMAD6) has been reported in the initiation and progression of human cancers by acting as a biological participant. However, the role of SMAD6 in RB has not been illustrated yet. METHODS: The expression of SMAD6 mRNA, miR-134-5p and DNM3OS was measured by RT-qPCR. SMAD6 protein levels were measured by western blot. The effects of SMAD6 depletion on RB cells were analyzed using CCK-8 and transwell assays. The key proteins related to epithelial-mesenchymal transition (EMT) was determined by western blot. The localization of DNM3OS was detected by nuclear/cytoplasmic assay. In addition, the direct interaction between miR-134-5p and SMAD6 or DNM3OS was confirmed with the application of dual-luciferase reporter assay. RESULTS: SMAD6 was upregulated in RB tissue samples and cell lines, and silencing SMAD6 suppressed cell proliferation, migration and EMT in RB. Mechanically, SMAD6 was positively regulated by lncRNA DNM3OS through competitively interacting with miR-134-5p. DNM3OS contributed to RB progression by SMAD6-mediated manner. CONCLUSIONS: This research unmasked a novel DNM3OS/miR-134-5p/SMAD6 pathway in RB, which might make contribution to treatment of RB.

18.
Aging (Albany NY) ; 11(21): 9442-9460, 2019 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-31694982

RESUMO

Transforming growth factor-ß1 (TGFß1)-induced differentiation into and the activation of myofibroblasts have been regarded as critical events in benign prostatic hyperplasia (BPH); however, the underlying mechanisms of BPH pathogenesis remain unclear. Microarray profiling, STRING analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation, and Gene Ontology (GO) enrichment analysis were performed to confirm the candidate genes and long non-coding RNA (lncRNAs) related to BPH. Collagen Type III (COL3A1) was significantly upregulated by TGFß1 in prostate stromal cells (PrSCs) and might be involved in DNM3OS function in myofibroblasts upon TGFß1 stimulation. Upon TGFß1 stimulation, COL3A1 protein was decreased by DNM3OS silencing. miR-29a and miR-29b could directly bind to the DNM3OS and COL3A1 3' untranslated region (UTR)s to negatively regulate their expression, and by serving as a competing endogenous RNAs (ceRNA), DNM3OS competed with COL3A1 for miR-29a/29b binding, therefore counteracting miR-29a/29b-mediated COL3A1 suppression. The effect of DNM3OS silencing on ECM components and TGFß1 downstream signaling was similar to that of the TGFß1 inhibitor SB431542. miR-361 could target DNM3OS and TGFß1; DNM3OS competed for miR-361 binding to counteract miR-361-mediated TGFß1 suppression. In conclusion, we identified DNM3OS as a specifically-upregulated lncRNA upon TGFß1 stimulation in PrSCs; by serving as a ceRNA for the miR-29a/29b cluster and miR-361, DNM3OS eliminated miRNA-mediated suppression of COL3A1 and TGFß1, thereby promoting TGFß1-induced PrSC transformation into myofibroblasts.


Assuntos
MicroRNAs/metabolismo , Hiperplasia Prostática/etiologia , Hiperplasia Prostática/metabolismo , RNA Longo não Codificante/metabolismo , Idoso , Colágeno Tipo III/metabolismo , Matriz Extracelular/metabolismo , Perfilação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Miofibroblastos , Análise de Sequência com Séries de Oligonucleotídeos , Cultura Primária de Células , Próstata/metabolismo , Próstata/patologia , Células Estromais/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
19.
Biochem Biophys Res Commun ; 511(1): 57-62, 2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30770102

RESUMO

Long non-coding RNAs (lncRNAs) act as tumor suppressors or oncogenes in tumor development and progression. In the present study, we explored the expression and biological role of the lncRNA DNM3OS in gastric cancer (GC). We observed that DNM3OS was upregulated in GC tissues and cell lines, and high DNM3OS expression was correlated with malignant features and served as an indicator of a poor prognosis for GC patients. DNM3OS knockdown inhibited the proliferation of GC cells, and reduced DNM3OS suppressed tumor growth in vivo. Moreover, DNM3OS depletion inhibited the migration and invasion of GC cells through the suppression of the Snail-mediated epithelial-mesenchymal transition (EMT). In conclusion, we demonstrated that DNM3OS serves as an oncogenic lncRNA in GC, and we implicated DNM3OS as a promising prognostic factor and a potential therapeutic target for GC patients.


Assuntos
Transição Epitelial-Mesenquimal , Invasividade Neoplásica/genética , RNA Longo não Codificante/genética , Fatores de Transcrição da Família Snail/genética , Neoplasias Gástricas/genética , Animais , Linhagem Celular Tumoral , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos Nus , Invasividade Neoplásica/patologia , Neoplasias Gástricas/patologia
20.
RNA Biol ; 12(3): 343-53, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25826666

RESUMO

microRNA is necessary for osteoclast differentiation, function and survival. It has been reported that miR-199/214 cluster plays important roles in vertebrate skeletal development and miR-214 inhibits osteoblast function by targeting ATF4. Here, we show that miR-214 is up-regulated during osteoclastogenesis from bone marrow monocytes (BMMs) with macrophage colony stimulating factor (M-CSF) and receptor activator of nuclear factor-κB ligand (RANKL) induction, which indicates that miR-214 plays a critical role in osteoclast differentiation. Overexpression of miR-214 in BMMs promotes osteoclastogenesis, whereas inhibition of miR-214 attenuates it. We further find that miR-214 functions through PI3K/Akt pathway by targeting phosphatase and tensin homolog (Pten). In vivo, osteoclast specific miR-214 transgenic mice (OC-TG214) exhibit down-regulated Pten levels, increased osteoclast activity, and reduced bone mineral density. These results reveal a crucial role of miR-214 in the differentiation of osteoclasts, which will provide a potential therapeutic target for osteoporosis.


Assuntos
MicroRNAs/genética , Osteoclastos/metabolismo , Osteoporose/genética , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Animais , Sequência de Bases , Densidade Óssea , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Bovinos , Diferenciação Celular , Regulação da Expressão Gênica , Fator Estimulador de Colônias de Macrófagos/farmacologia , Camundongos , Camundongos Transgênicos , MicroRNAs/agonistas , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Dados de Sequência Molecular , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Oligonucleotídeos/genética , Oligonucleotídeos/metabolismo , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteoporose/metabolismo , Osteoporose/patologia , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ligante RANK/farmacologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA