lncRNA H19

LncRNA H19
  • 文章类型: Journal Article
    肺癌在全球范围内一直是癌症相关死亡的主要原因。此外,它在所有类型的癌症中显示出最高的死亡率。5年内的生存率不到20%,主要是由于这种疾病通常在晚期被诊断出来,导致与早期阶段相比,治疗方案效果较差。原发性肺癌有两种主要类型:非小细胞肺癌,约占所有病例的80%-85%,和小细胞肺癌,根据癌症起源的特定细胞类型进行分类。对这种疾病生物学的理解和致癌驱动因素改变的识别已经显着改变了治疗方法的格局。长链非编码RNA(lncRNAs)通过不同的分子机制在调节各种生理和病理过程中起着至关重要的作用。在这些lncRNAs中,lncRNAH19,最初被鉴定为肿瘤胎儿转录本,由于其在许多肿瘤中的表达升高,因此引起了极大的关注。广泛的研究已经证实其参与肿瘤发生和恶性进展通过促进细胞生长,入侵,迁移,上皮-间质转化,转移,和治疗抵抗。这篇全面的综述旨在概述lncRNAH19的异常过表达及其有助于肺癌进展的分子途径。这篇综述的发现强调了进一步研究这种疾病的诊断和治疗的潜力,为未来的研究提供了有希望的途径。
    Lung cancer holds the position of being the primary cause of cancer-related fatalities on a global scale. Furthermore, it exhibits the highest mortality rate among all types of cancer. The survival rate within a span of 5 years is less than 20%, primarily due to the fact that the disease is often diagnosed at an advanced stage, resulting in less effective treatment options compared to earlier stages. There are two main types of primary lung cancer: nonsmall-cell lung cancer, which accounts for approximately 80%-85% of all cases, and small-cell lung cancer, which is categorized based on the specific type of cells in which the cancer originates. The understanding of the biology of this disease and the identification of oncogenic driver alterations have significantly transformed the landscape of therapeutic approaches. Long noncoding RNAs (lncRNAs) play a crucial role in regulating various physiological and pathological processes through diverse molecular mechanisms. Among these lncRNAs, lncRNA H19, initially identified as an oncofetal transcript, has garnered significant attention due to its elevated expression in numerous tumors. Extensive research has confirmed its involvement in tumorigenesis and malignant progression by promoting cell growth, invasion, migration, epithelial-mesenchymal transition, metastasis, and therapy resistance. This comprehensive review aims to provide an overview of the aberrant overexpression of lncRNA H19 and the molecular pathways through which it contributes to the advancement of lung cancer. The findings of this review highlight the potential for further investigation into the diagnosis and treatment of this disease, offering promising avenues for future research.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    长链非编码RNA(lncRNA)H19是一种广泛研究的lncRNA,与许多病理变化有关。我们以前的发现已经证明,慢性肾病患者血清lncRNAH19水平降低,lncRNAH19降低与肾小管间质纤维化密切相关。发展终末期肾病的重要步骤。尽管如此,lncRNAH19在肾小管间质纤维化中的确切功能和机制尚未完全理解。本工作利用单侧输尿管梗阻(UUO)和转化生长因子-β1(TGF-β1)刺激的HK-2细胞的小鼠模型来研究lncRNAH19在肾小管间质纤维化中的可能作用和机制。用TGF-β1刺激UUO和HK-2细胞的小鼠肾脏中lncRNAH19的水平降低。lncRNAH19在小鼠肾脏中的上调显著减轻了肾损伤,由UUO引发的纤维化和炎症。此外,HK-2细胞中lncRNAH19的增加减少了TGF-β1诱导的上皮-间质转化(EMT)。值得注意的是,lncRNAH19的上调降低了UUO和TGF-β1刺激的HK-2细胞小鼠肾脏中的脂质积累和三酰甘油含量,伴随着长链酰基辅酶A合成酶1(ACSL1)的上调。lncRNAH19被鉴定为microRNA-130a-3p的海绵,lncRNAH19通过它调节ACSL1的表达。microRNA-130a-3p的过表达逆转了lncRNAH19诱导的ACSL1表达增加。lncRNAH19过表达对EMT的抑制作用,ACSL1沉默或microRNA-130a-3p过表达减少了HK-2细胞中的炎症和脂质积累。总的来说,研究结果表明,lncRNAH19通过调节microRNA-130a-3p/ACSL1轴减少脂质沉积,从而改善肾小管间质纤维化。
    Long non-coding RNA (lncRNA) H19 is an extensively studied lncRNA that is related to numerous pathological changes. Our previous findings have documented that serum lncRNA H19 levels are decreased in patients with chronic kidney disorder and lncRNA H19 reduction is closely correlated with renal tubulointerstitial fibrosis, an essential step in developing end-stage kidney disease. Nonetheless, the precise function and mechanism of lncRNA H19 in renal tubulointerstitial fibrosis are not fully comprehended. The present work utilized a mouse model of unilateral ureteral obstruction (UUO) and transforming growth factor-β1 (TGF-β1)-stimulated HK-2 cells to investigate the possible role and mechanism of lncRNA H19 in renal tubulointerstitial fibrosis were investigated. Levels of lncRNA H19 decreased in kidneys of mice with UUO and HK-2 cells stimulated with TGF-β1. Up-regulation of lncRNA H19 in mouse kidneys remarkably relieved kidney injury, fibrosis and inflammation triggered by UUO. Moreover, the increase of lncRNA H19 in HK-2 cells reduced epithelial-to-mesenchymal transition (EMT) induced by TGF-β1. Notably, up-regulation of lncRNA H19 reduced lipid accumulation and triacylglycerol content in kidneys of mice with UUO and TGF-β1-stimulated HK-2 cells, accompanied by the up-regulation of long-chain acyl-CoA synthetase 1 (ACSL1). lncRNA H19 was identified as a sponge of microRNA-130a-3p, through which lncRNA H19 modulates the expression of ACSL1. The overexpression of microRNA-130a-3p reversed the lncRNA H19-induced increases in the expression of ACSL1. The suppressive effects of lncRNA H19 overexpression on the EMT, inflammation and lipid accumulation in HK-2 cells were diminished by ACSL1 silencing or microRNA-130a-3p overexpression. Overall, the findings showed that lncRNA H19 ameliorated renal tubulointerstitial fibrosis by reducing lipid deposition via modulation of the microRNA-130a-3p/ACSL1 axis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    在动脉粥样硬化的背景下,lncRNAH19与铁凋亡之间的精确关联仍不确定。
    这项研究旨在阐明潜在的过程,并提出新的方法来推进针对动脉粥样硬化的治疗干预措施。
    铁沉积的评估,这需要使用CCK-8和细胞内MDA的定量来评估细胞活力,GSH,和亚铁离子。同时,通过蛋白质印迹分析评估的蛋白质表达水平,同时还测定了lncRNAH19的表达水平。此外,用ox-LDL培养的HAEC受到Fer-1干扰。将HAECs暴露于ox-LDL,然后用H19shRNA和H19过表达载体pcDNA3.1转染。然后测量细胞中的铁死亡水平。然后,HAEC与ox-LDL孵育,然后用H19shRNA转染并用Erastin处理以评估铁凋亡水平,细胞活力,和炎症因子的产生。和血管发育的能力。
    ox-LDL组HAECs的存活率要低得多。Ox-LDL导致HAECs中ACSL4表达上调,而SLC7A11和GPX4的表达降低。
    lncRNAH19增强铁凋亡并加剧LDL诱导的动脉内皮细胞损伤。
    UNASSIGNED: The precise association between lncRNA H19 and ferroptosis in the context of atherosclerosis remains uncertain.
    UNASSIGNED: This study is to clarify the underlying process and propose novel approaches for the advancement of therapeutic interventions targeting atherosclerosis.
    UNASSIGNED: Assessment of ferroptosis, which entails the evaluation of cell viability using CCK-8 and the quantification of intracellular MDA, GSH, and ferrous ions. Simultaneously, the protein expression levels of assessed by western blot analysis, while the expression level of lncRNA H19 was also determined. Furthermore, HAECs that were cultured with ox-LDL were subjected to Fer-1 interference. HAECs were exposed to ox-LDL and then transfected with H19 shRNA and H19 overexpression vector pcDNA3.1. The level of ferroptosis in the cells was then measured. Then, HAECs were subjected to incubation with ox-LDL, followed by transfection with H19 shRNA and treated with Erastin to assess the levels of ferroptosis, cell viability, and inflammatory factor production. and the ability for blood vessel development.
    UNASSIGNED: The survival rate of HAECs in the ox-LDL group was much lower. Ox-LDL resulted in an upregulation of ACSL4 expression in HAECs, while the expression of SLC7A11 and GPX4 decreased.
    UNASSIGNED: lncRNA H19 enhances ferroptosis and exacerbates arterial endothelial cell damage induced by LDL.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:外周动脉疾病(PAD)是一种缺血性疾病,在全球范围内发病率不断上升。lncRNAH19(H19)富集在内皮祖细胞(EPCs)中,和焦凋亡抗性H19过表达EPCs(oe-H19-EPCs)的移植可能促进PAD的血管生成和血流恢复,尤其是严重肢体缺血(CLI)。
    方法:使用免疫荧光和流式细胞术对从人外周血中分离的EPC进行表征。用CCK8和EdU测定法测定细胞增殖。通过Transwell和伤口愈合测定评估细胞迁移。使用管形成测定评价血管生成潜力。蛋白质印迹法检测EPCs中的焦亡途径相关蛋白。使用荧光素酶测定法分析miR-107上H19和FADD的结合位点。在体内,Oe-H19-EPCs移植到小鼠缺血肢体模型中,通过激光多普勒成像检测血流。使用全转录组测序检查了oe-H19-EPCs对缺血性肢体的治疗作用背后的转录景观。
    结果:H19在EPC中的过表达导致增殖增加,迁移,和管形成能力。这些作用是通过焦亡途径介导的,受H19/miR-107/FADD轴调控。在小鼠缺血肢体模型中移植oe-H19-EPCs促进血管生成和血流恢复。全转录组测序表明,oe-H19-EPCs治疗后,缺血肢体血管发生途径显著激活。
    结论:H19过表达通过竞争性结合miR-107增加FADD水平,导致增殖增强,迁移,血管生成,和抑制EPCs的焦亡。这些作用最终促进CLI中血流的恢复。
    BACKGROUND: Peripheral artery disease (PAD) is an ischemic disease with a rising incidence worldwide. The lncRNA H19 (H19) is enriched in endothelial progenitor cells (EPCs), and transplantation of pyroptosis-resistant H19-overexpressed EPCs (oe-H19-EPCs) may promote vasculogenesis and blood flow recovery in PAD, especially with critical limb ischemia (CLI).
    METHODS: EPCs isolated from human peripheral blood was characterized using immunofluorescence and flow cytometry. Cell proliferation was determined with CCK8 and EdU assays. Cell migration was assessed by Transwell and wound healing assays. The angiogenic potential was evaluated using tube formation assay. The pyroptosis pathway-related protein in EPCs was detected by western blot. The binding sites of H19 and FADD on miR-107 were analyzed using Luciferase assays. In vivo, oe-H19-EPCs were transplanted into a mouse ischemic limb model, and blood flow was detected by laser Doppler imaging. The transcriptional landscape behind the therapeutic effects of oe-H19-EPCs on ischemic limbs were examined with whole transcriptome sequencing.
    RESULTS: Overexpression of H19 in EPCs led to an increase in proliferation, migration, and tube formation abilities. These effects were mediated through pyroptosis pathway, which is regulated by the H19/miR-107/FADD axis. Transplantation of oe-H19-EPCs in a mouse ischemic limb model promoted vasculogenesis and blood flow recovery. Whole transcriptome sequencing indicated significant activation of vasculogenesis pathway in the ischemic limbs following treatment with oe-H19-EPCs.
    CONCLUSIONS: Overexpression of H19 increases FADD level by competitively binding to miR-107, leading to enhanced proliferation, migration, vasculogenesis, and inhibition of pyroptosis in EPCs. These effects ultimately promote the recovery of blood flow in CLI.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    幽门螺杆菌(H.pylori),连同它的CagA,与造成DNA损伤有关,细胞周期停滞,凋亡,和胃癌的发展。尽管lncRNAH19在胃癌中大量表达并作为原癌基因发挥作用,目前尚不清楚lncRNAH19是否有助于幽门螺杆菌CagA的致癌过程。本研究探讨了H19在幽门螺杆菌诱导的DNA损伤反应和恶性肿瘤中的作用。观察到感染CagA+H.pylori菌株(GZ7/cagA)的细胞显示出显著较高的H19表达,导致γH2A增加。X和p-ATM表达下降,p53和Rad51表达下降。还观察到更快的细胞迁移和侵袭,H19基因在幽门螺杆菌中被逆转。YWHAZ被鉴定为H19靶蛋白,在H19敲低细胞中表达增加。GZ7/cagA感染对H19敲低诱导的YWHAZ表达增加有反应。此外,H19敲低刺激细胞进入G2期并减弱GZ7/cagA感染对细胞S期屏障的影响。结果提示幽门螺杆菌CagA可上调H19表达,参与DNA损伤反应,促进细胞迁移和侵袭,并可能通过调节YWHAZ影响细胞周期阻滞。
    Helicobacter pylori (H. pylori), together with its CagA, has been implicated in causing DNA damage, cell cycle arrest, apoptosis, and the development of gastric cancer. Although lncRNA H19 is abundantly expressed in gastric cancer and functions as a pro-oncogene, it remains unclear whether lncRNA H19 contributes to the oncogenic process of H. pylori CagA. This study investigates the role of H19 in the DNA damage response and malignancy induced by H. pylori. It was observed that cells infected with CagA+ H. pylori strain (GZ7/cagA) showed significantly higher H19 expression, resulting in increased γH2A.X and p-ATM expression and decreased p53 and Rad51 expression. Faster cell migration and invasion was also observed, which was reversed by H19 knockdown in H. pylori. YWHAZ was identified as an H19 target protein, and its expression was increased in H19 knockdown cells. GZ7/cagA infection responded to the increased YWHAZ expression induced by H19 knockdown. In addition, H19 knockdown stimulated cells to enter the G2-phase and attenuated the effect of GZ7/cagA infection on the cellular S-phase barrier. The results suggest that H. pylori CagA can upregulate H19 expression, participate in the DNA damage response and promote cell migration and invasion, and possibly affect cell cycle arrest via regulation of YWHAZ.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肺癌是全球死亡的主要原因,尤其是在有表皮生长因子受体(EGFR)突变的非小细胞肺癌(NSCLC)亚洲患者中.最初,第一代EGFR酪氨酸激酶抑制剂(TKIs)通常作为主要治疗选择;然而,遇到对这些药物的耐药性是一个重大障碍。因此,解决最初的阻力并确保持续有效已变得至关重要。最近的研究集中在长链非编码RNA(lncRNAs)在肿瘤耐药中的作用,尤其是lncRNAH19.β-榄香烯,来自姜黄芳香盐。,显示出强烈的抗肿瘤作用。然而,β-榄香烯之间的关系,NSCLC中的lncRNAH19和吉非替尼耐药尚不清楚。本研究旨在探讨β-榄香烯是否能增强吉非替尼耐药NSCLC细胞对吉非替尼的敏感性,并阐明其作用机制。使用细胞计数试剂盒-8(CCK8)测定评价吉非替尼和β-榄香烯对细胞活力的影响。此外,蛋白质印迹和qRT-PCR分析用于确定自噬相关蛋白和基因的表达水平,分别。对细胞增殖的影响通过集落形成试验来衡量,凋亡诱导通过流式细胞术定量。此外,使用裸鼠异种移植模型评估体内致瘤潜能.LC3B的表达水平,EGFR,通过免疫荧光检查Rab7蛋白。我们的发现阐明了对吉非替尼的抗性与自噬的失调和lncRNAH19的过表达密切相关。β-榄香烯和吉非替尼的协同给药显著减弱耐药细胞的增殖能力,加速凋亡过程,并抑制肺癌的体内增殖。值得注意的是,β-榄香烯极大地减少了lncRNAH19的表达,并降低了抗性细胞中的自噬活性,从而增强他们对吉非替尼的反应性。此外,β-榄香烯破坏了Rab7促进的EGFR降解途径,促进其重新定位到质膜。β-榄香烯是一种有前途的辅助治疗方法,用于规避非小细胞肺癌吉非替尼耐药,可能通过调节lncRNAH19介导的自噬。Rab7参与这一动态揭示了对肺癌耐药机制的新见解,为未来的治疗创新铺平道路。
    Lung cancer is a leading cause of mortality worldwide, especially among Asian patients with non-small cell lung cancer (NSCLC) who have epidermal growth factor receptor (EGFR) mutations. Initially, first-generation EGFR tyrosine kinase inhibitors (TKIs) are commonly administered as the primary treatment option; however, encountering resistance to these medications poses a significant obstacle. Hence, it has become crucial to address initial resistance and ensure continued effectiveness. Recent research has focused on the role of long noncoding RNAs (lncRNAs) in tumor drug resistance, especially lncRNA H19. β-elemene, derived from Curcuma aromatic Salisb., has shown strong anti-tumor effects. However, the relationship between β-elemene, lncRNA H19, and gefitinib resistance in NSCLC is unclear. This study aims to investigate whether β-elemene can enhance the sensitivity of gefitinib-resistant NSCLC cells to gefitinib and to elucidate its mechanism of action. The impact of gefitinib and β-elemene on cell viability was evaluated using the cell counting kit-8 (CCK8) assay. Furthermore, western blotting and qRT-PCR analysis were employed to determine the expression levels of autophagy-related proteins and genes, respectively. The influence on cellular proliferation was gauged through a colony-formation assay, and apoptosis induction was quantified via flow cytometry. Additionally, the tumorigenic potential in vivo was assessed using a xenograft model in nude mice. The expression levels of LC3B, EGFR, and Rab7 proteins were examined through immunofluorescence. Our findings elucidate that the resistance to gefitinib is intricately linked with the dysregulation of autophagy and the overexpression of lncRNA H19. The synergistic administration of β-elemene and gefitinib markedly attenuated the proliferative capacity of resistant cells, expedited apoptotic processes, and inhibited the in vivo proliferation of lung cancer. Notably, β-elemene profoundly diminished the expression of lncRNA H19 and curtailed autophagic activity in resistant cells, thereby bolstering their responsiveness to gefitinib. Moreover, β-elemene disrupted the Rab7-facilitated degradation pathway of EGFR, facilitating its repositioning to the plasma membrane. β-elemene emerges as a promising auxiliary therapeutic for circumventing gefitinib resistance in NSCLC, potentially through the regulation of lncRNA H19-mediated autophagy. The participation of Rab7 in this dynamic unveils novel insights into the resistance mechanisms operative in lung cancer, paving the way for future therapeutic innovations.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    双氯芬酸引起的小肠损伤称为双氯芬酸肠病。小檗碱(BBR),一类来源于小檗和黄柏的异喹啉类生物碱,广泛用于肠道疾病。本研究评估了BBR对双氯芬酸肠病肠黏膜机械屏障的保护作用及其可能的作用机制。体外动物实验表明,BBR下调了小肠和外泌体中长非编码RNAH19(lncRNAH19)的表达。在涉及外泌体和肠上皮细胞-6(IEC-6)细胞的共培养实验中,qRT-PCR的结果,西方印迹,免疫荧光实验表明,lncRNAH19在小肠中的表达升高,通过来自双氯芬酸基团的外泌体传递,抑制自噬相关蛋白5(Atg5)和轻链3(LC3)的表达水平,以及紧密连接(TJ)蛋白带闭塞-1(ZO-1),claudin-1和阻塞,相对于对照组。BBR治疗减弱外泌体lncRNAH19水平,上调Atg5和LC3的表达,增强TJ蛋白表达,并增加轻链3(LC3)-II/LC3-I的比例。这些发现显著阐明BBR通过抑制外泌体lncRNAH19促进IECs中自噬的恢复,从而减轻双氯芬酸肠病中肠粘膜机械屏障功能的损害。涉及外泌体lncRNAH19调节自噬的过程,从而影响肠粘膜机械屏障,为BBR在双氯芬酸肠病治疗中的应用提供了新的视角。
    Small intestine damage caused by diclofenac is called diclofenac enteropathy. Berberine (BBR), a class of isoquinoline alkaloids derived from Berberis vulgaris and Phellodendron amurense, is widely used in intestinal diseases. The present study evaluated the protective effect of BBR on the intestinal mucosal mechanical barrier in diclofenac enteropathy and its possible action mechanism. The in vitro animal experiment revealed that BBR downregulated the expression of long non-coding RNA H19 (lncRNA H19) in the small intestine and exosomes. In the co-culture experiment involving exosomes and intestinal epithelial cell-6 (IEC-6) cells, the results of qRT-PCR, western blotting, and immunofluorescence assays demonstrated that the elevated expression of lncRNA H19 in the small intestine, conveyed via exosomes derived from the diclofenac group, suppressed the expression levels of autophagy-associated protein 5 (Atg 5) and light chain 3 (LC 3), as well as and the tight junction (TJ) proteins zonula occludens-1 (ZO-1), claudin-1, and occluding, relative to the control group. BBR treatment attenuated exosomal lncRNA H19 levels, upregulated the expression of Atg5 and LC3 expression, enhanced TJ protein expression, and increased the light chain 3 (LC3)-II/LC3-I ratio. These findings significantly elucidated that BBR promoted the restoration of autophagy in IECs by inhibiting exosomal lncRNA H19, thereby mitigating the impairment of the intestinal mucosal mechanical barrier function in diclofenac enteropathy. The process involving exosomal lncRNA H19 regulating autophagy, thereby affecting the intestinal mucosal mechanical barrier, offers a novel perspective for the application of BBR in the treatment of diclofenac enteropathy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    单一固定理论不能完全解释脊髓损伤(SCI)后观察到的广泛骨丢失。骨髓间充质干细胞(BMSCs)具有自我更新能力和各种类型的分化潜能,在骨稳态中起着至关重要的作用。本研究旨在探讨长链非编码RNAH19在SCI后骨质疏松中的分子机制,为现有防治策略提供新的研究方向。我们使用小干扰RNA敲低H19表达并使用miR-29b-3p模拟物和抑制剂调节miR-29b-2p表达。西方印迹,实时荧光定量PCR,茜素红染色,碱性磷酸酶染色和双荧光素酶报告基因测定用于评估基因表达,成骨能力和结合位点。lncRNAH19在骨质疏松组的BMSCs中上调,而miR-29b-3p下调。我们鉴定了miR-29b-3p与lncRNAsH19和DKK1之间的结合位点。H19敲低促进BMSCs成骨分化,而miR-29b-3p抑制减弱了这种作用。我们在lncRNAsH19和DKK1中发现了miR-29b-3p的潜在结合位点。我们的发现表明,长链非编码RNAH19通过miR-29b-3p/DKK1轴和直接抑制β-catenin信号通路介导SCI后骨质疏松中的BMSCs成骨分化。
    Single immobilization theory cannot fully account for the extensive bone loss observed after spinal cord injury (SCI). Bone marrow mesenchymal stem cells (BMSCs) are crucial in bone homeostasis because they possess self-renewal capabilities and various types of differentiation potential. This study aimed to explore the molecular mechanism of long non-coding RNA H19 in osteoporosis after SCI and provide new research directions for existing prevention strategies. We used small interfering RNA to knockdown H19 expression and regulated miR-29b-2p expression using miR-29b-3p mimetics and inhibitors. Western blotting, real-time fluorescence quantitative PCR, Alizarin red staining, alkaline phosphatase staining and double-luciferase reporter gene assays were used to assess gene expression, osteogenic ability and binding sites. lncRNA H19 was upregulated in BMSCs from the osteoporosis group, whereas miR-29b-3p was downregulated. We identified the binding sites between miR-29b-3p and lncRNAs H19 and DKK1. H19 knockdown promoted BMSCs\' osteogenic differentiation, whereas miR-29b-3p inhibition attenuated this effect. We discovered potential binding sites for miR-29b-3p in lncRNAs H19 and DKK1. Our findings suggest that long non-coding RNA H19 mediates BMSCs\' osteogenic differentiation in osteoporosis after SCI through the miR-29b-3p/DKK1 axis and by directly inhibiting the β-catenin signalling pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    作为最早发现的lncRNA分子之一,lncRNAH19通常在胚胎发育过程中大量表达,并参与细胞分化和组织形成。近年来,lncRNAH19在肿瘤中的作用已被逐渐认识到。越来越多的证据表明,其异常表达与癌症的发展密切相关。LncRNAH19作为癌基因不仅促进生长,扩散,许多肿瘤的侵袭和转移,还会对治疗产生抗药性,影响患者预后和生存。因此,在这次审查中,我们总结了lncRNAH19参与肿瘤进展的广泛研究,并讨论了lncRNAH19作为关键靶基因,影响肿瘤对放疗的敏感性,化疗和免疫治疗通过参与多个细胞过程和调节多个信号通路,这为进一步研究癌症治疗提供了广阔的前景。
    As one of the earliest discovered lncRNA molecules, lncRNA H19 is usually expressed in large quantities during embryonic development and is involved in cell differentiation and tissue formation. In recent years, the role of lncRNA H19 in tumors has been gradually recognized. Increasing evidence suggests that its aberrant expression is closely related to cancer development. LncRNA H19 as an oncogene not only promotes the growth, proliferation, invasion and metastasis of many tumors, but also develops resistance to treatment, affecting patients\' prognosis and survival. Therefore, in this review, we summarise the extensive research on the involvement of lncRNA H19 in tumor progression and discuss how lncRNA H19, as a key target gene, affects tumor sensitivity to radiotherapy, chemotherapy and immunotherapy by participating in multiple cellular processes and regulating multiple signaling pathways, which provides a promising prospect for further research into the treatment of cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    心血管疾病是全世界死亡和残疾的主要原因。最近,越来越多的证据表明,长链非编码RNA(lncRNAs)在心血管疾病的发病机制中起关键作用,包括动脉粥样硬化,冠状动脉疾病,扩张型心肌病,糖尿病性心肌病,主动脉夹层,还有更多.LncRNAH19是第一个被描述为非编码蛋白质的mRNA样分子。大量研究发现lncRNAH19与心血管疾病的病理生理过程有关,它正在成为各种心脏病的潜在关键调节剂。在这次审查中,我们旨在总结lncRNAH19在心血管疾病中的作用,为其未来作为新的治疗靶点的潜在用途提供理论依据。
    Cardiovascular disease is a major cause of death and disability worldwide. Recently, increasing evidence has demonstrated that long non-coding RNAs (lncRNAs) play critical roles in the pathogenesis of cardiovascular diseases, including atherosclerosis, coronary artery disease, dilated cardiomyopathy, diabetic cardiomyopathy, aortic dissection, and more. LncRNA H19 was the first to be described as a non-protein-coding mRNA-like molecule. A large number of studies have found that lncRNA H19 is related to the pathophysiological processes of cardiovascular diseases, and it is emerging as a potential key regulator of various heart diseases. In this review, we aim to summarize the role of lncRNA H19 in cardiovascular diseases in order to provide a theoretical basis for its potential use as a new therapeutic target in the future.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号