Beta catenin

β 连环蛋白
  • 文章类型: Journal Article
    背景:牛黄(CB),用于中药,在各种癌症模型中表现出抗肿瘤作用。它还构成了称为PienTzeHuang的复合制剂的组成部分,用于治疗肝癌。然而,它对肝癌肿瘤微环境的影响,特别是在肿瘤相关巨噬细胞(TAMs)上,不是很了解。
    目的:阐明CB通过Wnt/β-catenin通路调控抑制M2-TAM极化的抗肝癌作用。
    方法:本研究使用UPLC-Q-TOF-MS鉴定了CB的活性成分,在裸鼠模型中评估了其抗肿瘤作用,并通过网络药理学阐明了潜在的机制,转录组学,和分子对接。体外试验用于研究含CB的血清对HepG2细胞和M2-TAMs的影响。通过实时逆转录酶-聚合酶链反应和Westernblot分析验证了Wnt通路的调节。
    结果:这项研究确定了CB中的22种活性成分,其中11在血液中检测到。临床前研究已证明CB有效抑制肝肿瘤生长的能力。采用网络药理学的综合方法,转录组学,和分子对接通过抑制M2-TAM极化将Wnt信号通路作为CB抗肿瘤活性的靶标。体外和体内实验进一步证实,CB显著阻碍M2-TAM极化并抑制Wnt/β-连环蛋白途径活化。当用Wnt激动剂SKL2001处理时,CB对M2-TAM的抑制作用被逆转,证实了其途径特异性。
    结论:这项研究表明,CB通过Wnt/β-catenin通路介导M2-TAM极化的抑制,有助于抑制肝癌的生长。
    BACKGROUND: Calculus bovis (CB), used in traditional Chinese medicine, exhibits anti-tumor effects in various cancer models. It also constitutes an integral component of a compound formulation known as Pien Tze Huang, which is indicated for the treatment of liver cancer. However, its impact on the liver cancer tumor microenvironment, particularly on tumor-associated macrophages (TAMs), is not well understood.
    OBJECTIVE: To elucidate the anti-liver cancer effect of CB by inhibiting M2-TAM polarization via Wnt/β-catenin pathway modulation.
    METHODS: This study identified the active components of CB using UPLC-Q-TOF-MS, evaluated its anti-neoplastic effects in a nude mouse model, and elucidated the underlying mechanisms via network pharmacology, transcriptomics, and molecular docking. In vitro assays were used to investigate the effects of CB-containing serum on HepG2 cells and M2-TAMs, and Wnt pathway modulation was validated by real-time reverse transcriptase-polymerase chain reaction and Western blot analysis.
    RESULTS: This study identified 22 active components in CB, 11 of which were detected in the bloodstream. Preclinical investigations have demonstrated the ability of CB to effectively inhibit liver tumor growth. An integrated approach employing network pharmacology, transcriptomics, and molecular docking implicated the Wnt signaling pathway as a target of the antineoplastic activity of CB by suppressing M2-TAM polarization. In vitro and in vivo experiments further confirmed that CB significantly hinders M2-TAM polarization and suppresses Wnt/β-catenin pathway activation. The inhibitory effect of CB on M2-TAMs was reversed when treated with the Wnt agonist SKL2001, confirming its pathway specificity.
    CONCLUSIONS: This study demonstrated that CB mediates inhibition of M2-TAM polarization through the Wnt/β-catenin pathway, contributing to the suppression of liver cancer growth.
    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
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    Objective: To explore the gene mutation characteristics and the relationship between gene mutations and long-term prognosis in clinical stage ⅠA lung adenocarcinoma patients. Methods: A retrospective analysis was conducted on 63 clinical stage ⅠA lung adenocarcinoma patients who underwent surgical resection at the Cancer Hospital of the Chinese Academy of Medical Sciences from January 2007 to October 2012, with documented postoperative recurrence or metastasis, as well as those who had a follow-up duration of 10 years or more without recurrence or metastasis. Whole exome sequencing (WES) technology was used to analyze the gene mutation profiles in tumor tissues and univariate and multivariate Cox regression analysis were used to clarify the influencing factors for patient prognosis. Results: After long term follow-up, 13 out of the 63 patients (21%) experienced recurrence or metastasis. WES technology analysis revealed that the most common tumor related gene mutations occurred in epidermal growth factor receptor (EGFR), with a mutation rate of 65.1% (41/63), followed by tumor protein p53 (TP53), fatatypical cadherin 1 (FAT1), low density lipoprotein receptor-related protein 1B (LRP1B), mechanistic target of rapamycin (MTOR), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit gamma (PIK3CG), and SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily A, member 4 (SMARCA4), with mutation rates of 30.2% (19/63), 20.6% (13/63), 15.9% (10/63), 15.9% (10/63), 15.9% (10/63), and 15.9% (10/63), respectively. Multivariate Cox regression analysis showed that PIK3CG mutations (HR=21.52, 95% CI: 3.19-145.01),smoothened (SMO) mutations (HR=35.28, 95% CI: 3.12-398.39), catenin beta 1 (CTNNB1) mutations (HR=332.86, 95% CI: 15.76-7 029.05), colony stimulating factor 1 receptor (CSF1R) mutations (HR=8 109.60, 95% CI: 114.19-575 955.17), and v-Raf murine sarcoma viral oncogene homolog B (BRAF) mutations (HR=23.65, 95% CI: 1.86-300.43) were independent risk factors affecting the prognosis of clinical stage ⅠA lung adenocarcinoma patients. Conclusions: PIK3CG, SMO, CTNNB1, CSF1R, BRAF gene mutations are closely related to long-term recurrence or metastasis in clinical stage ⅠA lung adenocarcinoma. Patients with these gene mutations should be given closer clinical attention.
    目的: 探讨临床ⅠA期肺腺癌的基因突变特征及其与患者预后的关系,为早期肺腺癌患者的个体化治疗提供依据。 方法: 收集2007年1月至2012年10月在中国医学科学院肿瘤医院接受手术切除且随访达10年以上或随访期间出现复发或转移的临床ⅠA期肺腺癌患者63例,采用全外显子组测序(WES)技术分析肺癌组织的基因突变谱,采用单因素和多因素Cox回归分析明确患者预后影响因素。 结果: 在随访期间,63例患者中13例(20.6%)出现复发或转移。WES技术分析显示,肺癌组织中表皮生长因子受体突变频率最高,达65.1%(41/63),其次为肿瘤蛋白p53、异常类脂肪酸1、低密度脂蛋白受体相关蛋白1B、雷帕霉素机械靶点、磷脂酰肌醇4,5-双磷酸3-激酶催化亚单位γ(PIK3CG)及与SWI/SNF相关基质相关的依赖于肌动蛋白的染色质调节因子亚家族A成员4,突变频率分别为30.2%(19/63)、20.6%(13/63)、15.9%(10/63)、15.9%(10/63)、15.9%(10/63)和15.9%(10/63)。多因素Cox回归分析显示,PIK3CG突变(HR=21.52,95%CI:3.19~145.01)、平滑蛋白(SMO)突变(HR=35.28,95%CI:3.12~398.39)、β-连环蛋白1(CTNNB1)突变(HR=332.86,95%CI:15.76~7 029.05)、集落刺激因子1受体(CSF1R)突变(HR=8 109.60,95%CI:114.19~575 955.17)、v-Raf小鼠肉瘤病毒癌基因同源B(BRAF)突变(HR=23.65,95%CI:1.86~300.43)为临床ⅠA期肺腺癌患者预后的独立危险因素。 结论: PIK3CG、SMO、CTNNB1、CSF1R、BRAF基因突变与临床ⅠA期肺腺癌的远期复发和转移密切相关,应给予具有这些基因突变的肺腺癌患者更为密切的临床关注。.
    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
    肝星状细胞(HSC)活化是肝纤维化(LF)的重要病理进程。调节HSC活化和LF的分子机制尚不完全清楚。这里,我们探讨了转录因子SRY相关的高迁移率族蛋白7(SOX7)对HSC激活和LF的影响,以及潜在的分子机制。我们发现SOX7在人和小鼠纤维化肝脏中的表达水平降低,特别是在纤维化病灶。SOX7在原代活化的HSC和TGF-β1刺激的LX-2细胞中也下调。SOX7敲低可促进LX-2细胞的活化和增殖,同时抑制其凋亡。另一方面,SOX7的过表达抑制了HSC的活化和增殖。机械上,SOX7通过降低TGF-β1诱导的β-连环蛋白的表达和Smad2和Smad3的磷酸化来减弱HSC活化和LF。此外,使用AAV8-SOX7小鼠模型的SOX7的过表达改善了在体内响应于CCl4处理的LF的程度。总的来说,SOX7抑制HSC激活和LF。因此,以SOX7为目标,可能是一种潜在的新策略,以防止LF。
    Hepatic stellate cell (HSC) activation is the essential pathological process of liver fibrosis (LF). The molecular mechanisms regulating HSC activation and LF are incompletely understood. Here, we explored the effect of transcription factor SRY-related high mobility group box 7 (SOX7) on HSC activation and LF, and the underlying molecular mechanism. We found the expression levels of SOX7 were decreased in human and mouse fibrotic livers, particularly at the fibrotic foci. SOX7 was also downregulated in primary activated HSCs and TGF-β1 stimulated LX-2 cells. SOX7 knockdown promoted activation and proliferation of LX-2 cells while inhibiting their apoptosis. On the other hand, overexpression of SOX7 suppressed the activation and proliferation of HSCs. Mechanistically, SOX7 attenuates HSC activation and LF by decreasing the expression of β-catenin and phosphorylation of Smad2 and Smad3 induced by TGF-β1. Furthermore, overexpression of SOX7 using AAV8-SOX7 mouse models ameliorated the extent of LF in response to CCl4 treatment in vivo. Collectively, SOX7 suppressed HSC activation and LF. Targeting SOX7, therefore, could be a potential novel strategy to protect against LF.
    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
    角膜上皮位于眼球的最前表面,可防止外部刺激。角膜上皮的发育和角膜稳态的维持对于维持视力至关重要。最近通过对眼表疾病的深入研究发现,Wnt/β-catenin信号通路对于角膜上皮细胞的生长和分层以及内皮细胞稳定性的控制是必需的。此外,Wnt/β-catenin信号通路与圆锥角膜等常见角膜疾病的发展直接相关,真菌性角膜炎,和角膜新生血管形成。本文主要综述了Wnt/β-catenin信号通路在发育过程中的作用,稳态,角膜的病理学,希望为角膜上皮的研究和相关疾病的治疗提供新的见解。
    The corneal epithelium is located on the most anterior surface of the eyeball and protects against external stimuli. The development of the corneal epithelium and the maintenance of corneal homeostasis are essential for the maintenance of visual acuity. It has been discovered recently via the in-depth investigation of ocular surface illnesses that the Wnt/β-catenin signaling pathway is necessary for the growth and stratification of corneal epithelial cells as well as the control of endothelial cell stability. In addition, the Wnt/β-catenin signaling pathway is directly linked to the development of common corneal illnesses such as keratoconus, fungal keratitis, and corneal neovascularization. This review mainly summarizes the role of the Wnt/β-catenin signaling pathway in the development, homeostasis, and pathobiology of cornea, hoping to provide new insights into the study of corneal epithelium and the treatment of related diseases.
    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
    泛素化,一种普遍且高度动态的可逆翻译后修饰,受到去泛素化酶(DUBs)超家族的严格调控。其中,含OTU结构域的泛素醛结合蛋白1(OTUB1)是OTU去泛素家族的关键成员,在各种癌症中作为肿瘤调节剂发挥关键作用。然而,其在BLCA(BLCA)中的具体参与及其临床意义仍然不明确。本研究旨在阐明OTUB1在BLCA中的生物学功能及其对临床预后的影响。我们的调查显示,BLCA中OTUB1的表达增加,与不利的临床结果相关。通过体内和体外实验,我们证明,增加OTUB1水平促进BLCA肿瘤发生和进展,同时赋予顺铂治疗耐药性。值得注意的是,我们建立了一个涉及OTUB1、β-catenin、坏死,BLCA,描述它们之间的监管相互作用。机械上,我们发现OTUB1通过去泛素化和稳定β-catenin发挥其影响,导致其核易位。随后,核β-catenin增强c-myc和cyclinD1的转录活性,同时抑制RIPK3和MLKL的表达,从而促进BLCA进展和顺铂耐药。重要的是,我们的临床数据提示OTUB1/β-catenin/RIPK3/MLKL轴有望成为BLCA的潜在生物标志物.
    Ubiquitination, a prevalent and highly dynamic reversible post-translational modification, is tightly regulated by the deubiquitinating enzymes (DUBs) superfamily. Among them, OTU Domain-Containing Ubiquitin Aldehyde-Binding Protein 1 (OTUB1) stands out as a critical member of the OTU deubiquitinating family, playing a pivotal role as a tumor regulator across various cancers. However, its specific involvement in BLCA (BLCA) and its clinical significance have remained ambiguous. This study aimed to elucidate the biofunctions of OTUB1 in BLCA and its implications for clinical prognosis. Our investigation revealed heightened OTUB1 expression in BLCA, correlating with unfavorable clinical outcomes. Through in vivo and in vitro experiments, we demonstrated that increased OTUB1 levels promote BLCA tumorigenesis and progression, along with conferring resistance to cisplatin treatment. Notably, we established a comprehensive network involving OTUB1, β-catenin, necroptosis, and BLCA, delineating their regulatory interplay. Mechanistically, we uncovered that OTUB1 exerts its influence by deubiquitinating and stabilizing β-catenin, leading to its nuclear translocation. Subsequently, nuclear β-catenin enhances the transcriptional activity of c-myc and cyclin D1 while suppressing the expression of RIPK3 and MLKL, thereby fostering BLCA progression and cisplatin resistance. Importantly, our clinical data suggest that the OTUB1/β-catenin/RIPK3/MLKL axis holds promise as a potential biomarker for BLCA.
    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
    背景:牙周膜干细胞(PDLSCs)在修复牙周破坏中的作用至关重要,但过度的氧化应激(OS)会损害它们的功能。诺亚达明(NOCA),环状铁载体,已被证明具有抗癌和抗菌特性。本研究旨在探讨NOCA对OS诱导的PDLSCs细胞功能障碍的保护机制。
    方法:使用CCK-8测定法评估NOCA对PDLSC的细胞毒性。然后用过氧化氢(H2O2)处理PDLSC以诱导OS。ROS水平,细胞活力,治疗后使用相关试剂盒分析抗氧化因子的表达。使用小分子抑制剂U0126和XAV-939分别阻断ERK信号传导和Wnt途径。使用矿化结节的碱性磷酸酶(ALP)活性染色和茜素红S(ARS)染色评估成骨分化。通过实时定量聚合酶链反应(RT-qPCR)或蛋白质印迹(WB)分析确定成骨基因标记和ERK途径的表达水平。通过蛋白质印迹和共聚焦显微镜检查β-catenin核定位。
    结果:NOCA在浓度低于20µM时无明显的细胞毒性,并有效抑制H2O2诱导的PDLSCs中的OS。NOCA还恢复了ALP活性,矿化结节形成,和成骨标志物在H2O2刺激的PDLSCs中的表达。机械上,NOCA增加p-ERK水平并促进β-catenin易位进入细胞核;然而,阻断ERK途径破坏了NOCA提供的成骨保护,并损害了其在OS条件下在PDLSCs中诱导β-catenin核易位的能力。
    结论:NOCA通过调节ERK/Wnt信号通路保护PDLSCs抵抗H2O2诱导的OS,并有效恢复PDLSCs成骨分化受损。
    BACKGROUND: The role of periodontal ligament stem cells (PDLSCs) in repairing periodontal destruction is crucial, but their functions can be impaired by excessive oxidative stress (OS). Nocardamine (NOCA), a cyclic siderophore, has been shown to possess anti-cancer and anti-bacterial properties. This study aimed to investigate the protective mechanisms of NOCA against OS-induced cellular dysfunction in PDLSCs.
    METHODS: The cytotoxicity of NOCA on PDLSCs was assessed using a CCK-8 assay. PDLSCs were then treated with hydrogen peroxide (H2O2) to induce OS. ROS levels, cell viability, and antioxidant factor expression were analyzed using relevant kits after treatment. Small molecule inhibitors U0126 and XAV-939 were employed to block ERK signaling and Wnt pathways respectively. Osteogenic differentiation was assessed using alkaline phosphatase (ALP) activity staining and Alizarin Red S (ARS) staining of mineralized nodules. Expression levels of osteogenic gene markers and ERK pathway were determined via real-time quantitative polymerase chain reaction (RT-qPCR) or western blot (WB) analysis. β-catenin nuclear localization was examined by western blotting and confocal microscopy.
    RESULTS: NOCA exhibited no significant cytotoxicity at concentrations below 20 µM and effectively inhibited H2O2-induced OS in PDLSCs. NOCA also restored ALP activity, mineralized nodule formation, and the expression of osteogenic markers in H2O2-stimulated PDLSCs. Mechanistically, NOCA increased p-ERK level and promoted β-catenin translocation into the nucleus; however, blocking ERK pathway disrupted the osteogenic protection provided by NOCA and impaired its ability to induce β-catenin nuclear translocation under OS conditions in PDLSCs.
    CONCLUSIONS: NOCA protected PDLSCs against H2O2-induced OS and effectively restored impaired osteogenic differentiation in PDLSCs by modulating the ERK/Wnt signaling 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
    背景:大约25-30%的急性髓性白血病(AML)患者存在FMS样受体酪氨酸激酶-3(FLT3)突变,这些突变导致疾病进展和不良预后。长期暴露于FLT3酪氨酸激酶抑制剂(TKIs)通常由于不同的代偿生存信号而导致有限的临床反应。因此,迫切需要阐明FLT3TKI耐药的潜在机制.异常调节的鞘脂代谢经常导致癌症进展和不良的治疗反应。然而,在FLT3突变的AML中,其与TKI敏感性的关系尚不清楚.因此,我们旨在评估AML中FLT3TKI耐药的机制.
    方法:我们进行了脂质组学分析,RNA-seq,qRT-PCR,和酶联免疫吸附试验,以确定索拉非尼耐药的潜在驱动因素。FLT3信号被索拉非尼或奎扎替尼抑制,通过使用拮抗剂或通过敲除抑制SPHK1。通过细胞计数试剂盒-8,PI染色,在FLT3突变和野生型AML细胞系中评估细胞生长和凋亡,和膜联蛋白-V/7AAD测定。采用蛋白质印迹和免疫荧光分析,通过使用SPHK1过表达和外源S1P的拯救实验来探索潜在的分子机制。以及S1P2,β-连环蛋白的抑制剂,PP2A,和GSK3β。异种移植鼠模型,患者样本,和公开可用的数据进行了分析,以证实我们的体外结果。
    结果:我们证明长期索拉非尼治疗可上调SPHK1/1-磷酸鞘氨醇(S1P)信号传导,它又通过S1P2受体积极调节β-连环蛋白信号传导以抵消TKI介导的对FLT3突变的AML细胞的抑制。SPHK1的遗传或药理学抑制在体外有效增强了TKI介导的FLT3突变的AML细胞的增殖抑制和凋亡诱导。SPHK1敲低增强索拉非尼功效并改善AML异种移植小鼠的存活率。机械上,靶向SPHK1/S1P/S1P2信号与FLT3TKIs协同作用,通过激活蛋白磷酸酶2A(PP2A)-糖原合酶激酶3β(GSK3β)途径来抑制β-catenin活性。
    结论:这些发现确立了鞘脂代谢酶SPHK1作为TKI敏感性的调节因子,并表明将SPHK1抑制与TKIs结合可能是治疗FLT3突变的AML的有效方法。
    BACKGROUND: Approximately 25-30% of patients with acute myeloid leukemia (AML) have FMS-like receptor tyrosine kinase-3 (FLT3) mutations that contribute to disease progression and poor prognosis. Prolonged exposure to FLT3 tyrosine kinase inhibitors (TKIs) often results in limited clinical responses due to diverse compensatory survival signals. Therefore, there is an urgent need to elucidate the mechanisms underlying FLT3 TKI resistance. Dysregulated sphingolipid metabolism frequently contributes to cancer progression and a poor therapeutic response. However, its relationship with TKI sensitivity in FLT3-mutated AML remains unknown. Thus, we aimed to assess mechanisms of FLT3 TKI resistance in AML.
    METHODS: We performed lipidomics profiling, RNA-seq, qRT-PCR, and enzyme-linked immunosorbent assays to determine potential drivers of sorafenib resistance. FLT3 signaling was inhibited by sorafenib or quizartinib, and SPHK1 was inhibited by using an antagonist or via knockdown. Cell growth and apoptosis were assessed in FLT3-mutated and wild-type AML cell lines via Cell counting kit-8, PI staining, and Annexin-V/7AAD assays. Western blotting and immunofluorescence assays were employed to explore the underlying molecular mechanisms through rescue experiments using SPHK1 overexpression and exogenous S1P, as well as inhibitors of S1P2, β-catenin, PP2A, and GSK3β. Xenograft murine model, patient samples, and publicly available data were analyzed to corroborate our in vitro results.
    RESULTS: We demonstrate that long-term sorafenib treatment upregulates SPHK1/sphingosine-1-phosphate (S1P) signaling, which in turn positively modulates β-catenin signaling to counteract TKI-mediated suppression of FLT3-mutated AML cells via the S1P2 receptor. Genetic or pharmacological inhibition of SPHK1 potently enhanced the TKI-mediated inhibition of proliferation and apoptosis induction in FLT3-mutated AML cells in vitro. SPHK1 knockdown enhanced sorafenib efficacy and improved survival of AML-xenografted mice. Mechanistically, targeting the SPHK1/S1P/S1P2 signaling synergizes with FLT3 TKIs to inhibit β-catenin activity by activating the protein phosphatase 2 A (PP2A)-glycogen synthase kinase 3β (GSK3β) pathway.
    CONCLUSIONS: These findings establish the sphingolipid metabolic enzyme SPHK1 as a regulator of TKI sensitivity and suggest that combining SPHK1 inhibition with TKIs could be an effective approach for treating FLT3-mutated AML.
    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
    背景:心房颤动(AF)与卒中风险和死亡率增加相关。据报道,房颤大鼠心房纤维化的过程受β-catenin调节。然而,人类房颤患者这一过程的病理生理机制尚不清楚。本研究旨在探讨β-catenin参与人右心耳(hRAA)组织心房纤维化的可能机制。
    方法:我们比较了房颤和窦性心律(SR)患者hRAA组织中β-catenin表达的差异。还在小鼠和原代细胞中探索了β-catenin在AF发展中的可能功能。
    结果:首先,AF组心肌细胞缝隙连接膜之间的间隙较宽。其次,缝隙连接功能相关蛋白的表达,Connexin40和Connexin43减少,而β-catenin及其结合伴侣E-cadherin在hRAA和AF组心肌细胞中的表达增加。第三,β-catenin与E-cadherin共定位在SR组心肌细胞的质膜上,而在房颤组,它们在细胞内分离和积累。此外,糖原合成酶激酶3β(GSK-3β)和腺瘤性结肠息肉病(APC)的表达,参与β-连环蛋白的降解,在AF组的hRAA组织和心肌细胞中降低。最后,在AF模型小鼠中,抑制β-catenin的表达后,心房纤维化和AF的发展被证明可以被阻止。
    结论:基于人类心房病理和分子分析,我们的研究结果提供了β-catenin与心房纤维化和房颤进展相关的证据.
    BACKGROUND: Atrial fibrillation (AF) is associated with increased risk of stroke and mortality. It has been reported that the process of atrial fibrosis was regulated by β-catenin in rats with AF. However, pathophysiological mechanisms of this process in human with AF remain unclear. This study aims to investigate the possible mechanisms of β-catenin in participating in the atrial fibrosis using human right atrial appendage (hRAA) tissues .
    METHODS: We compared the difference of β-catenin expression in hRAA tissues between the patients with AF and sinus rhythm (SR). The possible function of β-catenin in the development of AF was also explored in mice and primary cells.
    RESULTS: Firstly, the space between the membrane of the gap junctions of cardiomyocytes was wider in the AF group. Secondly, the expression of the gap junction function related proteins, Connexin40 and Connexin43, was decreased, while the expression of β-catenin and its binding partner E-cadherin was increased in hRAA and cardiomyocytes of the AF group. Thirdly, β-catenin colocalized with E-cadherin on the plasma membrane of cardiomyocytes in the SR group, while they were dissociated and accumulated intracellularly in the AF group. Furthermore, the expression of glycogen synthase kinase 3β (GSK-3β) and Adenomatous Polyposis Coli (APC), which participated in the degradation of β-catenin, was decreased in hRAA tissues and cardiomyocytes of the AF group. Finally, the development of atrial fibrosis and AF were proved to be prevented after inhibiting β-catenin expression in the AF model mice.
    CONCLUSIONS: Based on human atrial pathological and molecular analyses, our findings provided evidence that β-catenin was associated with atrial fibrosis and AF progression.
    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
    胃肠道癌症(GIC)是威胁全球人类健康的高度流行的癌症。据报道,Wnt/β-catenin信号通路在GIC的致癌作用中起关键作用。目前,针对GIC中Wnt/β-catenin信号传导的许多干预措施正在临床试验中进行测试,并获得了有希望的结果。不幸的是,目前尚无临床批准的有效靶向该途径的药物.本综述旨在评估靶向Wnt/β-catenin信号通路的临床治疗对GIC的影响。通过整合来自生物信息学数据库的数据和过去五年的最新文献,我们研究了GIC中Wnt/β-catenin通路基因和蛋白的异质性表达和调控机制。具体来说,我们专注于表达模式,突变频率,和临床预后,以了解它们对治疗策略的影响。此外,我们讨论了最近针对该途径的临床试验。了解目前正在进行临床研究的抑制剂可能有助于优化基础研究和临床策略。我们希望阐明针对Wnt/β-catenin通路的患者的精准治疗分层的现状将指导GIC的精准医学的未来创新。
    Gastrointestinal cancers (GICs) are highly prevalent cancers that threaten human health worldwide. The Wnt/β-catenin signaling pathway has been reported to play a pivotal role in the carcinogenesis of GICs. Numerous interventions targeting the Wnt/β-catenin signaling in GICs are currently being tested in clinical trials with promising results. Unfortunately, there are no clinically approved drugs that effectively target this pathway. This comprehensive review aims to evaluate the impact of clinical therapies targeting the Wnt/β-catenin signaling pathway in GICs. By integrating data from bioinformatics databases and recent literature from the past five years, we examine the heterogeneous expression and regulatory mechanisms of Wnt/β-catenin pathway genes and proteins in GICs. Specifically, we focus on expression patterns, mutation frequencies, and clinical prognoses to understand their implications for treatment strategies. Additionally, we discuss recent clinical trial efforts targeting this pathway. Understanding the inhibitors currently under clinical investigation may help optimize foundational research and clinical strategies. We hope that elucidating the current status of precision therapeutic stratification for patients targeting the Wnt/β-catenin pathway will guide future innovations in precision medicine for GICs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号