Invasion and metastasis

侵袭和转移
  • 文章类型: Journal Article
    本研究旨在通过网络药理学预测和实验验证,探讨山奈酚抑制胃癌(GC)细胞侵袭转移的机制。它通过PPI网络分析识别核心靶标,并发现山奈酚与这些靶标结合良好。体外实验表明山奈酚能抑制其增殖,菌落形成,GC细胞的迁移和侵袭。蛋白质印迹显示山奈酚可降低AKT和GSK3β磷酸化,导致侵袭相关基因SRC表达降低,MMP9、CXCR4、KDR、MMP2。总的来说,山奈酚可能通过AKT/GSK3β信号通路阻止GC细胞的迁移和侵袭。
    This study aims to explore the mechanisms of the inhibitory effect of kaempferol on the invasion and metastasis of gastric cancer (GC) cells through network pharmacology prediction and experimental verification. It identifies core targets via PPI network analysis and finds that kaempferol binds to these targets well. In vitro experiments showed that kaempferol could inhibit the proliferation, colony formation, migration and invasion of GC cells. Western blotting indicated kaempferol may reduce AKT and GSK3β phosphorylation, leading to lower expression of invasion-related genes SRC, MMP9, CXCR4, KDR, and MMP2. Overall, kaempferol may prevent migration and invasion of GC cells via the AKT/GSK3β signaling pathway.
    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
    含锌指MIZ型1(ZMIZ1)是与激活的STATs(PIAS)家族的蛋白质抑制剂相关的转录共激活因子。越来越多的证据表明,ZMIZ1在癌症的发生和发展中起着至关重要的作用。ZMIZ1在舌鳞状细胞癌(TSCC)中的功能及其在该疾病中的作用机制尚未完全阐明。我们使用免疫组织化学对20名患者来源的ZMIZ1蛋白表达进行了定性分析,石蜡包埋的TSCC组织切片。我们使用RNAi在CAL-27TSCC细胞系中敲低ZMIZ1表达,并定量ZMIZ1敲低对增殖的影响,通过CCK-8,划痕分析和流式细胞术,分别。我们使用qRT-PCR和蛋白质印迹来研究ZMIZ1在该细胞系中的作用。最后,我们建立了裸鼠肺转移模型,以复制体外结果。ZMIZ1蛋白在TSCC病例组织样品中明显更丰富。ZMIZ1敲低可降低TSCC肿瘤细胞的侵袭和转移,促进细胞凋亡。ZMIZ1敲低与Notch信号通路相关因子Jagged1和Notch1、侵袭转移相关因子MKP-1、SSBP2和MMP7的表达下调有关,在mRNA水平。体外和体内数据表明,敲低ZMIZ1可能通过调节Notch信号传导来抑制TSCC的侵袭和转移。因此,ZMIZ1抑制可以代表TSCC的新治疗靶标。
    Zinc finger MIZ-type containing 1 (ZMIZ1) is a transcriptional coactivator related to the protein inhibitors of activated STATs (PIAS) family. Mounting evidence suggests that ZMIZ1 plays a crucial role in the occurrence and development of cancers. The function of ZMIZ1 in tongue squamous cell carcinoma (TSCC) and the mechanisms underpinning its role in this disease have not been fully clarified. We performed qualitative ZMIZ1 protein expression analyses using immunohistochemistry in 20 patient-derived, paraffin-embedded TSCC tissue sections. We used RNAi to knock down ZMIZ1 expression in the CAL-27 TSCC cell line and quantified the impact of ZMIZ1 knock down on proliferation, migration and apoptosis via CCK-8, scratch assay and flow cytometry, respectively. We used qRT-PCR and western blotting to investigate the role of ZMIZ1 in this cell line. Finally, we established a model of lung metastasis in nude mice to replicate the in vitro results. ZMIZ1 protein was significantly more abundant in TSCC case tissue samples. ZMIZ1 knockdown reduced the invasion and metastases of TSCC tumor cells and promoted apoptosis. ZMIZ1 knockdown was associated with the down-regulation of Notch signaling pathway related factors Jagged1 and Notch1, and invasion and metastasis related factors MKP-1, SSBP2 and MMP7 in vitro and in vivo, at the mRNA level. In vitro and in vivo data suggest that knock down of ZMIZ1 may inhibit TSCC invasion and metastasis by modulating Notch signaling. ZMIZ1 inhibition may therefore represent a new therapeutic target for TSCC.
    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:肺癌仍然是癌症相关死亡的首要原因,侵袭和转移对患者预后影响深远。N-乙酰转移酶10(NAT10)催化真核RNA中的专有N(4)-乙酰胞苷(ac4C)修饰。NAT10失调与各种疾病有关,然而,其在非小细胞肺癌(NSCLC)侵袭和转移中的作用尚不清楚。我们的研究探讨了NAT10在非小细胞肺癌中的临床意义和功能方面。
    方法:我们使用癌症基因组图谱(TCGA)和一组98例NSCLC患者研究了NAT10的临床相关性。采用WB,qRT-PCR,和IHC分析,我们评估了NAT10在NSCLC组织中的表达,支气管上皮细胞(BECs),NSCLC细胞系,和小鼠异种移植物。Further,敲低和过表达技术(siRNA,shRNA,和质粒)用于评估NAT10的作用。进行了一系列的试验,包括CCK-8,集落形成,伤口愈合,和transwell分析,阐明NAT10在增殖中的作用,入侵,和转移。此外,我们利用肺癌患者来源的3D类器官,小鼠异种移植模型,和Remodelin(NAT10抑制剂)来证实这些发现。
    结果:我们的研究显示NAT10在非小细胞肺癌组织中高表达,细胞系和小鼠异种移植模型。高NAT10水平与晚期T期相关,淋巴结转移和整体生存率差。NAT10击倒抑制了增殖,入侵,和移民,而NAT10过表达产生相反的效果。此外,NAT10水平降低与E-cadherin水平升高相关,而N-cadherin和波形蛋白表达降低,而升高的NAT10表达显示出对比结果。值得注意的是,重塑蛋白有效减弱NSCLC增殖,入侵,通过上皮-间质转化(EMT)途径抑制NAT10和迁移。
    结论:我们的数据强调NAT10是NSCLC的潜在治疗靶点,提出了通过NAT10抑制靶向干预肺癌的途径。
    BACKGROUND: Lung cancer remains the foremost reason of cancer-related mortality, with invasion and metastasis profoundly influencing patient prognosis. N-acetyltransferase 10 (NAT10) catalyzes the exclusive N (4)-acetylcytidine (ac4C) modification in eukaryotic RNA. NAT10 dysregulation is linked to various diseases, yet its role in non-small cell lung cancer (NSCLC) invasion and metastasis remains unclear. Our study delves into the clinical significance and functional aspects of NAT10 in NSCLC.
    METHODS: We investigated NAT10\'s clinical relevance using The Cancer Genome Atlas (TCGA) and a group of 98 NSCLC patients. Employing WB, qRT-PCR, and IHC analyses, we assessed NAT10 expression in NSCLC tissues, bronchial epithelial cells (BECs), NSCLC cell lines, and mouse xenografts. Further, knockdown and overexpression techniques (siRNA, shRNA, and plasmid) were employed to evaluate NAT10\'s effects. A series of assays were carried out, including CCK-8, colony formation, wound healing, and transwell assays, to elucidate NAT10\'s role in proliferation, invasion, and metastasis. Additionally, we utilized lung cancer patient-derived 3D organoids, mouse xenograft models, and Remodelin (NAT10 inhibitor) to corroborate these findings.
    RESULTS: Our investigations revealed high NAT10 expression in NSCLC tissues, cell lines and mouse xenograft models. High NAT10 level correlated with advanced T stage, lymph node metastasis and poor overall survive. NAT10 knockdown curtailed proliferation, invasion, and migration, whereas NAT10 overexpression yielded contrary effects. Furthermore, diminished NAT10 levels correlated with increased E-cadherin level whereas decreased N-cadherin and vimentin expressions, while heightened NAT10 expression displayed contrasting results. Notably, Remodelin efficiently attenuated NSCLC proliferation, invasion, and migration by inhibiting NAT10 through the epithelial-mesenchymal transition (EMT) pathway.
    CONCLUSIONS: Our data underscore NAT10 as a potential therapeutic target for NSCLC, presenting avenues for targeted intervention against lung cancer through NAT10 inhibition.
    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
    全球范围内,乳腺癌是女性中最常见的癌症。乳腺癌的肿瘤微环境常表现为缺氧。缺氧诱导因子1-α,转录因子,在乳腺癌中被过度表达和激活,通过介导一系列反应在缺氧微环境中发挥关键作用。缺氧诱导因子1-α参与调节下游通路和靶基因,这在缺氧条件下至关重要,包括糖酵解,血管生成,和转移。这些过程通过管理与肿瘤侵袭相关的癌症相关活动,显著促进乳腺癌进展。转移,免疫逃避,和抗药性,导致患者预后不良。因此,低氧诱导因子1-α作为癌症治疗的潜在靶点引起了人们的极大兴趣.目前,针对缺氧诱导因子1-α的药物研究主要处于临床前阶段,强调需要深入了解HIF-1α及其调控途径。预计未来将在临床试验中引入有效的HIF-1α抑制剂,为乳腺癌患者带来新的希望。因此,本文对HIF-1α的结构和功能进行综述,它在推进乳腺癌中的作用,以及对抗HIF-1α依赖性耐药性的策略,强调其治疗潜力。
    Globally, breast cancer stands as the most prevalent form of cancer among women. The tumor microenvironment of breast cancer often exhibits hypoxia. Hypoxia-inducible factor 1-alpha, a transcription factor, is found to be overexpressed and activated in breast cancer, playing a pivotal role in the anoxic microenvironment by mediating a series of reactions. Hypoxia-inducible factor 1-alpha is involved in regulating downstream pathways and target genes, which are crucial in hypoxic conditions, including glycolysis, angiogenesis, and metastasis. These processes significantly contribute to breast cancer progression by managing cancer-related activities linked to tumor invasion, metastasis, immune evasion, and drug resistance, resulting in poor prognosis for patients. Consequently, there is a significant interest in Hypoxia-inducible factor 1-alpha as a potential target for cancer therapy. Presently, research on drugs targeting Hypoxia-inducible factor 1-alpha is predominantly in the preclinical phase, highlighting the need for an in-depth understanding of HIF-1α and its regulatory pathway. It is anticipated that the future will see the introduction of effective HIF-1α inhibitors into clinical trials, offering new hope for breast cancer patients. Therefore, this review focuses on the structure and function of HIF-1α, its role in advancing breast cancer, and strategies to combat HIF-1α-dependent drug resistance, underlining its therapeutic potential.
    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
    白细胞介素-6(IL-6)和缺氧诱导因子-1α(HIF-1α)在上皮间质转化(EMT)和肿瘤发生发展中起重要作用。以前的研究表明,IL-6促进EMT,入侵,通过激活STAT3/HIF-1α通路在上皮性卵巢癌(EOC)细胞中的转移。MicroRNA(miRNA)是一种非编码小RNA,在肿瘤的发生发展中起着重要作用。值得注意的是,let-7和miR-200家族在EOC中显著改变。然而,IL-6是否通过STAT3/HIF-1α信号调节Let-7和miR-200家族的表达以诱导EOC中的EMT仍知之甚少.在这项研究中,我们使用两种EOC细胞系进行了体外和体内研究,SKOV3和OVCAR3细胞。我们的发现表明,IL-6下调Let-7c和miR-200c的mRNA水平,同时通过STAT3/HIF-1α信号在EOC细胞和体内上调其靶基因HMGA2和ZEB1。此外,探讨HIF-1α对miRNAs的调控作用,可以利用外源性HIF阻断剂YC-1和内源性HIF-1α的高表达或抑制。两种方法都可以证实下游分子HIF-1α抑制Let-7c和miR-200c的表达和功能。进一步的机制研究表明,Let-7c或miR-200c的过表达可以逆转IL-6诱导的EOC细胞的恶性演变,包括EMT,入侵,和转移。因此,我们的结果表明,IL-6通过STAT3/HIF-1α途径调节Let-7c和miR-200c的表达,从而促进EMT,入侵,和EOC细胞中的转移。
    Interleukin-6 (IL-6) and hypoxia-inducible factor-1α (HIF-1α) play important roles in epithelial-mesenchymal transformation (EMT) and tumor development. Previous studies have demonstrated that IL-6 promotes EMT, invasion, and metastasis in epithelial ovarian cancer (EOC) cells by activating the STAT3/HIF-1α pathway. MicroRNA (miRNA) is non-coding small RNAs that also play an important role in tumor development. Notably, Let-7 and miR-200 families are prominently altered in EOC. However, whether IL-6 regulates the expression of Let-7 and miR-200 families through the STAT3/HIF-1α signaling to induce EMT in EOC remains poorly understood. In this study, we conducted in vitro and in vivo investigations using two EOC cell lines, SKOV3, and OVCAR3 cells. Our findings demonstrate that IL-6 down-regulates the mRNA levels of Let-7c and miR-200c while up-regulating their target genes HMGA2 and ZEB1 through the STAT3/HIF-1α signaling in EOC cells and in vivo. Additionally, to explore the regulatory role of HIF-1α on miRNAs, both exogenous HIF blockers YC-1 and endogenous high expression or inhibition of HIF-1α can be utilized. Both approaches can confirm that the downstream molecule HIF-1α inhibits the expression and function of Let-7c and miR-200c. Further mechanistic research revealed that the overexpression of Let-7c or miR-200c can reverse the malignant evolution of EOC cells induced by IL-6, including EMT, invasion, and metastasis. Consequently, our results suggest that IL-6 regulates the expression of Let-7c and miR-200c through the STAT3/HIF-1α pathway, thereby promoting EMT, invasion, and metastasis in EOC cells.
    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: Preprint
    组蛋白去乙酰化酶(HDAC)是通过从组蛋白上的赖氨酸残基中去除乙酰基来调节细胞的基因表达和表观遗传变化的酶之一,允许组蛋白更紧密地包裹DNA并抑制肿瘤抑制基因。HDAC抑制剂通过限制HDAC酶的作用机制,在抑制肿瘤细胞增殖中发挥重要作用,导致向赖氨酸添加乙酰基。Mocetinostat,也以其化学名称(MGCD0103)而闻名,是一种新型的同种型选择性HDAC酶,其明确靶向HDAC同种型抑制Class1(HDAC1,2,3,8)和IV类(HDAC11)酶。它于2010年被批准用于治疗霍奇金淋巴瘤的II期试验。我们的研究表明,不同剂量的Mocetinostat抑制胶质母细胞瘤细胞的生长,转移,并诱导胶质母细胞瘤细胞C6和T98G的凋亡和分化。Westernblot已显示MGCD0103具有许多控制胶质母细胞瘤癌细胞的生物学活性。MGCD0103可以调节细胞中几种途径的分子机制,例如抑制PI3K/AKT途径和抑制HDAC1酶活性,这些酶活性负责癌症起始和进展中的许多生物过程。高剂量的Mocetinostat药物通过增加促凋亡蛋白(BAX)和降低抗凋亡蛋白水平(Bid,Bcl2)。此外,莫西替诺他上调抑癌基因的表达,下调E2f1转录因子的基因表达。此外,通过激活分化标志物GFAP并阻止未分化标志物表达(Id2,N-Myc)促进MGCDO103诱导的分化。MGCD0103是一种有效的抗癌药物,在治疗胶质母细胞瘤细胞中至关重要。
    Histon deacetylase (HDAC) enzyme is one of the enzymes involved in regulating gene expression and epigenetic alternation of cells by removing acetyl groups from lysine residue on a histone, allowing the histones to wrap the DNA more tightly and suppressing a tumor-suppressing gene. HDAC inhibitors play an important role in inhibiting the proliferation of tumor cells by restricting the mechanism of action of HDAC enzyme, leading to the addition of acetyl groups to lysine. Mocetinostat, also known by its chemical name (MGCD0103), is a novel isotype selective HDAC enzyme that explicitly targets HDAC isoforms inhibiting Class1(HDAC 1,2,3,8) and Class IV (HDAC11) enzymes. It was approved for treating the phase II trial of Hodgkin\'s lymphoma in 2010. Our study revealed that different doses of Mocetinostat inhibit the growth of glioblastoma cells, metastasis, and angiogenesis and induce the apoptosis and differentiation of glioblastoma cells C6 and T98G. Western blot has shown that MGCD0103 has many biological activities to control glioblastoma cancer cells. MGCD0103 can modulate the molecular mechanism for several pathways in cells, such as inhibition of the PI3K/AKT pathway and suppression of HDAC1 enzyme activity in charge of many biological processes in the initiation and progression of cancer. The high doses of Mocetinostat drug significantly induce apoptosis and suppress cancer cell proliferation through increased pro-apoptotic proteins (BAX) and a down level of anti-apoptotic proteins(Bid, Bcl2). Also, the mocetinostat upregulated the expression of the tumor suppressor gene and downregulated the gene expression of the E2f1 transcription factor. Additionally, MGCDO103-induced differentiation was facilitated by activating the differentiation marker GFAP and preventing the undifferentiation marker from expression (Id2, N-Myc). The MGCD0103 is a potent anticancer drug crucial in treating glioblastoma cells.
    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
    HMGA2,一个关键的转录因子,作为多种侵袭性恶性肿瘤进展的多功能调节因子。在这项研究中,质谱用于鉴定可能与HMGA2相互作用的泛素特异性蛋白酶,并且USP48被鉴定为HMGA2的去泛素化酶。USP48的强制表达通过抑制HMGA2的降解而显著增加其蛋白水平,而USP48的剥夺促进了HMGA2的降解,从而抑制肿瘤的侵袭和转移。我们发现USP48在赖氨酸258处经历SUMO化,这增强了其对HMGA2的结合亲和力。通过随后的小分子表型筛选,我们确定DUB-IN-2是一种非常有效的USP48药物抑制剂.有趣的是,靶向USP48的小分子抑制剂诱导HMGA2的去稳定化。临床上,癌组织中USP48或HMGA2的上调指示患有结直肠癌(CRC)的患者的不良预后。总的来说,我们的研究不仅阐明了参与HMGA2稳定性的DUB的调控机制,而且验证了USP48作为CRC的潜在治疗靶点,但也将DUB-IN-2确定为USP48的有效抑制剂和有希望的CRC治疗候选物。
    HMGA2, a pivotal transcription factor, functions as a versatile regulator implicated in the progression of diverse aggressive malignancies. In this study, mass spectrometry was employed to identify ubiquitin-specific proteases that potentially interact with HMGA2, and USP48 was identified as a deubiquitinating enzyme of HMGA2. The enforced expression of USP48 significantly increased HMGA2 protein levels by inhibiting its degradation, while the deprivation of USP48 promoted HMGA2 degradation, thereby suppressing tumor invasion and metastasis. We discovered that USP48 undergoes SUMOylation at lysine 258, which enhances its binding affinity to HMGA2. Through subsequent phenotypic screening of small molecules, we identified DUB-IN-2 as a remarkably potent pharmacological inhibitor of USP48. Interestingly, the small-molecule inhibitor targeting USP48 induces destabilization of HMGA2. Clinically, upregulation of USP48 or HMGA2 in cancerous tissues is indicative of poor prognosis for patients with colorectal cancer (CRC). Collectively, our study not only elucidates the regulatory mechanism of DUBs involved in HMGA2 stability and validates USP48 as a potential therapeutic target for CRC, but also identifies DUB-IN-2 as a potent inhibitor of USP48 and a promising candidate for CRC treatment.
    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
    环状RNA(circularRNAs,circRNAs)在癌症的发生和发展中起重要作用。探讨并阐明黑色素瘤特异性环状RNA的临床意义及其潜在的分子机制。通过qRT-PCR和ISH确认黑素瘤细胞和组织中的CircROR1表达。进行qRT-PCR和Western印迹以测量CCNE1,KAT2A,MMP9和TIMP2。MTT,进行Transwell和伤口愈合试验以评估细胞增殖,侵袭和转移。建立异种移植小鼠模型以进一步验证体内CircROR1/CCNE1轴。进行RNA下拉和RIP测定以检测KAT2A和CircROR1的直接相互作用。使用ChIP测定法来研究CCNE1启动子中H3K9ac乙酰化的富集。CircROR1在转移性黑色素瘤细胞和组织中显著上调,促进扩散,体外侵袭和转移以及体内肿瘤生长。CircROR1过表达增加CCNE1和MMP9蛋白表达,降低TIMP2蛋白表达。功能性挽救试验表明CircROR1在通过CCNE1促进恶性进展中发挥作用。CircROR1与KAT2A蛋白特异性结合而不影响其表达。CircROR1过表达通过募集KAT2A增加了CCNE1启动子区的H3K9ac修饰水平,从而上调CCNE1表达。CircROR1通过KAT2A介导的组蛋白乙酰化上调CCNE1表达。我们的研究证实了CircROR1在黑色素瘤侵袭和转移中的关键作用,CircROR1可以作为黑色素瘤治疗的潜在治疗靶点。
    Circular RNAs (circRNAs) play important roles in cancer occurrence and progression. To explore and elucidate the clinical significance of specific circular RNA in melanoma and its potential molecular mechanism. CircROR1 expression in melanoma cells and tissues was confirmed by qRT-PCR and ISH. qRT-PCR and Western blotting were performed to measure the levels of CCNE1, KAT2A, MMP9 and TIMP2. MTT, Transwell and wound healing assays were performed to evaluate cell proliferation, invasion and metastasis. A xenograft mouse model was established to further verify the CircROR1/CCNE1 axis in vivo. RNA pull-down and RIP assays were performed to detect the direct interaction KAT2A and CircROR1. A ChIP assay was used to investigate the enrichment of H3K9ac acetylation in the CCNE1 promoter. CircROR1 was significantly upregulated in metastatic melanoma cells and tissues, promoting proliferation, invasion and metastasis in vitro and tumour growth in vivo. CircROR1 overexpression increased CCNE1 and MMP9 protein expression and decreased TIMP2 protein expression. Functional rescue assays demonstrated that CircROR1 played a role in promoting malignant progression through CCNE1. CircROR1 specifically bound to the KAT2A protein without affecting its expression. CircROR1 overexpression increased the level of H3K9ac modification in the CCNE1 promoter region by recruiting KAT2A, thus upregulating CCNE1 expression. CircROR1 upregulates CCNE1 expression through KAT2A-mediated histone acetylation. Our research confirms the critical role of CircROR1 in melanoma invasion and metastasis, and CircROR1 could serve as a potential therapeutic target for melanoma treatment.
    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
    中药(TCM)已用于治疗三阴性乳腺癌(TNBC),预后不良的乳腺癌亚型。临床研究证实,三英方配方(SYF),中药处方,具有明显的抑制乳腺癌复发和转移的作用,延长患者生存期,减少临床症状。然而,其活性成分和分子机制尚不清楚。在这项研究中,组成SYF的每种草药的活性成分及其靶蛋白从传统中药系统药理学数据库获得。乳腺癌相关基因从GeneCards数据库获得。通过分析上述数据来鉴定与乳腺癌中SYF治疗相关的主要靶标和途径。通过分子对接分析,我们发现活性成分槲皮素和木犀草素与关键靶标KDR1,PPARG,SOD1和VCAM1。体外实验验证SYF能降低细胞增殖,迁移,和TNBC细胞的侵袭能力。使用TNBC异种移植小鼠模型,我们证明SYF在体内可以延缓肿瘤的生长并有效抑制乳腺癌肺转移的发生。PPARG,SOD1、KDR1和VCAM1均受SYF调控,在SYF介导的抑制TNBC复发和转移中起重要作用。
    Traditional Chinese medicine (TCM) has been used to treat triple-negative breast cancer (TNBC), a breast cancer subtype with poor prognosis. Clinical studies have verified that the Sanyingfang formula (SYF), a TCM prescription, has obvious effects on inhibiting breast cancer recurrence and metastasis, prolonging patient survival, and reducing clinical symptoms. However, its active ingredients and molecular mechanisms are still unclear. In this study, the active ingredients of each herbal medicine composing SYF and their target proteins are obtained from the Traditional Chinese Medicine Systems Pharmacology database. Breast cancer-related genes are obtained from the GeneCards database. Major targets and pathways related to SYF treatment in breast cancer are identified by analyzing the above data. By conducting molecular docking analysis, we find that the active ingredients quercetin and luteolin bind well to the key targets KDR1, PPARG, SOD1, and VCAM1. In vitro experiments verify that SYF can reduce the proliferation, migration, and invasion ability of TNBC cells. Using a TNBC xenograft mouse model, we show that SYF could delay tumor growth and effectively inhibit the occurrence of breast cancer lung metastasis in vivo. PPARG, SOD1, KDR1, and VCAM1 are all regulated by SYF and may play important roles in SYF-mediated inhibition of TNBC recurrence and metastasis.
    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:异常可变剪接(AS)有助于肝内侵袭和转移扩散,导致肝细胞癌(HCC)的高致死率。
    目的:本研究旨在探讨UPF3B-S(一种截短的致癌剪接变体)在HCC转移中的功能意义。
    方法:采用Basescope法分析UPF3B-SmRNA在组织和细胞中的表达。RNA免疫沉淀,体外和体内模型用于探讨UPF3B-S的作用和潜在的机制。
    结果:我们显示剪接因子HnRNPR通过其RRM2结构域与UPF3B的pre-mRNA结合,产生外显子8排斥截短的剪接变体UPF3B-S。UPF3B-S的高表达与HCC患者的肿瘤转移和不利的总生存期相关。UPF3B-S的敲减显着抑制体外和体内HCC细胞的侵袭和迁移能力。机械上,UPF3B-S蛋白靶向CDH1mRNA的3'-UTR以增强CDH1mRNA的降解,这导致E-cadherin的下调和上皮-间质转化的激活。UPF3B-S的过表达增强LATS1的去磷酸化和YAP1的核积累以触发Hippo信号通路。
    结论:我们的研究结果表明,HnRNPR诱导的UPF3B-S通过耗尽CDH1mRNA和调节YAP1-Hippo信号促进HCC侵袭和转移。UPF3B-S可能作为侵袭性HCC临床管理的有希望的生物标志物。
    BACKGROUND: Abnormal alternative splicing (AS) contributes to aggressive intrahepatic invasion and metastatic spread, leading to the high lethality of hepatocellular carcinoma (HCC).
    OBJECTIVE: This study aims to investigate the functional implications of UPF3B-S (a truncated oncogenic splice variant) in HCC metastasis.
    METHODS: Basescope assay was performed to analyze the expression of UPF3B-S mRNA in tissues and cells. RNA immunoprecipitation, and in vitro and in vivo models were used to explore the role of UPF3B-S and the underlying mechanisms.
    RESULTS: We show that splicing factor HnRNPR binds to the pre-mRNA of UPF3B via its RRM2 domain to generate an exon 8 exclusion truncated splice variant UPF3B-S. High expression of UPF3B-S is correlated with tumor metastasis and unfavorable overall survival in patients with HCC. The knockdown of UPF3B-S markedly suppresses the invasive and migratory capacities of HCC cells in vitro and in vivo. Mechanistically, UPF3B-S protein targets the 3\'-UTR of CDH1 mRNA to enhance the degradation of CDH1 mRNA, which results in the downregulation of E-cadherin and the activation of epithelial-mesenchymal transition. Overexpression of UPF3B-S enhances the dephosphorylation of LATS1 and the nuclear accumulation of YAP1 to trigger the Hippo signaling pathway.
    CONCLUSIONS: Our findings suggest that HnRNPR-induced UPF3B-S promotes HCC invasion and metastasis by exhausting CDH1 mRNA and modulating YAP1-Hippo signaling. UPF3B-S could potentially serve as a promising biomarker for the clinical management of invasive HCC.
    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