Smyd2

SMYD2
  • 文章类型: Journal Article
    先天性巨结肠(HSCR)的发病机制复杂。最近,已经发现组蛋白修饰可以改变遗传易感性并在增殖中起重要作用,神经c细胞的分化和迁移。H3K36甲基化在基因转录激活和表达中起重要作用,但其在HSCR中的致病机制尚未被研究。本研究旨在阐明其在HSCR中的作用和分子机制。蛋白质印迹分析,免疫组织化学(IHC)和逆转录定量PCR(RT-qPCR)用于研究儿童扩张和狭窄结肠组织切片中H3K36甲基化和甲基转移酶的水平。我们证实SMYD2是H3K36甲基化差异的主要原因,并影响HSCR中的细胞增殖和迁移。随后,m6ARNA甲基化的定量检测显示SMYD2可以改变m6A甲基化水平。蛋白质印迹分析,RT-qPCR,免疫共沉淀(co-IP),和免疫荧光共定位用于证实SMYD2可以调节METTL3表达并影响m6A甲基化,影响细胞增殖和迁移。这些结果证实,H3K36甲基转移酶SMYD2可以通过调节METTL3影响赫氏弹簧病中的细胞增殖和迁移。我们的研究表明,H3K36甲基化在HSCR中起重要作用,证实甲基转移酶SMYD2可以经由过程调控METTL3表达来影响m6A甲基化程度和肠神经系统发育。
    The pathogenesis of Hirschsprung\'s disease (HSCR) is complex. Recently, it has been found that histone modifications can alter genetic susceptibility and play important roles in the proliferation, differentiation and migration of neural crest cells. H3K36 methylation plays a significant role in gene transcriptional activation and expression, but its pathogenic mechanism in HSCR has not yet been studied. This study aimed to elucidate its role and molecular mechanism in HSCR. Western blot analysis, immunohistochemistry (IHC) and reverse transcription-quantitative PCR (RT‒qPCR) were used to investigate H3K36 methylation and methyltransferase levels in dilated and stenotic colon tissue sections from children with. We confirm that SMYD2 is the primary cause of differential H3K36 methylation and influences cell proliferation and migration in HSCR. Subsequently, quantitative detection of m6A RNA methylation revealed that SMYD2 can alter m6A methylation levels. Western blot analysis, RT-qPCR, co-immunoprecipitation (co-IP), and immunofluorescence colocalization were utilized to confirm that SMYD2 can regulate METTL3 expression and affect m6A methylation, affecting cell proliferation and migration. These results confirm that the H3K36 methyltransferase SMYD2 can affect cell proliferation and migration in Hirschsprung\'s disease by regulating METTL3. Our study suggested that H3K36 methylation plays an important role in HSCR, confirming that the methyltransferase SMYD2 can affect m6A methylation levels and intestinal nervous system development by regulating METTL3 expression.
    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:胰腺癌(PC)的特征是预后不良和治疗选择有限。Ferroptosis在癌症中起着重要作用,含有SET和MYND结构域的蛋白2(SMYD2)在各种癌症中广泛表达。然而,SMYD2在调节PC铁凋亡中的作用仍未被探索。本研究旨在探讨SMYD2在介导铁凋亡中的作用及其在PC进展中的机制意义。
    方法:SMYD2,c-Myc,和NCOA4在PC组织中进行评估,和肿瘤周围组织。在人PC细胞系中进一步分析SMYD2表达。在BxPC3细胞,c-Myc的表达,NCOA4,自噬相关蛋白,和线粒体形态,在用si-SMYD2转染并用自噬抑制剂和铁凋亡抑制剂处理后进行评估。使用流式细胞术和ELISA测定定量铁凋亡水平。进行RNA免疫沉淀以阐明c-Myc和NCOA4mRNA之间的相互作用。构建异种移植小鼠模型以验证SMYD2敲低对PC生长的影响。
    结果:发现SMYD2和c-Myc在PC组织中高表达,而NCOA4显示表达降低。在研究的PC细胞系中,BxPC3细胞表现出最高的SMYD2表达。SMYD2敲除导致c-Myc水平降低,NCOA4表达增加,自噬相关蛋白表达减少,线粒体收缩,铁中毒水平升高。此外,确定了c-Myc和NCOA4之间的相互作用。在体内,SMYD2敲低抑制肿瘤生长。
    结论:靶向SMYD2通过c-Myc/NCOA4轴促进铁蛋白吞噬依赖性铁凋亡抑制PC进展。这些发现为PC的潜在诊断和治疗策略提供了见解。
    BACKGROUND: Pancreatic cancer (PC) is characterized by a poor prognosis and limited treatment options. Ferroptosis plays an important role in cancer, SET and MYND domain-containing protein 2 (SMYD2) is widely expressed in various cancers. However, the role of SMYD2 in regulating ferroptosis in PC remains unexplored. This study aimed to investigate the role of SMYD2 in mediating ferroptosis and its mechanistic implications in PC progression.
    METHODS: The levels of SMYD2, c-Myc, and NCOA4 were assessed in PC tissues, and peritumoral tissues. SMYD2 expression was further analyzed in human PC cell lines. In BxPC3 cells, the expression of c-Myc, NCOA4, autophagy-related proteins, and mitochondrial morphology, was evaluated following transfection with si-SMYD2 and treatment with autophagy inhibitors and ferroptosis inhibitors. Ferroptosis levels were quantified using flow cytometry and ELISA assays. RNA immunoprecipitation was conducted to elucidate the interaction between c-Myc and NCOA4 mRNA. A xenograft mouse model was constructed to validate the impact of SMYD2 knockdown on PC growth.
    RESULTS: SMYD2 and c-Myc were found to be highly expressed in PC tissues, while NCOA4 showed reduced expression. Among the PC cell lines studied, BxPC3 cells exhibited the highest SMYD2 expression. SMYD2 knockdown led to decreased c-Myc levels, increased NCOA4 expression, reduced autophagy-related protein expression, mitochondrial shrinkage, and heightened ferroptosis levels. Additionally, an interaction between c-Myc and NCOA4 was identified. In vivo, SMYD2 knockdown inhibited tumor growth.
    CONCLUSIONS: Targeting SMYD2 inhibits PC progression by promoting ferritinophagy-dependent ferroptosis through the c-Myc/NCOA4 axis. These findings provide insights into potential diagnostic and therapeutic strategies for PC.
    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
    百草枯(PQ)中毒导致肺部不可逆转的纤维化,死亡率高,没有已知的解毒剂。在这项研究中,我们研究了SET和MYND结构域包含2(SMYD2)对PQ诱导的肺纤维化(PF)的影响及其潜在机制。我们通过腹膜内注射PQ(20mg/kg)和体外PQ(25μM)损伤的MLE-12细胞模型建立了体内PQ诱导的PF小鼠模型。在给药的第15天,组织损伤,炎症,和纤维化小鼠使用各种方法进行评估,包括常规血细胞计数,血液生物化学,血气分析,西方印迹,H&E染色,ELISA,Masson染色,和免疫荧光。研究结果表明,AZ505给药减轻了组织损伤,炎症,和PQ中毒小鼠的胶原沉积。机械上,体内和体外实验均表明,AZ505治疗通过下调GLI发病机制相关2(GLIPR2)和ERK/p38途径来抑制PQ诱导的上皮-间质转化(EMT)过程。进一步的研究表明,SMYD2抑制降低GLIPR2甲基化并促进GLIPR2泛素化,在暴露于PQ的MLE-12细胞中导致GLIPR2不稳定。此外,体外进行的拯救实验表明,GLIPR2过表达消除了AZ505对ERK/p38途径和EMT的抑制作用。我们的结果表明,SMYD2抑制剂AZ505可以通过调节PQ诱导的PF中的GLIPR2/ERK/p38轴来抑制EMT过程。
    Paraquat (PQ) poisoning leads to irreversible fibrosis in the lungs with high mortality and no known antidote. In this study, we investigated the effect of the SET and MYND domain containing 2 (SMYD2) on PQ-induced pulmonary fibrosis (PF) and its potential mechanisms. We established an in vivo PQ-induced PF mouse model by intraperitoneal injection of PQ (20 mg/kg) and in vitro PQ (25 μM)-injured MLE-12 cell model. On the 15th day of administration, tissue injury, inflammation, and fibrosis in mice were evaluated using various methods including routine blood counts, blood biochemistry, blood gas analysis, western blotting, H&E staining, ELISA, Masson staining, and immunofluorescence. The findings indicated that AZ505 administration mitigated tissue damage, inflammation, and collagen deposition in PQ-poisoned mice. Mechanistically, both in vivo and in vitro experiments revealed that AZ505 treatment suppressed the PQ-induced epithelial-mesenchymal transition (EMT) process by downregulating GLI pathogenesis related 2 (GLIPR2) and ERK/p38 pathway. Further investigations demonstrated that SMYD2 inhibition decreased GLIPR2 methylation and facilitated GLIPR2 ubiquitination, leading to GLIPR2 destabilization in PQ-exposed MLE-12 cells. Moreover, rescue experiments conducted in vitro demonstrated that GLIPR2 overexpression eliminated the inhibitory effect of AZ505 on the ERK/p38 pathway and EMT. Our results reveal that the SMYD2 inhibitor AZ505 may act as a novel therapeutic candidate to suppress the EMT process by modulating the GLIPR2/ERK/p38 axis in PQ-induced PF.
    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
    通过精确协调干细胞和祖细胞的增殖和分化来实现组织形成和器官稳态。虽然这些过程的失调会导致退行性疾病或癌症,它们的分子相互作用尚不清楚。在这里,我们表明人多能干细胞(hPSC)自我更新向分化的转变与不同细胞周期蛋白依赖性激酶抑制剂(CDKIs)的诱导有关。在hPSC中,激活素/节点/TGFβ信号通过SMAD2/3-NANOG-OCT4-EZH2-SNON转录复合物将CDKIs维持在平衡状态。随着逐渐分化,CDKIs由SMAD2/3-SMYD2和发育调节因子如EOMES之间的连续转录复合物诱导,从而延长G1期。这个,反过来,通过阻断其接头磷酸化诱导SMAD2/3转录活性。这种SMAD2/3-CDKI正反馈回路驱动了可用于生产用于治疗应用的细胞的多能性和逐步细胞命运规范的退出。我们的研究揭示了细胞命运规范如何与细胞周期动力学相互关联的基本机制,并为控制组织自我形成的自主电路提供了见解。
    Tissue formation and organ homeostasis is achieved by precise coordination of proliferation and differentiation of stem cells and progenitors. While deregulation of these processes can result in degenerative disease or cancer, their molecular interplays remain unclear. Here we show that the switch of human pluripotent stem cell (hPSC) self-renewal to differentiation is associated with the induction of distinct cyclin dependent kinase inhibitors (CDKIs). In hPSCs, Activin/Nodal/TGFβ signalling maintains CDKIs in a poised state via SMAD2/3-NANOG-OCT4-EZH2-SNON transcriptional complex. Upon gradual differentiation, CDKIs are induced by successive transcriptional complexes between SMAD2/3-SMYD2 and developmental regulators such as EOMES, thereby lengthening the G1 phase. This, in turn, induces SMAD2/3 transcriptional activity by blocking its linker phosphorylation. Such SMAD2/3-CDKI positive feedback loops drive the exit from pluripotency and stepwise cell fate specification that could be harnessed for producing cells for therapeutic applications. Our study uncovers fundamental mechanisms how cell fate specification is interconnected to cell cycle dynamics and provides insight to autonomous circuitries governing tissue self-formation.
    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
    PIK3CA的激活突变常见于雌激素受体阳性(ER+)乳腺癌,磷脂酰肌醇3-激酶(PI3K)抑制剂alpelisib与抗ER抑制剂的组合被批准用于治疗。我们先前已经证明PI3K途径通过染色质修饰剂KMT2D的磷酸化来调节ER活性。这里,我们在KMT2D上发现了一个甲基化位点,在K1330与S1331直接相邻,由赖氨酸甲基转移酶SMYD2催化。SMYD2丢失减弱alpelisib诱导的KMT2D染色质结合和alpelisib介导的基因表达变化,包括ER依赖性转录。SMYD2的敲除或药理抑制使乳腺癌细胞敏感,患者来源的类器官,和肿瘤对PI3K/AKT的抑制和内分泌治疗部分通过KMT2DK1330甲基化。一起,我们的发现揭示了翻译后修饰之间的调节串扰,这些修饰可以微调染色质的KMT2D功能。这提供了SMYD2抑制剂与PI3Kα/AKT抑制剂组合用于治疗ER+/PIK3CA突变型乳腺癌的基本原理。
    Activating mutations in PIK3CA are frequently found in estrogen-receptor-positive (ER+) breast cancer, and the combination of the phosphatidylinositol 3-kinase (PI3K) inhibitor alpelisib with anti-ER inhibitors is approved for therapy. We have previously demonstrated that the PI3K pathway regulates ER activity through phosphorylation of the chromatin modifier KMT2D. Here, we discovered a methylation site on KMT2D, at K1330 directly adjacent to S1331, catalyzed by the lysine methyltransferase SMYD2. SMYD2 loss attenuates alpelisib-induced KMT2D chromatin binding and alpelisib-mediated changes in gene expression, including ER-dependent transcription. Knockdown or pharmacological inhibition of SMYD2 sensitizes breast cancer cells, patient-derived organoids, and tumors to PI3K/AKT inhibition and endocrine therapy in part through KMT2D K1330 methylation. Together, our findings uncover a regulatory crosstalk between post-translational modifications that fine-tunes KMT2D function at the chromatin. This provides a rationale for the use of SMYD2 inhibitors in combination with PI3Kα/AKT inhibitors in the treatment of ER+/PIK3CA mutant breast cancer.
    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
    冠状动脉再狭窄是冠心病患者远期预后不良的重要原因。这里,我们显示,赖氨酸甲基转移酶SMYD2在细胞核中的表达在血清和PDGF-BB诱导的血管平滑肌细胞(VSMC)中显著升高,以及颈动脉损伤引起的新生内膜增生组织。Smyd2在VSMC中的过表达(Smyd2-vTg)促进,但用其特异性抑制剂LLY-507或SMYD2敲低治疗显著抑制小鼠VSMC表型转换和颈动脉损伤诱导的新内膜形成。转录组测序显示SMYD2敲低抑制血清反应因子(SRF)靶基因的表达,并且SRF过表达在很大程度上逆转了SMYD2敲低对VSMC增殖的抑制作用。HDAC3直接与SRF相互作用并使其脱乙酰,从而增强VSMC中的SRF转录活性。此外,SMYD2通过在其启动子处H3K36的三甲基化促进HDAC3表达。HDAC3的特异性抑制剂RGFP966不仅能抵消SMYD2过表达对VSMC的促增殖作用,但也抑制小鼠颈动脉损伤诱导的新内膜形成。HDAC3以脱乙酰酶活性依赖性方式部分消除了SMYD2敲低对VSMC增殖的抑制作用。我们的结果表明,SMYD2-HDAC3-SRF轴构成了调节VSMC表型转换和新生内膜增生的新型和关键的表观遗传机制。
    Coronary restenosis is an important cause of poor long-term prognosis in patients with coronary heart disease. Here, we show that lysine methyltransferase SMYD2 expression in the nucleus is significantly elevated in serum- and PDGF-BB-induced vascular smooth muscle cells (VSMCs), and in tissues of carotid artery injury-induced neointimal hyperplasia. Smyd2 overexpression in VSMCs (Smyd2-vTg) facilitates, but treatment with its specific inhibitor LLY-507 or SMYD2 knockdown significantly inhibits VSMC phenotypic switching and carotid artery injury-induced neointima formation in mice. Transcriptome sequencing revealed that SMYD2 knockdown represses the expression of serum response factor (SRF) target genes and that SRF overexpression largely reverses the inhibitory effect of SMYD2 knockdown on VSMC proliferation. HDAC3 directly interacts with and deacetylates SRF, which enhances SRF transcriptional activity in VSMCs. Moreover, SMYD2 promotes HDAC3 expression via tri-methylation of H3K36 at its promoter. RGFP966, a specific inhibitor of HDAC3, not only counteracts the pro-proliferation effect of SMYD2 overexpression on VSMCs, but also inhibits carotid artery injury-induced neointima formation in mice. HDAC3 partially abolishes the inhibitory effect of SMYD2 knockdown on VSMC proliferation in a deacetylase activity-dependent manner. Our results reveal that the SMYD2-HDAC3-SRF axis constitutes a novel and critical epigenetic mechanism that regulates VSMC phenotypic switching and neointimal hyperplasia.
    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
    据报道,中性粒细胞胞外陷阱(NETs)的形成与癌症转移有关。本研究旨在探讨NETs对胃癌(GC)细胞转移的影响及其机制。在GC患者的血浆中测量NETs。然后,用NETs处理GC细胞以评估细胞活力,迁移,使用细胞计数试剂盒8和Transwell测定法进行侵袭,建立肝转移和异种移植肿瘤小鼠模型以评估肿瘤生长和转移。使用乙酰化RNA免疫沉淀评估由NAT10介导的SET和含MYND结构域2(SMYD2)的N4-乙酰胞苷(ac4C)修饰。结果显示,GC患者血浆中NETs水平升高,特别是那些转移性GC。此外,GC细胞与NETs共同处理可促进细胞活力,移民和入侵,而NAT10或SMYD2敲除消除了这种作用。NAT10还促进了SMYD2的ac4C修饰,从而增加了SMYD2的稳定性。此外,NETs在体内促进GC细胞在肝脏中的转移。总的来说,本研究的结果表明,NETs通过NAT10介导的ac4C修饰SMYD2促进GC细胞转移。这些发现表明,抑制NETs的形成可能是减弱GC进展的有效方法。
    It has been reported that the formation of neutrophil extracellular traps (NETs) is associated with cancer metastasis. The current study aimed to explore the effects of NETs on gastric cancer (GC) cell metastasis and uncover their underlying mechanism. NETs were measured in the plasma of patients with GC. Then, GC cells were treated with NETs to assess cell viability, migration, and invasion using cell counting kit 8 and Transwell assay, The liver metastasis and xenograft tumor mouse models were established to assess tumor growth and metastasis. The N4-acetylcytidine (ac4C) modification of SET and MYND domain containing 2 (SMYD2) mediated by NAT10 was evaluated using acetylated RNA immunoprecipitation. The results showed that the level of NETs was increased in the plasma of patients with GC, particularly in those with metastatic GC. In addition, GC cell co-treatment with NETs promoted cell viability, migration and invasion, while NAT10 or SMYD2 knockdown abrogated this effect. NAT10 also promoted the ac4C modification of SMYD2, thus increasing SMYD2 stability. Furthermore, NETs promoted the metastasis of GC cells in the liver in vivo. Overall, the results of the present study demonstrated that NETs promoted GC cell metastasis via the NAT10-mediated ac4C modification of SMYD2. These findings suggested that inhibiting the formation of NETs could be an effective approach for attenuating GC progression.
    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:SMYD2是含有SET和MYND结构域的家族SMYD的蛋白质。它可以甲基化各种组蛋白和非组蛋白癌症相关蛋白的赖氨酸残基,在肿瘤发生中起关键作用。尽管新出现的证据支持SMYD2在癌症进展中的关联,但其最终效果尚不清楚。因此,需要进一步研究该基因与癌症进展的关系。在目前的研究中,研究者利用TCGA数据确定SMYD2在11种癌症中的潜在致癌作用.转录表达,存活率,突变,丰富的途径,使用不同的生物信息学工具和服务器探索SMYD2的基因本体论。此外,我们还研究了多种癌症类型中SMYD2基因表达与免疫细胞浸润之间的相关性。
    结果:研究结果表明SMYD2的高表达与癌症事件显著相关。在CESC和KIRC,SMYD2的mRNA表达与总生存期(OS)显着相关。在BRCA,KIRC,COAD,和HNSC,SMYD2的mRNA表达与无病生存期(DFS)显着相关。我们发现了15个错觉,4截断,4融合,和1个剪接类型的突变。SMYD2的表达与6种癌症的肿瘤纯度和免疫细胞浸润显着相关。GNPAT基因与SMYD2高度相关。共表达基因的重要途径和基因本体论(GO)术语与癌症形成相关的各种过程相关。
    结论:总的来说,我们的数据驱动结果可能为了解SMYD2的致癌作用提供合理全面的见解.这表明SMYD2可能用作鉴定各种人类肿瘤的新生物标志物的重要靶标。
    BACKGROUND: SMYD2 is a protein of the SET and MYND domain-containing family SMYD. It can methylate the lysine residue of various histone and nonhistone cancer-related proteins and plays a critical role in tumorigenesis. Although emerging evidence supports the association of SMYD2 in the progression of cancers, but its definitive effect is not yet clear. Therefore, further study of the gene in relation with cancer progression needs to be conducted. In the current study, investigators used TCGA data to determine the potential carcinogenic effect of SMYD2 in 11 cancer types. The transcriptional expression, survival rate, mutations, enriched pathways, and Gene Ontology of the SMYD2 were explored using different bioinformatics tools and servers. In addition, we also examined the correlation between SMYD2 gene expression and immunocyte infiltration in multiple cancer types.
    RESULTS: Findings revealed that higher expression of SMYD2 was significantly correlated with cancer incidents. In CESC and KIRC, the mRNA expression of SMYD2 was significantly correlated with overall survival (OS). In BRCA, KIRC, COAD, and HNSC, the mRNA expression of SMYD2 was significantly correlated with disease-free survival (DFS). We detected 15 missense, 4 truncating, 4 fusions, and 1 splice type of mutation. The expression of SMYD2 was significantly correlated with tumor purity and immunocyte infiltration in six cancer types. The gene GNPAT was highly associated with SMYD2. Significant pathways and Gene Ontology (GO) terms for co-expressed genes were associated to various processes linked with cancer formation.
    CONCLUSIONS: Collectively, our data-driven results may provide reasonably comprehensive insights for understanding the carcinogenic effect of SMYD2. It suggests that SMYD2 might be used as a significant target for identifying new biomarkers for various human tumors.
    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
    尽管吉西他滨(GEM)是晚期胰腺腺癌(PAAD)的一线药物,GEM耐药的发展严重限制了这种化疗的有效性.这项研究调查了生态型病毒整合位点2A(EVI2A)在PAAD中对GEM的抗性和免疫逃避的机制。使用GEO数据集预测GEM抗性相关生物标志物,产生GEM抗性PAAD细胞。发现EVI2A在GEM抗性PAAD细胞中高表达。功能分析显示,EVI2A促进GEM抗性细胞的增殖和运动,并阻止细胞凋亡。此外,EVI2A降低T细胞效应物活化。SMYD2在GEM抗性细胞中过表达,和SMYD2增强了EVI2A的H3K36me2改性,从而促进EVI2A表达。SMYD2降低了GEM抗性细胞的敏感性,被EVI2A击倒逆转。SMYD2增加了M2巨噬细胞的数量(与PAAD细胞共培养),并减少了T细胞效应子的激活(与巨噬细胞上清液共培养),EVI2A敲除后,M2巨噬细胞数量减少,T细胞效应子被激活。我们的研究结果表明,EVI2A,由SMYD2-H3K36me2表观遗传轴操纵,促进PAAD中GEM抵抗和M2巨噬细胞介导的免疫逃避。因此,EVI2A可能是克服PAAD中GEM耐药性和免疫抑制环境的治疗靶标。
    Although gemcitabine (GEM) is the first‑line drug for advanced pancreatic adenocarcinoma (PAAD), the development of GEM resistance severely limits the effectiveness of this chemotherapy. This study investigated the mechanisms of ecotropic viral integration site 2 A (EVI2A) for resistance to GEM and immune evasion in PAAD. GEM resistance-related biomarkers were predicted using GEO datasets, and GEM-resistant PAAD cells were generated. EVI2A was found expressed highly in GEM-resistant PAAD cells. Gain-of-function analyses revealed that EVI2A encouraged the proliferation and motility of GEM-resistant cells and prevented apoptosis. In addition, EVI2A reduced T cell effector activation. SMYD2 was overexpressed in GEM-resistant cells, and SMYD2 enhanced H3K36me2 modification of EVI2A, thereby promoting EVI2A expression. SMYD2 reduced the sensitivity of GEM-resistant cells, which was reversed by EVI2A knockdown. SMYD2 increased the amount of M2 macrophages (co-cultured with PAAD cells) and decreased T cell effector activation (co-cultured with macrophage supernatant), and the number of M2 macrophages was decreased and T cell effectors were activated following EVI2A knockdown. Our findings indicate that EVI2A, manipulated by the SMYD2-H3K36me2 epigenetic axis, promoted GEM resistance and M2 macrophage-mediated immune evasion in PAAD. Therefore, EVI2A might represent a therapeutic target for overcoming GEM resistance and immunosuppressive environment in PAAD.
    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
    基于负载药物的纳米颗粒的癌症治疗可以克服传统化疗方法中药物毒性作用的问题。在这项研究中,我们装载了LLY-507,一种有效的SMYD2抑制剂,一种甲基转移酶,在氧化铁纳米颗粒(IONP)上。通过显微分析对制备的纳米粒子进行了表征,装载效率,和药物释放研究。显微镜检查显示平均晶粒尺寸为44nm。LLY-507-IONP的体外作用,通过MTT分析(A549细胞)和溶血研究确定LLY-507和IONP。IONP在血液中具有几乎阴性的溶血活性。细胞活力测定揭示了单独的LLY-507和装载LLY-507的IONP对A549的IC50值;装载在NP上的药物的较低值(单独0.71μg/mL和装载在NP上的0.53μg/mL)显示出强的协同抗癌潜力。我们进一步测试了负载的NP在尿烷诱导的肺癌小鼠模型中的作用(在三项独立试验中,n=40只小鼠,对照组20只小鼠),以检查SMYD2在肺癌发展的各个时间点的作用。由于LLY-507导致的SMYD2的丢失抑制了肿瘤的生长,肺气肿,出血,和相当大的拥堵。因此,可以得出结论,SMYD2抑制剂对小鼠肺具有抗炎作用,并通过抑制SMYD2蛋白抑制肿瘤生长。
    Cancer therapies based on nanoparticles with a loaded drug can overcome the problem of the drug\'s toxic effects in the traditional chemotherapeutic approach. In this study, we loaded LLY-507, a potent inhibitor of SMYD2, a methyltransferase enzyme, on iron oxide nanoparticles (IONPs). The prepared nanoparticles were characterized by microscopic analysis, loading efficiency, and drug release studies. Microscopic examination revealed an average grain size of 44 nm. The in vitro effect of LLY-507-IONPs, LLY-507, and IONPs was determined by MTT analysis (A549 cells) and hemolysis studies. IONPs have almost negative hemolytic activity in blood. The cell viability assay revealed IC50 values of both LLY-507 alone and LLY-507-loaded IONPs against A549; the lower value of the drug loaded on NPs (0.71 µg/mL alone and 0.53 µg/mL loaded on NPs) shows strong synergistic anticancer potential. We further tested the role of loaded NPs in a urethane-induced lung cancer mouse model (n = 40 mice in three independent trials, 20 mice in control group) to check the role of SMYD2 at various time points of lung cancer development. The loss of SMYD2 due to LLY-507 suppressed tumor growth, emphysema, hemorrhage, and congestion considerably. Hence, it can be concluded that the SMYD2 inhibitor has an anti-inflammatory effect on the mouse lung and suppresses tumor growth by inhibiting the SMYD2 protein.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号