LY294002

LY294002
  • 文章类型: Journal Article
    背景:现有研究表明,阻断磷酸肌醇3-激酶(PI3K)活性有助于过敏性哮喘的炎症溶液,但这种抑制作用是否直接减弱体内嗜中性粒细胞气道炎症仍不清楚。我们探索了PI3K特异性抑制剂LY294002对中性粒细胞性气道炎症进展的药理作用,并研究了其潜在机制。
    结果:雌性C57BL/6小鼠在第0天和第6天用卵清蛋白(OVA)和脂多糖(LPS)鼻内致敏,并在第14-17天用OVA攻击以建立中性粒细胞气道疾病模型。在挑战阶段,一部分小鼠用LY294002气管内治疗.我们发现LY294002的治疗可减轻炎性小鼠的临床症状。组织学研究显示LY294002显著抑制炎症细胞浸润和粘液产生。该治疗还显著抑制OVA-LPS诱导的炎症细胞计数增加,尤其是中性粒细胞计数,支气管肺泡灌洗液(BALF)中IL-17水平。与未处理的小鼠相比,LY294002处理的小鼠在BALF中表现出显著增加的IL-10水平。此外,LY294002降低IL-6和IL-17的血浆浓度。LY29402的抗炎作用与NLRP3炎性体的下调相关。
    结论:我们的研究结果表明LY294002是中性粒细胞气道炎症的潜在药理学靶点。
    BACKGROUND: Existing investigations suggest that the blockade of phosphoinositide 3-kinase (PI3K) activity contributes to inflammatory solution in allergic asthma, but whether this inhibition directly attenuates neutrophilic airway inflammation in vivo is still unclear. We explored the pharmacological effects of LY294002, a specific inhibitor of PI3K on the progression of neutrophilic airway inflammation and investigated the underlying mechanism.
    RESULTS: Female C57BL/6 mice were intranasally sensitized with ovalbumin (OVA) together with lipopolysaccharide (LPS) on days 0 and 6, and challenged with OVA on days 14-17 to establish a neutrophilic airway disease model. In the challenge phase, a subset of mice was treated intratracheally with LY294002. We found that treatment of LY294002 attenuates clinic symptoms of inflammatory mice. Histological studies showed that LY294002 significantly inhibited inflammatory cell infiltration and mucus production. The treatment also significantly inhibited OVA-LPS induced increases in inflammatory cell counts, especially neutrophil counts, and IL-17 levels in bronchoalveolar lavage fluid (BALF). LY294002 treated mice exhibited significantly increased IL-10 levels in BALF compared to the untreated mice. In addition, LY294002 reduced the plasma concentrations of IL-6 and IL-17. The anti-inflammatory effects of LY29402 were correlated with the downregulation of NLRP3 inflammasome.
    CONCLUSIONS: Our findings suggested that LY294002 as a potential pharmacological target for neutrophilic airway inflammation.
    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:结直肠癌(CRC)的转移是阻碍其治疗效果并导致高死亡率的重要障碍之一。结直肠癌患者手术切除率低,预后差。PSAT1是参与丝氨酸生物合成的酶。研究表明,PSAT1在调节肿瘤转移中起着至关重要的作用。上皮-间充质转化(EMT)是上皮细胞获得间充质表型的细胞重编程过程。它是促进细胞转移和恶性肿瘤进展的关键过程。大肠癌中PSAT1与EMT的关系,以及潜在的分子机制,仍然是神秘的,值得彻底探索。这些发现表明,PSAT1可能是减轻结直肠癌转移的有希望的治疗靶标,并且在即将进行的研究中具有作为有价值的预后生物标志物的潜力。材料和方法:利用TCGA数据集结合临床CRC标本,我们最初的重点是深入检查CRC中PSAT1的表达,特别探索其与患者经历的不良预后结局的潜在相关性。此外,我们通过利用siRNA敲低技术对PSAT1对CRC的调控影响进行了全面调查.在体外实验领域,我们仔细评估了PSAT1对CRC进展各个方面的影响,包括细胞迁移,入侵,扩散,和殖民地的形成。为了阐明复杂的影响,我们采用了包含一系列检测和分析的多方面方法.这些包括伤口愈合试验,transwell分析,利用细胞计数试剂盒-8(CCK-8)测定,和集落形成测定。通过使用这种多样化的调查技术,我们能够全面理解PSAT1在结直肠癌发病机制中的多方面作用.这种多种多样的分析极大地促进了我们对结直肠癌发病机制中的复杂机制的深入理解。使用WB和PCR实验,我们发现PSAT1在调节CRC的EMT发展中发挥作用。在机制方面,我们发现PSAT1通过调节Pl3K/AKT信号通路影响EMT。结果:我们的研究揭示了CRC标本中PSAT1表达的显著下调。重要的是,这种下调与CRC患者的不良预后呈显著正相关.功能上,我们的研究表明,在体外环境中,siRNA介导的PSAT1敲低可显著增强CRC发病机制的各个关键方面.具体来说,这包括大幅促进CRC细胞迁移,入侵,扩散,和殖民地的形成。此外,PSAT1的沉默也证明了EMT过程的实质性促进。有趣的是,我们的研究揭示了迄今为止尚未探索的PSAT1在CRC和EMT中的调控作用机制.我们已经建立,第一次,PSAT1通过调节PI3K/AKT信号通路的激活发挥其影响。这种机制见解为理解CRC进展和PSAT1介导的EMT诱导的分子基础提供了有价值的贡献。结论:一致地,我们的研究结果揭示了以前未知的PSAT1/PI3K/AKT轴在CRCEMT过程启动中的重要作用.此外,我们的发现引入了一种新的生物标志物,对CRC的临床诊断和治疗具有潜在意义.
    Background: The metastasis of colorectal cancer (CRC) is one of the significant barriers impeding its treated consequence and bring about high mortality, less surgical resection rate and poor prognosis of CRC patients. PSAT1 is an enzyme involved in serine biosynthesis. The studies showed that PSAT1 plays the part of a crucial character in the regulation of tumor metastasis. And Epithelial-Mesenchymal Transition (EMT) is a process of cell reprogramming in which epithelialcells obtain mesenchymal phenotypes. It is a crucial course in promoting cell metastasis and the progression of malignant tumors. The relationship between PSAT1 and EMT in colorectal cancer, as well as the underlying molecular mechanisms, remains enigmatic and warrants thorough exploration. These findings suggest that PSAT1 may serve as a promising therapeutic target for mitigating colorectal cancer metastasis and holds the potential to emerge as a valuable prognostic biomarker in forthcoming research endeavors. Materials and Methods: Utilizing TCGA dataset in conjunction with clinical CRC specimens, our initial focus was directed towards an in-depth examination of PSAT1 expression within CRC, specifically exploring its potential correlation with the adverse prognostic outcomes experienced by patients. Furthermore, we conducted a comprehensive investigation into the regulatory influence exerted by PSAT1 on CRC through the utilization of siRNA knockdown techniques. In the realm of in vitro experimentation, we meticulously evaluated the impact of PSAT1 on various facets of CRC progression, including cell migration, invasion, proliferation, and colony formation. In order to elucidate the intricate effects in question, we adopted a multifaceted methodology that encompassed a range of assays and analyses. These included wound healing assays, transwell assays, utilization of the Cell Counting Kit-8 (CCK-8) assay, and colony formation assays. By employing this diverse array of investigative techniques, we were able to achieve a comprehensive comprehension of the multifaceted role that PSAT1 plays in the pathogenesis of colorectal cancer. This multifarious analysis greatly contributed to our in-depth understanding of the complex mechanisms at play in colorectal cancer pathogenesis. Using WB and PCR experiments, we found that PSAT1 has a role in regulating EMT development in CRC.In terms of mechanism, we found that PSAT1 affected EMT by Regulating Pl3K/AKT Signaling Pathway. Results: Our investigation revealed a noteworthy down-regulation of PSAT1 expression in CRC specimens. Importantly, this down-regulation exhibited a significant positive correlation with the unfavorable prognosis of patients afflicted with CRC. Functionally, our study showcased that the siRNA-mediated knockdown of PSAT1 markedly enhanced various key aspects of CRC pathogenesis in an in vitro setting. Specifically, this included a substantial promotion of CRC cell migration, invasion, proliferation, and colony formation. Moreover, the silencing of PSAT1 also demonstrated a substantial promotion of the EMT process. Intriguingly, our research unveiled a hitherto unexplored mechanism underlying the regulatory role of PSAT1 in CRC and EMT. We have established, for the first time, that PSAT1 exerts its influence by modulating the activation of the PI3K/AKT Signaling Pathway. This mechanistic insight provides a valuable contribution to the understanding of the molecular underpinnings of CRC progression and EMT induction mediated by PSAT1. Conclusions: In unison, our research findings shed light on the previously uncharted and significant role of the PSAT1/PI3K/AKT axis in the initiation of the EMT process in CRC. Furthermore, our discoveries introduce a novel biomarker with potential implications for the clinical diagnosis and treatment of CRC.
    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
    目的:本研究旨在阐明间充质干细胞(MSCs)如何通过PI3K-AKT通路调节线粒体动力学,有效减轻来曲唑诱导的多囊卵巢综合征(PCOS)的病理变化。
    方法:32只雌性大鼠随机分为4个实验组,PCOS,PCOS+MSCs,和PCOS+MSCs+LY294002。Sham组接受0.5%w/v羧甲基纤维素(CMC);PCOS组接受来曲唑(1mg/kg,每天)在0.5%CMC中21天。PCOS+MSC组中的动物接受1×106个MSC/大鼠(i.p,)在研究的第22天。在PCOS+MSCs+LY294002组中,大鼠在MSC移植前40分钟接受LY294002(PI3K-AKT抑制剂)。线粒体动态基因表达,线粒体膜电位(MMP),柠檬酸合酶(CS)活性,氧化应激,炎症,卵巢组织学参数,血清激素水平,胰岛素抵抗的稳态模型评估(HOMA-IR),胰岛素和葡萄糖浓度,在实验结束时评估p-PI3K和p-AKT蛋白水平。
    结果:PCOS大鼠显示线粒体动力学和组织学改变的显著破坏,降低MMP,CS,卵巢超氧化物歧化酶(SOD)和雌激素水平。他们的胰岛素和葡萄糖浓度也有显著上升,HOMA-IR,睾酮水平,肿瘤坏死因子-α(TNF-α)和白细胞介素-6(IL-6)水平,与Sham组相比,卵巢丙二醛(MDA)含量以及p-PI3K和p-AKT蛋白水平显着降低。在PCOS+MSCs组中,MSCs移植可以改善上述参数。施用LY294002(PI3K-AKT通路抑制剂)使线粒体动态标志物恶化,氧化应激状态,炎症标志物,荷尔蒙水平,葡萄糖,与PCOS+MSCs组相比,胰岛素水平和卵泡发育。
    结论:这项研究表明,MSC移植在调节线粒体动力学方面具有保护作用,促进线粒体生物发生,在PCOS模型中,与氧化还原状态和炎症反应的竞争主要通过PI3K-AKT通路介导.
    OBJECTIVE: The present study aimed to elucidate how mesenchymal stem cells (MSCs) application could efficiently attenuate pathological changes of letrozole-induced poly cystic ovary syndrome (PCOS) by modulating mitochondrial dynamic via PI3K-AKT pathway.
    METHODS: Thirty-two female rats were randomly divided into four experimental groups: Sham, PCOS, PCOS + MSCs, and PCOS + MSCs + LY294002. The Sham group received 0.5% w/v carboxymethyl cellulose (CMC); the PCOS group received letrozole (1 mg/kg, daily) in 0.5% CMC for 21 days. Animals in the PCOS + MSCs group received 1 × 106 MSCs/rat (i.p,) on the 22th day of the study. In the PCOS + MSCs + LY294002 group, rats received LY294002 (PI3K-AKT inhibitor) 40 min before MSC transplantation. Mitochondrial dynamic gene expression, mitochondrial membrane potential (MMP), citrate synthase (CS) activity, oxidative stress, inflammation, ovarian histological parameters, serum hormone levels, homeostatic model assessment for insulin resistance (HOMA-IR), insulin and glucose concentrations, p-PI3K and p-AKT protein levels were evaluated at the end of the experiment.
    RESULTS: PCOS rats showed a significant disruption of mitochondrial dynamics and histological changes, lower MMP, CS, ovary super oxide dismutase (SOD) and estrogen level. They also had a notable rise in insulin and glucose concentrations, HOMA-IR, testosterone level, tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) levels, ovarian malondialdehyde (MDA) content as well as a notable decrease in p-PI3K and p-AKT protein levels compared to the Sham group. In the PCOS + MSCs group, the transplantation of MSCs could improve the above parameters. Administration of LY294002 (PI3K-AKT pathway inhibitor) deteriorated mitochondrial dynamic markers, oxidative stress status, inflammation markers, hormonal levels, glucose, and insulin levels and follicular development compared to the PCOS + MSCs group.
    CONCLUSIONS: This study demonstrated that the protective effects of MSC transplantation in regulating mitochondrial dynamics, promoting mitochondrial biogenesis, competing with redox status and inflammation response were mainly mediated through the PI3K-AKT pathway in the PCOS model.
    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
    目的:自身免疫性甲状腺炎(AIT)是最常见的甲状腺疾病;没有预防疾病进展的措施.本研究试图确定Notch信号传导通过激活下游磷脂酰肌醇-3激酶/蛋白激酶/雷帕霉素复合物1的机制靶标(PI3K/AKT/mTORC1)途径来调节T辅助性17(Th17)细胞的分化。实验性自身免疫性甲状腺炎(EAT)的甲状腺损伤。
    方法:体内实验,小鼠随机分为4组:对照组,EAT组,和两组用LY294002治疗(pTg分别加25mg/kg或50mg/kgLY294002)。评估了甲状腺炎的程度,和Th17细胞的百分比,白细胞介素-17A(IL-17A)的表达,并对Notch-PI3K信号通路的主要成分进行检测。体外实验,使用两种不同剂量的LY294002(25和50μM)干预EAT小鼠的脾单核细胞(SMC),以进一步评估Notch-PI3K通路对Th17细胞的调节作用。
    结果:我们的数据表明Th17细胞的浸润和IL-17A的表达,缺口,多毛和分裂1(Hes1),p-AKT(Ser473),p-AKT(Thr308),p‑mTOR(Ser2448),在EAT小鼠中S6K1和S6K2显著增加。PI3K通路被阻断后,甲状腺炎的程度明显减轻,Th17细胞的比例,IL-17A的表达,上述Notch-PI3K通路相关分子呈剂量依赖性下降。此外,Th17细胞比例与血清甲状腺球蛋白抗体(TgAb)浓度呈正相关,IL-17A,和Notch-PI3K通路相关分子的mRNA水平。
    结论:Notch信号通过Hes-磷酸酶和张力蛋白同源物(PTEN)调节下游PI3K/AKT/mTORC1通路促进Th17细胞分泌IL-17A,参与甲状腺自身免疫损伤,PI3K通路抑制剂可能通过影响Th17细胞分化对AIT发挥重要作用。
    OBJECTIVE: Autoimmune Thyroiditis (AIT) is the most common thyroid disease; however, there were no measures to prevent the progression of the disease. The present study attempts to identify that Notch signaling regulates the differentiation of T helper 17 (Th17) cells by activating downstream Phosphatidylinositol-3 kinase/protein kinase/mechanistic target of rapamycin complex 1 (PI3K/AKT/mTORC1) pathway participating in the thyroid injury of the experimental autoimmune thyroiditis (EAT).
    METHODS: In vivo experiments, mice were randomly divided into 4 groups: a control group, an EAT group, and two groups with LY294002 treatment (pTg plus 25 mg/kg or 50 mg/kg LY294002, respectively). The degrees of thyroiditis were evaluated, and the percentage of Th17 cells, expression of interleukin-17A (IL-17A), and the main components of the Notch-PI3K signaling pathway were detected in different groups. In vitro experiments, two different dosages of LY294002 (25 and 50 μM) were used to intervene splenic mononuclear cells (SMCs) from EAT mice to further evaluate the regulatory effect of Notch-PI3K pathway on Th17 cells.
    RESULTS: Our data demonstrate that the infiltration of Th17 cells and the expressions of IL-17A, Notch, hairy and split 1 (Hes1), p‑AKT (Ser473), p‑AKT (Thr308), p‑mTOR (Ser2448), S6K1, and S6K2 increased remarkably in EAT mice. After PI3K pathway was blocked, the degrees of thyroiditis were significantly alleviated, and the proportion of Th17 cells, the expression of IL-17A, and the above Notch-PI3K pathway-related molecules decreased in a dose-dependent manner. Additionally, the proportion of Th17 cells was positively correlated with the concentration of serum thyroglobulin antibody (TgAb), IL-17A, and Notch-PI3K pathway-related molecules mRNA levels.
    CONCLUSIONS: Notch signal promotes the secretion of IL-17A from Th17 cells by regulating the downstream PI3K/AKT/mTORC1 pathway through Hes-Phosphatase and tensin homolog (PTEN) and participates in thyroid autoimmune damage, and the PI3K pathway inhibitor may play important effects on AIT by affecting Th17 cells differentiation.
    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
    呋喃香豆素是植物界中天然存在的化合物,其特点是低分子量,化学结构简单,在大多数有机溶剂中溶解度高。此外,它们具有广泛的活性,它们的性质取决于所连接的取代基的位置和类型。因此,我们研究的目的是研究呋喃香豆素(欧前胡素,异欧前胡素,Bergapten,和黄毒素)与人类多形性胶质母细胞瘤(T98G)和间变性星形细胞瘤(MOGGCCM)细胞系有关。测试化合物首次与LY294002(PI3K抑制剂)和索拉非尼(Raf抑制剂)组合使用。细胞凋亡,自噬,用Hoechst33342,吖啶橙,和碘化丙啶,分别。caspase3和Beclin1的水平通过免疫印迹和Raf和PI3K激酶的阻断来估计,使用特异性siRNA转染。划痕试验用于评估神经胶质瘤细胞的迁移潜力。我们的研究表明,呋喃香豆素的抗癌活性严格取决于存在,type,和取代基的位置。所获得的结果表明,通过在香豆素骨架的C8位置上存在异戊二烯基部分来确定实现更高的促凋亡活性。在间变性星形细胞瘤和胶质母细胞瘤中,欧前胡素诱导细胞凋亡最有效。此外,欧周素的用法,单独使用以及与索拉非尼或LY294002联合使用,可降低MOGGCCM和T98G细胞的迁移潜力。
    Furanocoumarins are naturally occurring compounds in the plant world, characterized by low molecular weight, simple chemical structure, and high solubility in most organic solvents. Additionally, they have a broad spectrum of activity, and their properties depend on the location and type of attached substituents. Therefore, the aim of our study was to investigate the anticancer activity of furanocoumarins (imperatorin, isoimperatorin, bergapten, and xanthotoxin) in relation to human glioblastoma multiforme (T98G) and anaplastic astrocytoma (MOGGCCM) cell lines. The tested compounds were used for the first time in combination with LY294002 (PI3K inhibitor) and sorafenib (Raf inhibitor). Apoptosis, autophagy, and necrosis were identified microscopically after straining with Hoechst 33342, acridine orange, and propidium iodide, respectively. The levels of caspase 3 and Beclin 1 were estimated by immunoblotting and for the blocking of Raf and PI3K kinases, the transfection with specific siRNA was used. The scratch test was used to assess the migration potential of glioma cells. Our studies showed that the anticancer activity of furanocoumarins strictly depended on the presence, type, and location of substituents. The obtained results suggest that achieving higher pro-apoptotic activity is determined by the presence of an isoprenyl moiety at the C8 position of the coumarin skeleton. In both anaplastic astrocytoma and glioblastoma, imperatorin was the most effective in induction apoptosis. Furthermore, the usage of imperatorin, alone and in combination with sorafenib or LY294002, decreased the migratory potential of MOGGCCM and T98G 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
    背景:最初在Th1细胞表面发现的T细胞免疫球蛋白和含粘蛋白结构域的分子3(TIM-3),负调节免疫反应并介导Th1细胞凋亡。越来越多的研究表明,TIM-3在免疫疾病的发生和发展中至关重要,癌症,和慢性传染病。然而,TIM-3对子宫内膜异位症的影响尚不清楚.
    方法:定量实时聚合酶链反应,西方印迹,和免疫组织化学用于测量子宫内膜异位症的TIM-3水平。细胞计数试剂盒-8,5-乙炔基-2'-脱氧尿苷,菌落形成,Transwell®迁移,Matrigel®入侵,和流式细胞术测定用于探索TIM-3的体外功能,和裸鼠异种移植实验用于评估其在体内的作用。根据RNAseq,筛选脑源性神经营养因子(BDNF)。通过转染质粒并在体内和体外添加抑制剂来确定特异性增殖相关信号分子的参与。
    结果:TIM-3mRNA和蛋白在在位和异位子宫内膜组织中的表达水平明显高于正常子宫内膜组织。通过检测TIM-3过表达和敲低对细胞增殖的影响,迁移,和体外侵袭,和体内病变的形成,我们发现TIM-3的表达与体外细胞增殖和克隆形成呈正相关,以及裸鼠的病变生长。通过添加磷脂酰肌醇3激酶/蛋白激酶B(PI3K/AKT)通路抑制剂LY294002,我们进一步验证了TIM-3通过PI3K途径促进体内和体外增殖。通过将质粒转染到ESC细胞中,并给予子宫内膜异位大鼠模型抑制剂,我们测试了TIM-3通过BDNF介导的PI3K/AKT轴调节增殖。
    结论:TIM-3可通过体内外BDNF介导的PI3K/AKT轴促进子宫内膜异位症的增殖,可能为子宫内膜异位症的治疗提供新的治疗靶点。
    BACKGROUND: T cell immunoglobulin and mucin domain-containing molecule-3 (TIM-3) initially discovered on the surface of Th1 cells, negatively regulates immune responses and mediates apoptosis of Th1 cells. An increasing number of studies have since shown that TIM-3 is crucial in the genesis and development of immune diseases, cancers, and chronic infectious illnesses. However, the effect of TIM-3 on endometriosis is still unknown.
    METHODS: Quantitative real-time polymerase chain reaction, western blotting, and immunohistochemistry were used to measure TIM-3 levels in endometriosis. Cell Counting Kit-8, 5-ethynyl-2\'-deoxyuridine, colony-forming, Transwell® migration, Matrigel® invasion, and flow cytometry assays were used to explore the function of TIM-3 in vitro, and xenograft experiments in nude mice were used to assess its role in vivo. According to the RNA seq, brain-derived neurotrophic factor (BDNF) was screened. The involvement of specific proliferation-related signaling molecules was determined by transfecting a plasmid and adding an inhibitor in vivo and in vitro.
    RESULTS: TIM-3 mRNA and protein expression levels were significantly higher in eutopic and ectopic endometrial tissues than in normal endometrial tissues. By examining the effects of TIM-3 overexpression and knockdown on cell proliferation, migration, and invasion in vitro, and lesions formation in vivo, we found that the expression of TIM-3 was positively correlated with cell proliferation and clone formation in vitro, as well as lesions growth in nude mice. By adding the phosphatidylinositol 3 kinase/protein kinase B(PI3K/AKT) pathway inhibitor LY294002 and knocking down PI3K, we further verified that TIM-3 promotes proliferation in vivo and in vitro via the PI3K pathway. By transfecting the plasmid into ESC cells and gave inhibitors to endometriotic rats models, we tested that TIM-3 regulates the proliferation by BDNF-mediated PI3K/AKT axis.
    CONCLUSIONS: TIM-3 can promote the proliferation of endometriosis by BDNF-mediated PI3K/AKT axis in vivo and in vitro, which may provide a new therapeutic target for the treatment of endometriosis.
    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
    骨髓增生异常综合征(MDS)向急性髓细胞性白血病(AML)的转化提出了重大的临床挑战。H3在赖氨酸27(H3K27me3)甲基化酶和去甲基化酶途径上的三甲基化参与MDS进程的调节。本研究调查了MEK/ERK和PI3K/AKT通路在MDS-AML转化中的功能机制。首先建立MDS-AML小鼠和SKM-1细胞模型,然后用MEK/ERK通路抑制剂治疗,PI3K/AKT通路抑制剂U0126,Ly294002或其组合。H3K27me3甲基化酶,使用蛋白质印迹分析确定了zeste同源物(EZH)1,EZH2,含去甲基酶Jumonji结构域的蛋白-3(JMJD3)和X染色体上普遍转录的四肽重复序列(UTX)和H3K27me3蛋白水平的增强子。细胞活力,周期分布和增殖使用CCK-8,流式细胞术,EdU和集落形成测定。测定临床样本和建立的模型中的ERK和AKT磷酸化水平,并对SKM-1细胞行为进行了评估。测量了H3K27me3甲基化酶和去甲基化酶以及无远端同源异型盒5(DLX5)的水平。结果显示,MDS和MDS-AML患者的ERK和AKT磷酸化水平升高,和老鼠模型。用MEK/ERK途径抑制剂U0126治疗,和PI3K/AKT途径抑制剂Ly294002,有效抑制MDS-AML小鼠的ERK和AKT磷酸化。观察到用U0126/Ly294002处理的MDS小鼠表现出降低的向AML的转化,延缓疾病转化,提高存活率。用U0126/Ly294002处理SKM‑1细胞导致细胞活力和增殖下降,并通过抑制ERK/PI3K磷酸化来增加细胞周期停滞。此外,用U0126/Ly294002治疗下调EZH2/EZH1表达,并上调JMJD3/UTX表达。当EZH2/EZH1过表达或JMJD3/UTX在SKM‑1细胞中被抑制时,U0126/Ly294002的作用无效。用U0126/Ly294002处理还导致DLX5启动子区域中的H3K27me3蛋白水平和H3K27me3水平降低,导致DLX5表达增加。总的来说,本研究的结果表明,U0126/Ly294002通过调节H3K27me3甲基化酶和去甲基化酶的水平参与MDS-AML转化,并调控DLX5的转录和表达。
    The transformation of myelodysplastic syndrome (MDS) into acute myeloid leukemia (AML) poses a significant clinical challenge. The trimethylation of H3 on lysine 27 (H3K27me3) methylase and de‑methylase pathway is involved in the regulation of MDS progression. The present study investigated the functional mechanisms of the MEK/ERK and PI3K/AKT pathways in the MDS‑to‑AML transformation. MDS‑AML mouse and SKM‑1 cell models were first established and this was followed by treatment with the MEK/ERK pathway inhibitor, U0126, the PI3K/AKT pathway inhibitor, Ly294002, or their combination. H3K27me3 methylase, enhancer of zeste homolog (EZH)1, EZH2, demethylase Jumonji domain‑containing protein‑3 (JMJD3) and ubiquitously transcribed tetratricopeptide repeat on chromosome X (UTX) and H3K27me3 protein levels were determined using western blot analysis. Cell viability, cycle distribution and proliferation were assessed using CCK‑8, flow cytometry, EdU and colony formation assays. The ERK and AKT phosphorylation levels in clinical samples and established models were determined, and SKM‑1 cell behaviors were assessed. The levels of H3K27me3 methylases and de‑methylases and distal‑less homeobox 5 (DLX5) were measured. The results revealed that the ERK and AKT phosphorylation levels were elevated in patients with MDS and MDS‑AML, and in mouse models. Treatment with U0126, a MEK/ERK pathway inhibitor, and Ly294002, a PI3K/AKT pathway inhibitor, effectively suppressed ERK and AKT phosphorylation in mice with MDS‑AML. It was observed that mice with MDS treated with U0126/Ly294002 exhibited reduced transformation to AML, delayed disease transformation and increased survival rates. Treatment of the SKM‑1 cells with U0126/Ly294002 led to a decrease in cell viability and proliferation, and to an increase in cell cycle arrest by suppressing ERK/PI3K phosphorylation. Moreover, treatment with U0126/Ly294002 downregulated EZH2/EZH1 expression, and upregulated JMJD3/UTX expression. The effects of U0126/Ly294002 were nullified when EZH2/EZH1 was overexpressed or when JMJD3/UTX was inhibited in the SKM‑1 cells. Treatment with U0126/Ly294002 also resulted in a decreased H3K27me3 protein level and H3K27me3 level in the DLX5 promoter region, leading to an increased DLX5 expression. Overall, the findings of the present study suggest that U0126/Ly294002 participates in MDS‑AML transformation by modulating the levels of H3K27me3 methylases and de‑methylases, and regulating DLX5 transcription and expression.
    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
    血管生成拟态(VM),侵袭性癌细胞形成管状结构的过程,在提供营养和逃生路线方面起着至关重要的作用。高可塑性肿瘤细胞,例如具有三阴性乳腺癌(TNBC)表型的患者,可以开发VM。然而,关于肿瘤微环境的细胞成分与TNBC细胞的VM能力之间的相互作用知之甚少。在这项研究中,我们分析了内皮细胞和基质细胞与TNBC细胞相互作用时诱导VM的能力,并分析了FGFR/PI3K/Akt通路在这一过程中的参与.使用荧光标记的TNBC细胞证实VM。只有内皮细胞触发了VM的形成,提示来自内皮来源的旁分泌/近分泌因子在VM发育中的主要作用。通过免疫细胞化学,qPCR,和分泌组分析,我们确定了VM形成癌细胞中促血管生成因子和干细胞标志物的表达增加.同样,由TNBC细胞引发的内皮细胞显示促血管生成分子的上调,包括FGF,VEGFA,和几种炎症细胞因子。AZD4547或LY294002阻止了内皮依赖性TNBC-VM的形成,这强烈表明FGFR/PI3K/Akt轴参与了该过程。鉴于VM与不良临床预后相关,靶向FGFR/PI3K/Akt的药理学可能有望治疗和预防TNBC肿瘤中的VM.
    Vasculogenic mimicry (VM), a process in which aggressive cancer cells form tube-like structures, plays a crucial role in providing nutrients and escape routes. Highly plastic tumor cells, such as those with the triple-negative breast cancer (TNBC) phenotype, can develop VM. However, little is known about the interplay between the cellular components of the tumor microenvironment and TNBC cells\' VM capacity. In this study, we analyzed the ability of endothelial and stromal cells to induce VM when interacting with TNBC cells and analyzed the involvement of the FGFR/PI3K/Akt pathway in this process. VM was corroborated using fluorescently labeled TNBC cells. Only endothelial cells triggered VM formation, suggesting a predominant role of paracrine/juxtacrine factors from an endothelial origin in VM development. Via immunocytochemistry, qPCR, and secretome analyses, we determined an increased expression of proangiogenic factors as well as stemness markers in VM-forming cancer cells. Similarly, endothelial cells primed by TNBC cells showed an upregulation of proangiogenic molecules, including FGF, VEGFA, and several inflammatory cytokines. Endothelium-dependent TNBC-VM formation was prevented by AZD4547 or LY294002, strongly suggesting the involvement of the FGFR/PI3K/Akt axis in this process. Given that VM is associated with poor clinical prognosis, targeting FGFR/PI3K/Akt pharmacologically may hold promise for treating and preventing VM in TNBC 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
    背景:卵泡发生是体细胞和卵母细胞之间相互作用的细胞信号的复杂网络。卵泡液(FF)中的许多成分在卵泡发生过程中动态变化,并在卵母细胞成熟中起积极作用。以前的研究报道,溶血磷脂酸(LPA)促进卵丘细胞的扩张,卵母细胞核成熟,和卵母细胞的体外成熟。
    结果:最初,LPA的表达在成熟FF中显著升高(P<0.0001)。然后,在人颗粒细胞(KGNs)中处理10μMLPA24小时会加重细胞增殖,随着自噬的增加,减少细胞凋亡。同时,我们证明LPA通过PI3K-AKT-mTOR信号通路介导的细胞功能作为PI3K抑制剂(LY294002)显著阻止LPA诱导的AKT,mTOR磷酸化和自噬激活。这种结果也通过免疫荧光染色和流式细胞术得到证实。此外,自噬抑制剂3甲基腺嘌呤(3MA)也可以减轻LPA的作用,通过PI3K-AKT-mTOR途径激活细胞凋亡。最后,我们发现用Ki16425或敲低LPAR1阻断,减轻LPA介导的KGN中的自噬激活,提示LPA通过激活LPAR1和PI3K-AKT-mTOR信号通路增强自噬。
    结论:本研究表明,在颗粒细胞中,LPA通过LPAR1激活PI3K-Akt-mTOR通路的增加,通过增强自噬抑制细胞凋亡,这可能在体内卵母细胞成熟中起作用。
    BACKGROUND: Folliculogenesis is a complex network of interacting cellular signals between somatic cells and oocytes. Many components in ovarian follicular fluid (FF) dynamically change during folliculogenesis and play a positive role in oocyte maturation. Previous studies have reported that lysophosphatidic acid (LPA) promotes cumulus cell expansion, oocyte nuclear maturation, and in vitro maturation of oocytes.
    RESULTS: Initially, the expression of LPA was raised in matured FF significantly (P < 0.0001). Then, 10 μM LPA treated for 24 h in human granulosa cells (KGNs) aggravated cell proliferation, with increased autophagy, and reduced apoptosis. Meanwhile, we demonstrated that LPA mediated cell function through the PI3K-AKT-mTOR signaling pathway as PI3K inhibitor (LY294002) significantly prevented LPA-induced AKT, mTOR phosphorylation and autophagy activation. Such results were also verified by immunofluorescence staining and flow cytometry. In addition, an autophagy inhibitor 3 methyladenine (3MA) could also alleviate the effects of LPA, by activating apoptosis through PI3K-AKT-mTOR pathways. Finally, we found blockade with Ki16425 or knockdown LPAR1, alleviated LPA mediated autophagy activation in KGNs, suggesting that LPA enhances autophagy through activation of the LPAR1 and PI3K-AKT-mTOR signaling pathways.
    CONCLUSIONS: This study demonstrates that increased LPA activated PI3K-Akt-mTOR pathway through LPAR1 in granulosa cells, suppressing apoptosis by enhancing autophagy, which might play a role in oocyte maturation in vivo.
    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
    Flash纳米沉淀(FNP)是一种湍流混合过程,能够重复地生产载有活性药物成分(API)的聚合物纳米颗粒。用这种方法生产的纳米颗粒由被亲水电晕包围的疏水核组成。FNP产生具有非常高加载水平的非离子疏水API的纳米颗粒。然而,具有可电离基团的疏水性化合物不能有效地掺入。为了克服这一点,可以将离子配对剂(IP)掺入到FNP制剂中以产生在混合期间有效沉淀的高度疏水性药物盐。我们证明了PI3K抑制剂的封装,LY294002,在聚(乙二醇)-b-聚(D,L乳酸)纳米颗粒。我们研究了在FNP过程中掺入两个疏水IP(棕榈酸(PA)和十六烷基膦酸(HDPA))如何影响LY294002负载和所得纳米颗粒的尺寸。还检查了有机溶剂选择对合成过程的影响。虽然任一疏水IP的存在有效地增加了LY294002在FNP过程中的包封,HDPA产生明确定义的胶体稳定颗粒,而PA导致不明确的聚集体。疏水性IP与FNP的结合为API的静脉内施用打开了大门,所述API先前由于其疏水性而被认为是不可用的。
    Flash nanoprecipitation (FNP) is a turbulent mixing process capable of reproducibly producing polymer nanoparticles loaded with active pharmaceutical ingredients (APIs). The nanoparticles produced with this method consist of a hydrophobic core surrounded by a hydrophilic corona. FNP produces nanoparticles with very high loading levels of nonionic hydrophobic APIs. However, hydrophobic compounds with ionizable groups are not as efficiently incorporated. To overcome this, ion pairing agents (IPs) can be incorporated into the FNP formulation to produce highly hydrophobic drug salts that efficiently precipitate during mixing. We demonstrate the encapsulation of the PI3K inhibitor, LY294002, within poly(ethylene glycol)-b-poly(D,L lactic acid) nanoparticles. We investigated how incorporating two hydrophobic IPs (palmitic acid (PA) and hexadecylphosphonic acid (HDPA)) during the FNP process affected the LY294002 loading and size of the resulting nanoparticles. The effect of organic solvent choice on the synthesis process was also examined. While the presence of either hydrophobic IP effectively increased the encapsulation of LY294002 during FNP, HDPA resulted in well-defined colloidally stable particles, while the PA resulted in ill-defined aggregates. The incorporation of hydrophobic IPs with FNP opens the door for the intravenous administration of APIs that were previously deemed unusable due to their hydrophobic nature.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号