MDR1

mdr1
  • 文章类型: Journal Article
    背景:吉西他滨是治疗胰腺癌所有阶段的基石药物,可以延长胰腺癌患者的生存期,但是胰腺癌患者对吉西他滨的耐药性阻碍了其疗效。尚未探索胰腺导管腺癌中早期生长反应1(EGR1)的过表达作为胰腺癌吉西他滨化学耐药的机制。吉西他滨化疗耐药的主要机制与药物摄取有关。新陈代谢,和行动。肿瘤多药耐药(MDR)在癌细胞中化疗的常见原因之一是转运蛋白通过诱导抗凋亡机制增加细胞内药物流出并降低药物浓度。据报道,吉西他滨以高亲和力结合MDR1。这项研究的目的是研究EGR1与MDR1联合调节胰腺癌细胞吉西他滨耐药性的潜在机制。
    方法:本研究使用以下体外和体内技术来探索EGR1与MDR1结合以调节胰腺癌细胞吉西他滨耐药性的潜在机制。细胞培养;通过功能分析的EGR1功能的体外和体内研究。使用ChIP测定法检测EGR1与MDR1启动子的结合。qRT-PCR,Westernblot法检测蛋白和mRNA表达;运用AnnexinV凋亡检测法检测细胞凋亡;CCK8、Edu法检测细胞增殖活力。建立胰腺癌皮下移植瘤动物模型,用苏木素伊红染色,免疫组化检测Ki-67表达。
    结果:我们发现,与正常胰腺导管上皮细胞相比,EGR1在不同胰腺癌细胞系中的表达增加。此外,吉西他滨治疗以剂量和时间依赖性方式诱导EGR1表达上调。EGR1在MDR1启动子序列中显著富集。敲低EGR1后,CFPAC-1和PANC-1细胞系中的细胞增殖受损,细胞凋亡增强,MDR1表达降低,从而部分逆转吉西他滨化学耐药。在动物实验中,与sh-NC组相比,EGR1敲除增强了吉西他滨对肿瘤生长的抑制作用。
    结论:我们的研究表明,EGR1可能参与MDR1的调节,以增强胰腺癌细胞的吉西他滨耐药性。EGR1可能是克服胰腺癌吉西他滨耐药的新治疗靶点。
    BACKGROUND: Gemcitabine is a cornerstone drug for the treatment of all stages of pancreatic cancer and can prolong the survival of patients with pancreatic cancer, but resistance to gemcitabine in pancreatic cancer patients hinders its efficacy. The overexpression of Early growth response 1(EGR1) in pancreatic ductal adenocarcinoma as a mechanism of gemcitabine chemoresistance in pancreatic cancer has not been explored. The major mechanisms of gemcitabine chemoresistance are related to drug uptake, metabolism, and action. One of the common causes of tumor multidrug resistance (MDR) to chemotherapy in cancer cells is that transporter proteins increase intracellular drug efflux and decrease drug concentrations by inducing anti-apoptotic mechanisms. It has been reported that gemcitabine binds to MDR1 with high affinity. The purpose of this research was to investigate the potential mechanisms by which EGR1 associates with MDR1 to regulate gemcitabine resistance in pancreatic cancer cells.
    METHODS: The following in vitro and in vivo techniques were used in this research to explore the potential mechanisms by which EGR1 binds to MDR1 to regulate gemcitabine resistance in pancreatic cancer cells. Cell culture; in vitro and in vivo study of EGR1 function by loss of function analysis. Binding of EGR1 to the MDR1 promoter was detected using the ChIP assay. qRT-PCR, Western blot assays to detect protein and mRNA expression; use of Annexin V apoptosis detection assay to test apoptosis; CCK8, Edu assay to test cell proliferation viability. The animal model of pancreatic cancer subcutaneous allograft was constructed and the tumours were stained with hematoxylin eosin and Ki-67 expression was detected using immunohistochemistry.
    RESULTS: We revealed that EGR1 expression was increased in different pancreatic cancer cell lines compared to normal pancreatic ductal epithelial cells. Moreover, gemcitabine treatment induced upregulation of EGR1 expression in a dose- and time-dependent manner. EGR1 is significantly enriched in the MDR1 promoter sequence.Upon knockdown of EGR1, cell proliferation was impaired in CFPAC-1 and PANC-1 cell lines, apoptosis was enhanced and MDR1 expression was decreased, thereby partially reversing gemcitabine chemoresistance. In animal experiments, knockdown of EGR1 enhanced the inhibitory effect of gemcitabine on tumor growth compared with the sh-NC group.
    CONCLUSIONS: Our study suggests that EGR1 may be involved in the regulation of MDR1 to enhance gemcitabine resistance in pancreatic cancer cells. EGR1 could be a novel therapeutic target to overcome gemcitabine resistance in pancreatic 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
    多药耐药(MDR)是乳腺癌化疗的主要限制。MDR的共同机制是各种抗癌药物可通过细胞膜蛋白P-糖蛋白(P-gp)外溢。这里,我们发现在耐药乳腺癌细胞中特异性检测到Shc3的异位过表达,因此,通过介导P-gp表达降低对化疗的敏感性并促进细胞迁移。然而,乳腺癌中P-gp和Shc3相互作用的分子机制尚不清楚.我们报道了另一种抗性机制,涉及Shc3上调后P-gp活性形式的增加。MCF-7/ADR和SK-BR-3细胞在敲除Shc3后可对阿霉素敏感。我们的结果表明ErbB2和EphA2之间的相互作用是间接的,并且受到Shc3的调节,这种复合物对于激活MAPK和AKT途径至关重要。同时,Shc3促进ErbB2核易位,随后通过ErbB2与COX2启动子结合来增加COX2表达。我们进一步证明,COX2表达与P-gp表达呈正相关,并且Shc3/ErbB2/COX2轴在体内上调P-gp活性。我们的结果显示了Shc3和ErbB2在调节乳腺癌细胞P-gp功效中的关键作用,并表明Shc3抑制可能增强对靶向癌基因成瘾途径的化疗药物的敏感性。
    Multidrug resistance (MDR) is a primary limitation of breast cancer chemotherapy. The common mechanism of MDR is various anticancer drugs can be effluxed by the cell membrane protein P-glycoprotein (P-gp). Here, we found that ectopic overexpression of Shc3 was detected specifically in drug-resistant breast cancer cells, consequently reducing sensitivity to chemotherapy and promoting cell migration by mediating P-gp expression. However, the molecular mechanism underlying the interplay between P-gp and Shc3 in breast cancer is unknown. We reported an additional resistance mechanism involving an increase in the active form of P-gp after Shc3 upregulation. MCF-7/ADR and SK-BR-3 cells can be sensitive to doxorubicin after knockdown of Shc3. Our results indicated that the interaction between ErbB2 and EphA2 is indirect and regulated by Shc3, and also, this complex is essential for activation of the MAPK and AKT pathways. Meanwhile, Shc3 promotes ErbB2 nuclear translocation, followed by a subsequent increase of the COX2 expression through ErbB2 binding to the COX2 promoter. We further demonstrated that COX2 expression was positively correlated with P-gp expression and the Shc3/ErbB2/COX2 axis upregulates P-gp activity in vivo. Our results show the crucial roles of Shc3 and ErbB2 in modulating P-gp efficacy in breast cancer cells and suggest that Shc3 inhibition may enhance the sensitivity to chemotherapeutic drugs that target oncogene addiction pathways.
    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:探讨飞燕子在肝癌治疗中的作用及其调控机制。
    方法:用不同浓度的飞跃素处理HepG2和HuH-7细胞。通过3-(4,5-二甲基-噻唑-2-基)-2,5-二苯基四唑溴化物测定法分析细胞活力。通过LC3b-绿色荧光蛋白(GFP)-Adv和LC3b-GFP-单体红色荧光蛋白-Adv转染的HepG2和HuH-7细胞分析细胞自噬和自噬通量,分别。通过Hoechst33342染色分析细胞凋亡,末端脱氧核苷酸转移酶dUTP缺口末端标记染色和DNA阶梯。细胞自噬,免疫印迹法检测细胞凋亡和存活相关蛋白的表达。
    结果:用不同浓度的delphinidin处理后,细胞存活率明显降低。Delphinidin可以阻断自噬通量,导致自噬体的显着增加,并导致细胞凋亡增加。菊酯与顺铂联合应用可促进抗肿瘤作用,降低顺铂在肿瘤细胞中的剂量。进一步的机制研究表明,delphinidin可以抑制肿瘤细胞中多药耐药基因1(MDR1)和促肿瘤转录辅因子DEAD-box解旋酶17(DDX17)的表达。DDX17的过表达可以逆转肿瘤细胞中delphinidin的抗肿瘤功能。
    结论:Delphinidin通过抑制MDR1和DDX17的表达诱导肿瘤细胞自噬流阻断和凋亡,具有很强的抗肿瘤作用。
    OBJECTIVE: To investigate the function and regulatory mechanisms of delphinidin in the treatment of hepatocellular carcinoma.
    METHODS: HepG2 and HuH-7 cells were treated with different concentrations of delphinidin. Cell viability was analysed by 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. The cell autophagy and autophagic flux were analysed by LC3b-green fluorescent protein (GFP)-Adv and LC3b-GFP-monomeric red fluorescent protein-Adv transfected HepG2 and HuH-7 cells, respectively. Cell apoptosis was analysed by Hoechst33342 staining, terminal deoxynucleotidyl transferase dUTP nick end labeling staining and DNA laddering. Cell autophagy, apoptosis and survival related protein expressions were detected by Western blotting.
    RESULTS: After treatment with different concentrations of delphinidin, the cell survival rate was significantly decreased. Delphinidin could block the autophagic flux, resulting in a significant increase in autophagosomes, and led to an increase in cell apoptosis. The combined application of delphinidin and cisplatin could promote the antitumour effect and reduce the dose of cisplatin in tumour cells. Further mechanism studies reveal that delphinidin could inhibit the multidrug resistance gene 1 (MDR1) and the tumour-promoting transcription cofactor DEAD-box helicase 17 (DDX17) expression in tumour cells. Overexpression of DDX17 could reverse delphinidin\'s antitumor function in tumour cells.
    CONCLUSIONS: Delphinidin has a strong anti-tumour effect by inducing tumour cell autophagic flux blockage and apoptosis by inhibiting of both MDR1 and DDX17 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
    Wistar和SpragueDawley是用于药物研究的最常见的大鼠品系,在临床前药物开发中可互换使用。没有研究评估Wistar和SpragueDawley大鼠在影响口服药物吸收的胃肠道因素方面是否相同,特别是与肠道转运蛋白有关。酶联免疫吸附测定(ELISA)和液相色谱-串联质谱(LC-MS/MS)是定量肠道蛋白质水平的两种可靠方法,具有各自独特的优势和局限性。在这项研究中,P-糖蛋白(P-gp),一个关键的外排运输车,使用ELISA和LC-MS/MS沿着雄性和雌性Wistar和SpragueDawley大鼠的整个肠道定量。这项工作表明SpragueDawley大鼠的基线P-gp表达高于Wistar大鼠。在空肠中发现P-gp表达的显着性别差异,使用这两种技术,雄性和雌性Wistar大鼠之间的回肠和结肠,男性表现出更高的P-gp水平。通过ELISA和LC-MS/MS,SpragueDawley大鼠P-gp表达无性别差异。两种方法对P-gp定量的趋势相似,但ELISA可以提供更快的数据采集。我们的发现报告了菌株之间的显着性别差异,并强调Wistar和SpragueDawley大鼠的P-gp表达不相等。由于人类在肠道P-gp水平上表现出明显的性别差异,因此,Wistar大鼠可能是更合适的临床前动物品系,以模拟雄性和雌性受试者中P-gp底物的口服药物吸收。
    Wistar and Sprague Dawley are the most common strains of rat used in pharmaceutical research and are used interchangeably in pre-clinical drug development. No studies have assessed whether Wistar and Sprague Dawley rats are equivalent in the gastrointestinal factors that influence oral drug absorption, specifically in relation to intestinal transporters. Enzyme-linked immunosorbent assay (ELISA) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) are two reliable methods for quantifying intestinal protein levels with their own distinct advantages and limitations. In this study, P-glycoprotein (P-gp), a key efflux transporter, was quantified using ELISA and LC-MS/MS along the complete intestinal tract of male and female Wistar and Sprague Dawley rats. This work presents that Sprague Dawley rats have innately higher baseline P-gp expression than Wistar rats. Significant sex differences in P-gp expression were identified in the jejunum, ileum and colon between male and female Wistar rats using both techniques, with males exhibiting higher P-gp levels. Sprague Dawley rats showed no sex differences in P-gp expression through ELISA and LC-MS/MS. Both methods demonstrated similar trends for P-gp quantification, but ELISA could offer faster data acquisition. Our findings report significant sex differences between the strains and highlight that Wistar and Sprague Dawley rats are not equivalent in their P-gp expression. As humans exhibit distinct sex differences in intestinal P-gp levels, Wistar rats may therefore be a more suitable pre-clinical animal strain to model oral drug absorption of P-gp substrates in male and female subjects.
    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
    间变性甲状腺癌(ATC)代表未分化,侵袭性和高转移性甲状腺癌的高死亡率。GAB1通过与激酶PI3K和磷酸酶SHP2的直接相互作用,紧密参与致癌信号的激活;然而,GAB1在ATC中的作用尚不清楚.GAB1在ATC中显著增加,伴有AKT激活。细胞增殖,ATC细胞中的GAB1敲低或PTC细胞中的过表达使迁移和侵袭受损或增强。此外,在ATC细胞中GAB1敲低抑制和在PTC细胞中的过表达促进了裸鼠甲状腺癌的生长。破坏GAB1和PI3K之间相互作用的GAB1突变未能恢复GAB1敲低ATC细胞中的细胞迁移和侵袭。RNA测序数据显示GAB1敲低部分重编程基因表达在ATC细胞回到正常甲状腺细胞。MDR1受AKT介导的GAB1转录调控。MDR1在ATC细胞中上调,MDR1敲低在ATC细胞中降低迁徙和侵袭。此外,MDR1过表达可恢复GAB1敲低ATC细胞的细胞迁移和侵袭以及肺转移。总的来说,GAB1在ATC中上调,通过调控MDR1表达促进AKT活化和细胞迁移侵袭。
    Anaplastic thyroid carcinoma (ATC) represents an undifferentiated, aggressive and highly metastatic form of thyroid cancer with high mortality. GAB1, through direct interaction with the kinase PI3K and phosphatase SHP2, is tightly involved in the activation of oncogenic signals; however, the role of GAB1 in ATC remains unclear. GAB1 was significantly increased in ATC, accompanied with AKT activation. Cell proliferation, migration and invasion were impaired or enhanced by GAB1 knockdown in ATC cells or overexpression in PTC cells. Moreover, GAB1 knockdown in ATC cells inhibited and overexpression in PTC cells promoted the growth of thyroid cancer in nude mice. GAB1 mutation disrupting the interaction between GAB1 and PI3K failed to restore cell migration and invasion in GAB1-knockdown ATC cells. RNA sequencing data showed GAB1-knockdown partially reprogramed gene expression in ATC cells back to that in normal thyroid cells. MDR1 was transcriptionally regulated by GAB1, which was mediated by AKT. MDR1 was upregulated in ATC cells and MDR1 knockdown in ATC cells decreased migration and invasion. In addition, MDR1 overexpression restored cell migration and invasion and lung metastasis of GAB1-knockdown ATC cells. Collectively, GAB1 is upregulated in ATC to promote AKT activation and cellular migration and invasion through regulating MDR1 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:顺铂(DDP)耐药性仍然是改善卵巢癌(OC)患者临床预后的关键挑战。Gli2过表达可导致OC细胞的DDP耐药,但具体的潜在监管机制仍不清楚。膜转运蛋白编码基因MDR1正调节各种癌症类型的化疗抗性。我们评估了MDR1作为潜在的Gli2下游靶标以及Gli2/MDR1轴在促进OC细胞DDP抗性中的贡献。
    方法:为了产生耐药的SKOV3/DDP细胞,SKOV3细胞在连续诱导下生长6个月,其中DDP浓度稳定增加。使用蛋白质印迹检测具有不同DDP敏感性的OC细胞中的Gli2表达。细胞计数试剂盒-8测定用于评估SKOV3,SKOV3/DDP的DDP敏感性,A2780和A2780/DDP细胞和逆转SKOV3/DDP和A2780/DDP细胞中的DDP抗性。使用5-乙炔基-2'-脱氧尿苷(EdU)掺入测定法分析细胞增殖。使用荧光素酶报告基因测定确定Gli2对MDR1的转录调节。最后,异种移植OC肿瘤在裸鼠中产生,然后用腹膜内DDP或磷酸盐缓冲盐水(PBS)注射处理,以研究Gli2是否影响体内OC中的DDP抗性。
    结果:DDP耐药的SKOV3/DDP和A2780/DDP细胞与DDP敏感的OC细胞相比,Gli2和MDR1的表达更高。SKOV3/DDP细胞中Gli2敲低显著降低MDR1表达,虽然它增加了DNA损伤,从而使OC细胞对DDP敏感。在SKOV3/DDP和A2780/DDP细胞中用Gli拮抗剂61抑制剂(GANT61)靶向Gli2表达后获得类似结果。在稳定过表达Gli2的细胞中,用梯度浓度的维拉帕米处理,MDR1抑制剂,显著抑制MDR1表达。我们的发现表明,MDR1表达的下调可能会逆转OC细胞对DDP的抗性。此外,双荧光素酶报告基因测定证实,MDR1是Gli2的直接下游靶标,Gli2正调控MDR1的表达。最后,裸鼠皮下异种移植证明Gli2在调节OC耐药中起关键作用。
    结论:我们确定了Hedgehog-Gli信号通过调节MDR1表达调节OC化学抗性的机制。因此,Gli2和MDR1是化疗耐药型OC患者的潜在生物标志物和治疗靶点。
    BACKGROUND: Cisplatin (DDP) resistance remains a key challenge in improving the clinical outcome of patients with ovarian cancer (OC). Gli2 overexpression can lead to DDP resistance in OC cells, but the specific underlying regulatory mechanism remains unclear. The membrane transporter encoding gene MDR1 positively regulates chemotherapy resistance in various cancer types. We evaluated MDR1 as a potential Gli2 downstream target and the contribution of the Gli2/MDR1 axis in promoting DDP resistance in OC cells.
    METHODS: To generate drug-resistant SKOV3/DDP cells, SKOV3 cells were grown for six months under continuous induction wherein the DDP concentration was steadily increased. Gli2 expression in OC cells with varying DDP sensitivities was detected using western blot. Cell counting kit-8 assays were used to assess the DDP sensitivity of SKOV3, SKOV3/DDP, A2780, and A2780/DDP cells and reversal of DDP resistance in SKOV3/DDP and A2780/DDP cells. Cell proliferation was analyzed using 5-ethynyl-2\'-deoxyuridine (EdU) incorporation assays. The transcriptional regulation of MDR1 by Gli2 was determined using luciferase reporter assays. Finally, xenograft OC tumors were generated in nude mice, which were then treated with intraperitoneal DDP or phosphate-buffered saline (PBS) injections to investigate if Gli2 affected DDP resistance in OC in vivo.
    RESULTS: DDP-resistant SKOV3/DDP and A2780/DDP cells showed higher expression of Gli2 and MDR1 as compared with that in DDP-sensitive OC cells. Gli2 knockdown in SKOV3/DDP cells significantly reduced MDR1 expression, whereas it increased DNA damage, thereby sensitizing OC cells to DDP. Similar results were obtained after targeting Gli2 expression with the Gli-antagonist 61 inhibitor (GANT61) in SKOV3/DDP and A2780/DDP cells. In cells stably overexpressing Gli2, treatment with gradient concentrations of verapamil, an MDR1 inhibitor, significantly inhibited MDR1 expression. Our findings indicate that downregulation of MDR1 expression may reverse OC cell resistance to DDP. Moreover, dual-luciferase reporter gene assays confirmed that MDR1 is a direct downstream target of Gli2, with Gli2 positively regulating MDR1 expression. Finally, subcutaneous xenotransplantation in nude mice demonstrated that Gli2 plays a key role in regulating OC drug resistance.
    CONCLUSIONS: We identified a mechanism by which Hedgehog-Gli signaling regulates OC chemoresistance by modulating MDR1 expression. Hence, Gli2 and MDR1 are potential biomarkers and therapeutic targets in patients with chemoresistant OC.
    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
    帕尔马霉素P3(PP3)降低氟康唑诱导的MDR1转录以逆转临床念珠菌菌株的唑耐药性。这里,我们证明,对于转录因子Mrr1具有功能获得突变的白色念珠菌菌株,PP3可恢复对几种抗真菌药物的敏感性.此外,PP3抑制携带过度活跃的MRR1等位基因的白色念珠菌菌株对Mdr1底物的外排。分子对接显示PP3是一种潜在的Mdr1阻断剂,可与Mdr1的底物结合袋结合。
    Palmarumycin P3 (PP3) reduces fluconazole-induced MDR1 transcription to reverse azole resistance in clinical Candida strains. Here, we demonstrated that PP3 restores the susceptibility to several antifungal drugs for Candida albicans strains with gain-of-function mutations in the transcription factor Mrr1. In addition, PP3 inhibits the efflux of Mdr1 substrates by C. albicans strains harboring hyperactive MRR1 alleles. Molecular docking revealed that PP3 is a potential Mdr1 blocker that binds to the substrate binding pocket of Mdr1.
    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
    肿瘤多药耐药(MDR)是干细胞样癌细胞中存在的,具有高度增殖和自我更新的干性。程序性细胞死亡4(PDCD4),作为促凋亡基因,它是否参与癌症的干性和顺铂耐药尚不清楚。在这里,我们发现Hela/DDP(顺铂耐药)细胞中的PDCD4表达低于亲本Hela细胞。此外,在Hela/DDP细胞中,耐药基因和典型的干性标志物水平显着升高。在体内,异种移植肿瘤试验证实PDCD4的敲低加速了移植肿瘤的生长。体外,集落形成和MTT实验表明,在有或没有顺铂的条件下,PDCD4过表达都会抑制细胞增殖。相比之下,PDCD4缺乏引起细胞增殖和顺铂耐药。在机制上,PDCD4降低了pAKT和pYB1的蛋白水平,伴随着MDR1表达的降低。相应地,荧光素酶报告基因分析显示PDCD4调控的MDR1启动子活性完全依赖于YB1。此外,Ch-IP,GST-下拉,和Co-IP分析提供了新的证据,表明PDCD4可以通过核仁定位信号(NOLS)片段直接与YB1结合,通过阻断YB1核易位导致YB1与MDR1启动子区的结合减少,触发MDR1转录降低。一起来看,PDCD4-pAKT-pYB1形成整合的分子网络,在干性相关顺铂耐药过程中调节MDR1转录。
    Cancer multidrug resistance (MDR) is existence in stem cell-like cancer cells characterized by stemness including high-proliferation and self-renewal. Programmed cell death 4 (PDCD4), as a proapoptotic gene, whether it engaged in cancer stemness and cisplatin resistance is still unknown. Here we showed that PDCD4 expressions in Hela/DDP (cisplatin resistance) cells were lower than in parental Hela cells. Moreover, the levels of drug resistance genes and typical stemness markers were markedly elevated in Hela/DDP cells. In vivo, xenograft tumor assay confirmed that knockdown of PDCD4 accelerated the grafted tumor growth. In vitro, colony formation and MTT assay demonstrated that PDCD4 overexpression inhibited cells proliferation in conditions with or without cisplatin. By contrast, PDCD4 deficiency provoked cell proliferation and cisplatin resistance. On mechanism, PDCD4 decreased the protein levels of pAKT and pYB1, accompanied by reduced MDR1 expression. Correspondingly, luciferase reporter assay showed PDCD4 regulated MDR1 promoter activity entirely relied on YB1. Furthermore, Ch-IP, GST-pulldown, and Co-IP assays provided novel evidence that PDCD4 could directly bind with YB1 by the nucleolar localization signal (NOLS) segment, causing the reduced YB1 binding into the MDR1 promoter region through blocking YB1 nucleus translocation, triggering the decreased MDR1 transcription. Taken together, PDCD4-pAKT-pYB1 forms the integrated molecular network to regulate MDR1 transcription during the process of stemness-associated cisplatin resistance.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Intestinal efflux transporters affect the gastrointestinal processing of many drugs but further data on their intestinal expression levels are required. Relative mRNA expression and relative and absolute protein expression data of transporters are commonly measured by real-time polymerase chain reaction (RT-PCR), Western blot and mass spectrometry-based targeted proteomics techniques. All of these methods, however, have their own strengths and limitations, and therefore, validation for optimized quantification methods is needed. As such, the identification of the most appropriate technique is necessary to effectively translate preclinical findings to first-in-human trials. In this study, the mRNA expression and protein levels of the efflux transporter P-glycoprotein (P-gp) in jejunal and ileal epithelia of 30 male and female human subjects, and the duodenal, jejunal, ileal and colonic tissues in 48 Wistar rats were quantified using RT-PCR, Western blot and liquid chromatography-tandem mass spectrometry (LC-MS/MS). A similar sex difference was observed in the expression of small intestinal P-gp in humans and Wistar rats where P-gp was higher in males than females with an increasing trend from the proximal to the distal parts in both species. A strong positive linear correlation was determined between the Western blot data and LC-MS/MS data in the small intestine of humans (R2 = 0.85). Conflicting results, however, were shown in rat small intestinal and colonic P-gp expression between the techniques (R2 = 0.29 and 0.05, respectively). In RT-PCR and Western blot, an internal reference protein is experimentally required; here, beta-actin was used which is innately variable along the intestinal tract. Quantification via LC-MS/MS can provide data on P-gp expression without the need for an internal reference protein and consequently, can give higher confidence on the expression levels of P-gp along the intestinal tract. Overall, these findings highlight similar trends between the species and suggest that the Wistar rat is an appropriate preclinical animal model to predict the oral drug absorption of P-gp substrates in the human small intestine.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Chemotherapy is the main therapy for gastric cancer (GC) both before and after surgery, but the emergence of multidrug resistance (MDR) often leads to disease progression and recurrence. P-glycoprotein, encoded by MDR1, is a well-known multidrug efflux transporter involved in drug resistance development. Pygo2 overexpression has been identified in several cancers. Previous studies have shown that abnormal expression of Pygo2 is related to tumorigenesis, chemoresistance, and tumor progression. In this study, to evaluate the underlying relationship between Pygo2 and MDR1 in GC, we constructed GC drug-resistant cell lines, SGC7901/cis-platinum (DDP), and collected tissue from GC patients\' pre-and post-chemotherapy. We found that Pygo2 was overexpressed in GC, especially in GC drug-resistant cell lines and GC patients who underwent neoadjuvant DDP-based chemotherapy. Pygo2 overexpression may precede MDR1 and correlates with MDR1 in GC patients. Furthermore, knock-down of Pygo2 induced downregulation of MDR1 and restored SGC7901/DDP\'s sensitivity to DDP. Further mechanistic analysis demonstrated that Pygo2 could modulate MDR1 transcription by binding to the MDR1 promoter region and promoting MDR1 activation. The overall findings reveal that Pygo2 may be a promising biomarker for monitoring drug resistance in GC by regulating MDR1.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号