Tamoxifen

他莫昔芬
  • 文章类型: Journal Article
    他莫昔芬是乳腺癌(BC)内分泌治疗中广泛使用的抗雌激素药物。它通过竞争性结合雌激素受体α(ERα)来阻断雌激素信号,从而抑制BC细胞的生长。然而,随着他莫昔芬的长期应用,一部分BC患者对他莫昔芬有耐药性,这导致低总生存率和无进展生存率。耐药的分子机制主要是由于ERα表达下调和PI3K/AKT/mTOR信号通路异常激活。此外,DNA甲基化介导的靶向基因表达下调是调控蛋白质表达的重要调控方式。在本次审查中,简要介绍了甲基化和他莫昔芬,其次是甲基化对他莫昔芬耐药性和敏感性的影响。最后,描述了他莫昔芬甲基化的临床应用,包括其作为预后指标的用途。最后,假设当甲基化与他莫昔芬联合使用时,它可以恢复他莫昔芬的抗性。
    Tamoxifen is a widely used anti‑estrogen drug in the endocrine therapy of breast cancer (BC). It blocks estrogen signaling by competitively binding to estrogen receptor α (ERα), thereby inhibiting the growth of BC cells. However, with the long‑term application of tamoxifen, a subset of patients with BC have shown resistance to tamoxifen, which leads to low overall survival and progression‑free survival. The molecular mechanism of resistance is mainly due to downregulation of ERα expression and abnormal activation of the PI3K/AKT/mTOR signaling pathway. Moreover, the downregulation of targeted gene expression mediated by DNA methylation is an important regulatory mode to control protein expression. In the present review, methylation and tamoxifen are briefly introduced, followed by a focus on the effect of methylation on tamoxifen resistance and sensitivity. Finally, the clinical application of methylation for tamoxifen is described, including its use as a prognostic indicator. Finally, it is hypothesized that when methylation is used in combination with tamoxifen, it could recover the resistance of tamoxifen.
    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
    目的:作为骨形态发生蛋白(BMP)的拮抗剂,Noggin促进乳腺癌的溶骨性骨转移。本研究旨在进一步剖析其在雌激素受体(ER)阳性乳腺癌中的作用。
    方法:在雌激素剥夺和用17-β-雌二醇(E2)处理的条件下,测定ER阳性乳腺癌细胞系(MCF-7和T-47D)中的Noggin表达。使用重组人BMP7(rhBMP7)和BMP受体抑制剂(LDN-193189)检查了雌激素调节的Noggin中Smad1/5/8的激活。在MCF-7和T-47D细胞系中评价Noggin对细胞功能的影响。使用MTT测定法在具有Noggin过表达的MCF-7和T-47D细胞中确定对他莫昔芬和化疗药物的反应。
    结果:Noggin在乳腺癌中的表达与ERα呈负相关。Noggin在雌激素剥夺后上调,此外,E2消除了这种影响,在雌激素剥夺的MCF-7和T-47D细胞中观察到磷酸化Smad1/5/8水平升高,分别由E2和LDN-193189阻止。E2和LDN-193189也阻止了BMP7诱导的Noggin表达和Smad1/5/8的激活。Noggin过表达导致MCF-7和T-47D细胞的增殖增加。过表达Noggin的MCF-7和T-47D细胞对他莫昔芬(TAM)表现出良好的耐受性,DTX,5-FU,但活细胞的百分比高于对照组。
    结论:Noggin的表达可以通过BMP/Smad信号的推断而被雌激素抑制。过表达Noggin促进MCF-7和T-47D细胞增殖,有助于耐药性。
    OBJECTIVE: As an antagonist of bone morphogenetic protein (BMP), Noggin facilitates osteolytic bone metastases from breast cancer. The present study aimed to further dissect its role in oestrogen receptor (ER) positive breast cancer.
    METHODS: Noggin expression in ER positive breast cancer cell lines (MCF-7 and T-47D) was determined under conditions of oestrogen deprivation and treatment with 17-β-oestradiol (E2). Activation of Smad1/5/8 in the oestrogen-regulated Noggin was examined using recombinant human BMP7 (rhBMP7) and a BMP receptor inhibitor (LDN-193189). The influence of Noggin on cellular functions was evaluated in MCF-7 and T-47D cell lines. Responses to tamoxifen and chemotherapy drugs were determined in MCF-7 and T-47D cells with Noggin over-expression using MTT assay.
    RESULTS: Noggin expression was negatively correlated with ERα in breast cancers. Noggin was up-regulated upon oestrogen deprivation, an effect that was eliminated by E2 Furthermore, increased levels of phosphorylated Smad1/5/8 were observed in the oestrogen-deprived MCF-7 and T-47D cells, which was prevented by E2 and LDN-193189, respectively. BMP7-induced Noggin expression and activation of Smad1/5/8 was also prevented by E2 and LDN-193189. Noggin over-expression resulted in an increase in the proliferation of both MCF-7 and T-47D cells. MCF-7 and T-47D cells over-expressing Noggin exhibited a good tolerance to tamoxifen (TAM), DTX, and 5-FU, but the percentage of viable cells was higher compared with the controls.
    CONCLUSIONS: Noggin expression can be repressed by oestrogen through inference with the BMP/Smad signalling. Over-expression of Noggin promotes the proliferation of MCF-7 and T-47D cells, contributing to drug resistance.
    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
    雌激素受体阳性(ER+)乳腺癌严重危害全球女性身心健康,ER靶向治疗至关重要。这里,我们发现柑橘富含多甲氧基黄酮(PMFs)的水解物(C-H)及其主要成分(金黄素和3-甲氧基金黄素)通过泛素-蛋白酶体途径有效降解ERα蛋白,从而损害ER+乳腺癌细胞的增殖。此外,我们的研究表明,C-H联合他莫昔芬(TAM)在体外抑制ER乳腺癌的细胞增殖。进一步证实,C-H降低了荷瘤129小鼠体内ER+乳腺癌的肿瘤生长,并提高了他莫昔芬的功效。我们的研究表明,柑橘PMFs通过靶向ERα蛋白降解在乳腺癌治疗中具有潜在的药物和保健产品应用。
    Estrogen receptor-positive (ER+) breast cancer seriously endangers the women\'s physical and mental health worldwide and ER targeting therapy is vital. Here, we found that a citrus polymethoxyflavones (PMFs)-rich hydrolysate (C-H) and its major components (nobiletin and 3-methoxynobiletin) potently degrade ERα protein via the ubiquitin-proteasome pathway, thereby impairing the proliferation of ER+ breast cancer cells. Moreover, our study exhibited that C-H combined with tamoxifen (TAM) inhibited the cell proliferation of ER+ breast cancer in vitro. It was further confirmed that C-H decreased tumor growth of ER+ breast cancer in tumor-bearing 129 mice in vivo and improved the efficacy of tamoxifen. Our study revealed that the citrus PMFs have potential applications as pharmaceutical and healthcare products in breast cancer treatment by targeting ERα protein degradation.
    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
    内分泌耐药对激素受体阳性和人上皮生长因子受体2阴性(HR+HER2-)乳腺癌患者构成重大临床挑战。雌激素受体(ER)和ERBB信号通路的失调与抗性发展有关;然而,这些途径的整合仍不清楚.虽然已知SMAD4在肿瘤发生中起着不同的作用,它与内分泌抵抗的关系知之甚少。这里,我们研究了SMAD4在HR+HER2-乳腺癌获得性内分泌耐药中的作用.全基因组CRISPR筛选将SMAD4鉴定为T47D细胞中4-羟基他莫昔芬(OHT)敏感性的调节剂。临床数据分析显示乳腺癌组织中SMAD4表达下调,与预后不良有关。内分泌治疗后,SMAD4的表达被进一步抑制。功能研究表明,SMAD4消耗通过增强ER和ERBB信号传导在体外和体内诱导内分泌抗性。ER和ERBB信号的伴随抑制导致异常的自噬激活。同时抑制ER,ERBB,和自噬途径协同影响SMAD4耗竭细胞。我们的发现揭示了内分泌治疗诱导的SMAD4下调通过整合ER和ERBB信号驱动获得性耐药的机制,并为内分泌耐药HR+HER2-乳腺癌患者提出了合理的治疗策略。
    Endocrine resistance poses a significant clinical challenge for patients with hormone receptor-positive and human epithelial growth factor receptor 2-negative (HR + HER2-) breast cancer. Dysregulation of estrogen receptor (ER) and ERBB signaling pathways is implicated in resistance development; however, the integration of these pathways remains unclear. While SMAD4 is known to play diverse roles in tumorigenesis, its involvement in endocrine resistance is poorly understood. Here, we investigate the role of SMAD4 in acquired endocrine resistance in HR + HER2- breast cancer. Genome-wide CRISPR screening identifies SMAD4 as a regulator of 4-hydroxytamoxifen (OHT) sensitivity in T47D cells. Clinical data analysis reveals downregulated SMAD4 expression in breast cancer tissues, correlating with poor prognosis. Following endocrine therapy, SMAD4 expression is further suppressed. Functional studies demonstrate that SMAD4 depletion induces endocrine resistance in vitro and in vivo by enhancing ER and ERBB signaling. Concomitant inhibition of ER and ERBB signaling leads to aberrant autophagy activation. Simultaneous inhibition of ER, ERBB, and autophagy pathways synergistically impacts SMAD4-depleted cells. Our findings unveil a mechanism whereby endocrine therapy-induced SMAD4 downregulation drives acquired resistance by integrating ER and ERBB signaling and suggest a rational treatment strategy for endocrine-resistant HR + HER2- breast cancer patients.
    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:尽管他莫昔芬在治疗雌激素受体α(ERα)阳性乳腺癌方面取得了成功,随后的他莫昔芬耐药性的发展是临床上的共同挑战。催乳素受体(PRLR)下游信号可能与ERα在乳腺癌进展中协同作用。然而,针对PRL-PRLR轴联合他莫昔芬的潜在效应尚未得到彻底研究.
    方法:从TCGA获得的高通量RNA-seq数据,分析了Metabric和GEO数据集,以探索乳腺癌细胞中PRLR的表达以及PRLR表达与他莫昔芬治疗的相关性。使用外源性或PRL过表达细胞模型来研究活化的PRLR途径在介导他莫昔芬不敏感性中的作用。免疫毒素靶向PRLR(N8-PE24)是用剪接-内皮素技术构建的,N8-PE24对乳腺癌的疗效进行了体外和体内评估,包括分析细胞生长或凋亡,3D球状体培养,和动物异种移植。
    结果:PRL激活的PRLR通路可显著降低ERα阳性乳腺癌细胞对他莫昔芬的敏感性。他莫昔芬处理可上调PRLR的转录,并可通过碱化溶酶体诱导PRLR蛋白在乳腺癌细胞中的大量积累。同时,通过长期他莫昔芬压力获得的他莫昔芬抗性MCF7表现出PRLR转录和蛋白质水平的上调。免疫毒素N8-PE24在体外和体内均增强了乳腺癌细胞对他莫昔芬的敏感性。在异种移植模型中,当治疗PRLR阳性三阴性乳腺癌时,N8-PE24显著增强他莫昔芬和紫杉醇的疗效。
    结论:PRL-PRLR轴可能与ERα阳性乳腺癌细胞中他莫昔芬不敏感相关。N8-PE24可以抑制乳腺癌细胞的生长,并提高PRLR阳性乳腺癌细胞对他莫昔芬和紫杉醇的药物敏感性。我们的研究为靶向PRLR治疗乳腺癌提供了新的视角。
    BACKGROUND: Though tamoxifen achieves success in treating estrogen receptor α (ERα)-positive breast cancer, the followed development of tamoxifen resistance is a common challenge in clinic. Signals downstream of prolactin receptor (PRLR) could synergize with ERα in breast cancer progression. However, the potential effect of targeting PRL-PRLR axis combined with tamoxifen has not been thoroughly investigated.
    METHODS: High-throughput RNA-seq data obtained from TCGA, Metabric and GEO datasets were analyzed to explore PRLR expression in breast cancer cell and the association of PRLR expression with tamoxifen treatment. Exogenous or PRL overexpression cell models were employed to investigate the role of activated PRLR pathway in mediating tamoxifen insensitivity. Immunotoxin targeting PRLR (N8-PE24) was constructed with splicing-intein technique, and the efficacy of N8-PE24 against breast cancer was evaluated using in vitro and in vivo methods, including analysis of cells growth or apoptosis, 3D spheroids culture, and animal xenografts.
    RESULTS: PRLR pathway activated by PRL could significantly decrease sensitivity of ERα-positive breast cancer cells to tamoxifen. Tamoxifen treatment upregulated transcription of PRLR and could induce significant accumulation of PRLR protein in breast cancer cells by alkalizing lysosomes. Meanwhile, tamoxifen-resistant MCF7 achieved by long-term tamoxifen pressure exhibited both upregulated transcription and protein level of PRLR. Immunotoxin N8-PE24 enhanced sensitivity of breast cancer cells to tamoxifen both in vitro and in vivo. In xenograft models, N8-PE24 significantly enhanced the efficacy of tamoxifen and paclitaxel when treating PRLR-positive triple-negative breast cancer.
    CONCLUSIONS: PRL-PRLR axis potentially associates with tamoxifen insensitivity in ERα-positive breast cancer cells. N8-PE24 could inhibit cell growth of the breast cancers and promote drug sensitivity of PRLR-positive breast cancer cells to tamoxifen and paclitaxel. Our study provides a new perspective for targeting PRLR to treat 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
    背景:在ASCO指南中,他莫昔芬(TAM)被推荐为雌激素受体(ER)阳性早期乳腺癌患者的一线治疗方案。我们的研究旨在分析接受TAM辅助内分泌治疗与没有TAM相比的成本效益。并评估美国ER阳性早期男性乳腺癌患者高依从性TAM与低依从性的成本效益。
    方法:构建了两个包含三个相互排斥的健康状况的马尔可夫模型:(1)第一个马尔可夫模型比较了添加TAM与不使用TAM的成本效益(TAM与不使用TAM);(2)第二个模型比较了接受高依从性和低依从性的TAM(高依从性TAM与低依从性TAM)的成本效益。两种模型的模拟时间范围是患者的寿命。两个模型的有效性和安全性数据来自真实世界的研究。模型输入来自美国网站和已发布的文献。两个模型的主要结果都包括总成本,质量调整寿命年(QALYs),和增量成本效益比(ICER)。
    结果:在第一个模型中,与非TAM相比,TAM每QALY的ICER为5707.29美元,这大大低于美国每QALY50,000.00美元的WTP门槛。概率敏感性分析结果表明,该策略的成本效益概率为100.00%。在第二个模型中,与低依从性TAM相比,高依从性TAM绝对占主导地位。当WTP设置为$50,000.00/QALY时,高依从性TAM具有比低依从性TAM高99.70%的概率。所有这些参数在其合理范围内并没有反向改变我们模型的结果。
    结论:我们的研究将为医师或患者在进行治疗决策时提供有价值的指导,并为决策时考虑将资金适当分配给这一特殊群体提供有效的参考。
    BACKGROUND: Tamoxifen (TAM) is recommended as the first-line strategy for men with estrogen receptor (ER)-positive early breast cancer who are candidates for adjuvant endocrine therapy in ASCO guideline. Our study aims to analyze the cost-effectiveness of receiving adjuvant endocrine therapy with TAM compared to no TAM, and to assess the cost-effectiveness of using TAM with high adherence over low adherence for ER-positive early male breast cancer in the USA.
    METHODS: Two Markov models comprising three mutually exclusive health states were constructed: (1) the first Markov model compared the cost-effectiveness of adding TAM with not using TAM (TAM versus Not-TAM); (2) the second model compared the cost-effectiveness of receiving TAM with high adherence and low adherence (High-adherence-TAM versus Low-adherence-TAM). The simulation time horizon for both models was the lifetime of patients. The efficacy and safety data of two models were elicited from the real-world studies. Model inputs were derived from the US website and published literature. The main outcomes of two models both included the total cost, quality-adjusted life-years (QALYs), and incremental cost-effectiveness ratios (ICERs).
    RESULTS: In the first model, TAM yielded an ICER of $5707.29 per QALY compared to Not-TAM, which was substantially below the WTP threshold of $50,000.00 per QALY in the USA. Probabilistic sensitivity analysis results demonstrated a 100.00% probability of cost-effectiveness for this strategy. In the second model, High-adherence-TAM was dominated absolutely compared to Low-adherence-TAM. The High-adherence-TAM was cost-effective with a 99.70% probability over Low-adherence-TAM when WTP was set as $50,000.00/QALY. All of these parameters within their plausible ranges did not reversely change the results of our models.
    CONCLUSIONS: Our study will offer valuable guidance for physicians or patients when making treatment decisions and provide an effective reference for decision-making to consider the appropriate allocation of funds to this special group.
    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
    乳腺癌是全球女性癌症相关死亡的主要原因,激素受体(HR)+是主要亚型。他莫昔芬(TAM)作为HR+乳腺癌的主要治疗方法。然而,耐药性往往导致复发,强调需要开发新的疗法来提高患者的生活质量和降低复发率。青蒿素(ART)已证明在抑制耐药细胞生长方面的功效,将艺术定位为抵消内分泌抵抗的可行选择。本研究通过生物信息学分析和实验验证相结合的方法探索了青蒿素和他莫昔芬之间的相互作用。五个特征基因(AR,cdkn1a,erbb2,esr1,hsp90aa1)和七个药物-疾病交叉基因(cyp2e1,rorc,mapk10,glp1r,egfr,PGR,mgll)使用WGCNA交叉分析鉴定。随后进行功能富集分析。我们的发现证实了他莫昔芬耐药和致敏患者的关键簇基因表达与免疫细胞浸润之间的显着相关性。scRNA-seq分析显示关键簇基因在上皮细胞中高表达,提示青蒿素对雌激素受体(ER)阳性BC组织中肿瘤细胞的特异性影响。分子靶标对接和青蒿素对LCC9细胞的体外实验表明,通过调节相关耐药基因,可以逆转耐药细胞的迁移和耐药。这些结果表明青蒿素可以潜在地逆转ER阳性乳腺癌中的他莫昔芬耐药性。
    Breast cancer is the leading cause of cancer-related deaths in women worldwide, with Hormone Receptor (HR)+ being the predominant subtype. Tamoxifen (TAM) serves as the primary treatment for HR+ breast cancer. However, drug resistance often leads to recurrence, underscoring the need to develop new therapies to enhance patient quality of life and reduce recurrence rates. Artemisinin (ART) has demonstrated efficacy in inhibiting the growth of drug-resistant cells, positioning art as a viable option for counteracting endocrine resistance. This study explored the interaction between artemisinin and tamoxifen through a combined approach of bioinformatics analysis and experimental validation. Five characterized genes (ar, cdkn1a, erbb2, esr1, hsp90aa1) and seven drug-disease crossover genes (cyp2e1, rorc, mapk10, glp1r, egfr, pgr, mgll) were identified using WGCNA crossover analysis. Subsequent functional enrichment analyses were conducted. Our findings confirm a significant correlation between key cluster gene expression and immune cell infiltration in tamoxifen-resistant and -sensitized patients. scRNA-seq analysis revealed high expression of key cluster genes in epithelial cells, suggesting artemisinin\'s specific impact on tumor cells in estrogen receptor (ER)-positive BC tissues. Molecular target docking and in vitro experiments with artemisinin on LCC9 cells demonstrated a reversal effect in reducing migratory and drug resistance of drug-resistant cells by modulating relevant drug resistance genes. These results indicate that artemisinin could potentially reverse tamoxifen resistance in ER-positive 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
    目的:雌激素受体阳性(ER+)乳腺癌发生和对内分泌治疗耐药的复杂分子机制仍然难以捉摸。在这项研究中,我们阐明了G蛋白偶联受体GPR81的关键作用,在ER+乳腺癌(BC)中,通过证明GPR81在他莫昔芬(TAM)抗性ER+BC细胞系和肿瘤样品中的低表达,以及潜在的分子机制。
    方法:使用MDA和FAβO测定法探索脂肪酸氧化(FAO)水平和脂质积累,BODIPY493/503染色,和脂质TOX染色。使用CYTO-ID检测和Western印迹测定自噬水平。通过CCK8实验研究了GPR81对BC中TAM抗性的影响,集落形成测定和异种移植小鼠模型。
    结果:TAM抗性BC细胞中GPR81的异常低表达破坏了Rap1通路,导致PPARα和CPT1上调。PPARα/CPT1的这种升高增强了粮农组织,阻碍脂质积累和脂滴(LD)形成,并随后抑制细胞自噬,最终促进TAM抗性BC细胞生长。此外,靶向GPR81和粮农组织成为一种有前途的治疗策略,作为GPR81激动剂和CPT1抑制剂依托莫克在体内有效抑制ER+BC细胞和肿瘤生长,TAM抗性ER+细胞对TAM处理再敏感。
    结论:我们的数据强调了GPR81在促进ER+乳腺肿瘤发生和对内分泌治疗耐药方面的关键和功能上的重要作用。GPR81和FAO水平在TAM抗性ER+BC的临床环境中显示出作为诊断生物标志物和治疗靶标的潜力。
    OBJECTIVE: The intricate molecular mechanisms underlying estrogen receptor-positive (ER+) breast carcinogenesis and resistance to endocrine therapy remain elusive. In this study, we elucidate the pivotal role of GPR81, a G protein-coupled receptor, in ER+ breast cancer (BC) by demonstrating low expression of GPR81 in tamoxifen (TAM)-resistant ER+ BC cell lines and tumor samples, along with the underlying molecular mechanisms.
    METHODS: Fatty acid oxidation (FAO) levels and lipid accumulation were explored using MDA and FAβO assay, BODIPY 493/503 staining, and Lipid TOX staining. Autophagy levels were assayed using CYTO-ID detection and Western blotting. The impact of GPR81 on TAM resistance in BC was investigated through CCK8 assay, colony formation assay and a xenograft mice model.
    RESULTS: Aberrantly low GPR81 expression in TAM-resistant BC cells disrupts the Rap1 pathway, leading to the upregulation of PPARα and CPT1. This elevation in PPARα/CPT1 enhances FAO, impedes lipid accumulation and lipid droplet (LD) formation, and subsequently inhibits cell autophagy, ultimately promoting TAM-resistant BC cell growth. Moreover, targeting GPR81 and FAO emerges as a promising therapeutic strategy, as the GPR81 agonist and the CPT1 inhibitor etomoxir effectively inhibit ER+ BC cell and tumor growth in vivo, re-sensitizing TAM-resistant ER+ cells to TAM treatment.
    CONCLUSIONS: Our data highlight the critical and functionally significant role of GPR81 in promoting ER+ breast tumorigenesis and resistance to endocrine therapy. GPR81 and FAO levels show potential as diagnostic biomarkers and therapeutic targets in clinical settings for TAM-resistant ER+ BC.
    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
    他莫昔芬(TAM)是一种经典的抗雌激素药物,通过竞争性结合雌激素受体α(ERα)来拮抗雌激素。然而,对TAM的耐药性仍然是乳腺癌治疗中的重大挑战。在这项研究中,我们旨在设计一种主动靶向给药系统来增强TAM对ER阳性乳腺癌细胞的增殖抑制作用。在这里,用染料木素(GEN)修饰壳聚糖(CS),获得主动靶向的GEN-CS。使用离子交联方法构建了负载TAM的纳米颗粒(TAM-GEN-CS-NP),以GEN-CS为载体材料,三磷酸钠(TPP)为交联剂。因此,TAM-GEN-CS-NP表现出平均尺寸为299.8nm的球形形态。包封率和载药量分别为85.77%和14.13µg/mg,分别。与免费TAM相比,TAM-GEN-CS-NP表现出明显的缓释性能。体外细胞实验表明,TAM-GEN-CS-NP对MCF-7细胞具有积极的靶向和增殖抑制作用。TAM和TAM-GEN-CS-NP的IC50分别为10.25µg/mL和7.22µg/mL,分别。更重要的是,TAM和GEN的组合指数(CI)值小于1,表明具有协同作用。因此,TAM-GEN-CS-NP具有通过主动靶向和协同治疗策略增强TAM治疗乳腺癌的潜力。
    Tamoxifen (TAM) is a classical anti-estrogenic drug that antagonizes estrogen by competitively binding to estrogen receptor α (ERα). However, drug resistance to TAM remains a significant challenge in breast cancer treatment. In this study, we aimed to design an actively targeted drug delivery system to enhance the proliferation inhibitory effects of TAM on ER positive breast cancer cells. Herein, chitosan (CS) was modified with genistein (GEN) to obtain the actively targeted GEN-CS. The TAM-loaded nanoparticles (TAM-GEN-CS-NPs) were constructed using an ionic-crosslinking method, with GEN-CS as the carrier material and sodium tripolyphosphate (TPP) as the crosslinking agent. As a result, TAM-GEN-CS-NPs exhibited a spherical morphology with an average size of 299.8 nm. The encapsulation efficiency and drug loading content were 85.77% and 14.13 µg/mg, respectively. Compared with free TAM, TAM-GEN-CS-NPs displayed obvious slow-release performance. In vitro cellular assays demonstrated that TAM-GEN-CS-NPs had active targeting and proliferation inhibitory effects on MCF-7 cells. The IC50 of TAM and TAM-GEN-CS-NPs were 10.25 µg/mL and 7.22 µg/mL, respectively. More importantly, the combination index (CI) value of TAM and GEN was less than 1, indicating synergistic effects. Therefore, TAM-GEN-CS-NPs hold the potential to enhance TAM therapy for breast cancer through active targeting and synergistic treatment strategies.
    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
    他莫昔芬是一种雌激素受体调节剂,据报道可缓解小鼠肝脏脂质积聚,但机制尚不清楚。过氧化物酶体脂肪酸β-氧化是长链脂肪酸超负荷的主要代谢途径。由于长链脂肪酸是肝脏脂质积累的原因,过氧化物酶体脂肪酸β-氧化的激活可能是代谢相关脂肪肝疾病的一种新的治疗策略.在这项研究中,我们基于过氧化物酶体脂肪酸β-氧化的激活,研究了他莫昔芬抗肝脏脂质积累的机制。在无性别差异的高脂饮食小鼠中,他莫昔芬降低肝脏长链脂肪酸并减轻肝脏脂质积累。体外,他莫昔芬保护原代肝细胞免受棕榈酸诱导的脂毒性。机械上,从肝脏分离的肝细胞的RNA序列显示过氧化物酶体脂肪酸β-氧化被他莫昔芬激活。烯酰辅酶A水合酶和3-羟基酰基辅酶A水合酶的蛋白和mRNA表达在体内和体外均显着增加。原代肝细胞中的小干扰RNA烯酰CoA水合酶和3-羟酰基CoA水合酶消除了他莫昔芬在脂质积累中的治疗作用。总之,我们的结果表明,他莫昔芬可以通过激活烯酰辅酶A水合酶和3-羟酰基辅酶A水合酶介导的过氧化物酶体脂肪酸β-氧化来减轻高脂饮食小鼠的肝脏脂质积累.
    Tamoxifen is an estrogen receptor modulator that has been reported to alleviate hepatic lipid accumulation in mice, but the mechanism is still unclear. Peroxisome fatty acid β-oxidation is the main metabolic pathway for the overload of long-chain fatty acids. As long-chain fatty acids are a cause of hepatic lipid accumulation, the activation of peroxisome fatty acid β-oxidation might be a novel therapeutic strategy for metabolic associated fatty liver disease. In this study, we investigated the mechanism of tamoxifen against hepatic lipid accumulation based on the activation of peroxisome fatty acid β-oxidation. Tamoxifen reduced liver long-chain fatty acids and relieved hepatic lipid accumulation in high fat diet mice without sex difference. In vitro, tamoxifen protected primary hepatocytes against palmitic acid-induced lipotoxicity. Mechanistically, the RNA-sequence of hepatocytes isolated from the liver revealed that peroxisome fatty acid β-oxidation was activated by tamoxifen. Protein and mRNA expression of enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase were significantly increased in vivo and in vitro. Small interfering RNA enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase in primary hepatocytes abolished the therapeutic effects of tamoxifen in lipid accumulation. In conclusion, our results indicated that tamoxifen could relieve hepatic lipid accumulation in high fat diet mice based on the activation of enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase-mediated peroxisome fatty acids β-oxidation.
    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