Xenograft Model Antitumor Assays

异种移植模型抗肿瘤试验
  • 文章类型: Journal Article
    三阴性乳腺癌(TNBC)细胞中的高基底自噬和增强的线粒体裂变支持细胞迁移并促进癌细胞代谢的可塑性。这里,我们提出了一种靶向Drp1介导的线粒体裂变和自噬通路的TNBC治疗新的联合治疗方法.硫化氢(H2S)介导了无数的生物过程,包括自噬和线粒体功能。在这项研究中,我们证明了5-(4-羟基苯基)-3H-1,2-二硫醇-3-硫酮(ADT-OH),最广泛使用的持续释放H2S供体之一,在体外和体内没有增殖抑制的情况下有效抑制TNBC细胞的转移。ADT-OH治疗通过抑制自噬体形成来改善自噬通量,并通过降低动力蛋白相关蛋白1(Drp1)的表达和增加线粒体融合蛋白(Mfn2)的表达来诱导线粒体延伸。同时,ADT-OH下调线粒体自噬通量并抑制线粒体功能,最终导致抑制TNBC细胞的迁移和侵袭。在体内,ADT-OH的腹膜内给药在三种不同的动物模型中显示出有效的抗转移活性,MDA-MB-231原位异种移植模型,4T1-Luci原位模型和4T1-Luci尾静脉转移模型。然而,ADT-OH具有极低的水溶性,这是其有效性的一个重大障碍。因此,我们证明了用羟丙基-β-环糊精(CD)吸收可以显着提高ADT-OH在水中的溶解度。值得注意的是,获得的CD-ADT-OH在体内表现出优于ADT-OH的抗癌作用。总之,这项研究描述了哺乳动物线粒体裂变和自噬的新型调节因子,具有作为转移性TNBC的实验治疗剂的潜在用途。
    High basal autophagy and enhanced mitochondrial fission in triple-negative breast cancer (TNBC) cells support cell migration and promote plasticity of cancer cell metabolism. Here, we suggest a novel combination therapy approach for the treatment of TNBC that targets Drp1-mediated mitochondrial fission and autophagy pathways. Hydrogen sulfide (H2S) mediates a myriad of biological processes, including autophagy and mitochondrial function. In this study, we demonstrated that 5-(4-hydroxyphenyl)-3H-1,2-dithiole-3-thione (ADT-OH), one of the most widely utilized sustained-release H2S donors, effectively suppresses metastasis of TNBC cells in the absence of proliferation inhibition in vitro and in vivo. ADT-OH treatment ameliorated autophagy flux by suppressing autophagosome formation and induced mitochondrial elongation through decreasing expression of dynamin-related protein 1 (Drp1) and increasing expression of mitochondrial fusion protein (Mfn2). At the same time, ADT-OH downregulated mitophagy flux and inhibited mitochondrial function, eventually leading to the inhibition of migration and invasion in TNBC cells. In vivo, intraperitoneal administration of ADT-OH revealed a potent anti-metastatic activity in three different animal models, the MDA-MB-231 orthotopic xenograft model, the 4T1-Luci orthotopic model and the 4T1-Luci tail vein metastasis model. However, ADT-OH has an extremely low water solubility, which is a significant barrier to its effectiveness. Thus, we demonstrated that the solubility of ADT-OH in water can be improved significantly by absorption with hydroxypropyl-β-cyclodextrin (CD). Remarkably, the obtained CD-ADT-OH demonstrated superior anti-cancer effect to ADT-OH in vivo. Altogether, this study describes a novel regulator of mammalian mitochondrial fission and autophagy, with potential utility as an experimental therapeutic agent for metastatic TNBC.
    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
    骨肉瘤(OS)是一种高度恶性的原发性骨肿瘤,是年轻人癌症相关死亡的主要原因。GNE-477属于第二代mTOR抑制剂,在治疗OS方面具有广阔的潜力,但剂量耐受性和药物毒性限制了其开发和利用。本研究旨在制备一种用于GNE-477的新型H2O2刺激响应性十二烷酸(DA)-苯基硼酸酯-葡聚糖(DA-B-DEX)聚合物胶束递送系统,并评估其功效。对聚合物胶束进行了形貌表征,大小和临界胶束浓度。建立了GNE‑477加载的DA‑B‑DEX(GNE‑477@DBD)肿瘤靶向给药系统,并测量了GNE‑477的释放。利用荧光示踪技术分析了三种OS细胞系(MG‑63,U2OS和143B细胞)对GNE‑477@DBD的细胞摄取。羟基化的DA‑B成功接枝到葡聚糖上,接枝率为3%,适合形成两亲性胶束。暴露于H2O2后,DA-B-DEX胶束破裂并迅速释放药物,导致细胞对GNE‑477@DBD的摄取增加,持续释放GNE‑477。体外实验,包括MTT测定,流式细胞术,蛋白质印迹和RT-qPCR,证明GNE‑477@DBD抑制肿瘤细胞活力,细胞周期停滞在G1期,诱导细胞凋亡并阻断PI3K/Akt/mTOR级联反应。在体内,通过观察小鼠肿瘤生长情况和H&E染色结果,GNE‑477@DBD组比游离药物组表现出更积极的治疗结果,对其他器官几乎没有不良反应.总之,H2O2响应型DA‑B‑DEX为OS治疗的疏水性抗肿瘤药物提供了有前途的递送系统。
    Osteosarcoma (OS) is a highly malignant primary bone neoplasm that is the leading cause of cancer‑associated death in young people. GNE‑477 belongs to the second generation of mTOR inhibitors and possesses promising potential in the treatment of OS but dose tolerance and drug toxicity limit its development and utilization. The present study aimed to prepare a novel H2O2 stimulus‑responsive dodecanoic acid (DA)‑phenylborate ester‑dextran (DA‑B‑DEX) polymeric micelle delivery system for GNE‑477 and evaluate its efficacy. The polymer micelles were characterized by morphology, size and critical micelle concentration. The GNE‑477 loaded DA‑B‑DEX (GNE‑477@DBD) tumor‑targeting drug delivery system was established and the release of GNE‑477 was measured. The cellular uptake of GNE‑477@DBD by three OS cell lines (MG‑63, U2OS and 143B cells) was analyzed utilizing a fluorescent tracer technique. The hydroxylated DA‑B was successfully grafted onto dextran at a grafting rate of 3%, suitable for forming amphiphilic micelles. Following exposure to H2O2, the DA‑B‑DEX micelles ruptured and released the drug rapidly, leading to increased uptake of GNE‑477@DBD by cells with sustained release of GNE‑477. The in vitro experiments, including MTT assay, flow cytometry, western blotting and RT‑qPCR, demonstrated that GNE‑477@DBD inhibited tumor cell viability, arrested cell cycle in G1 phase, induced apoptosis and blocked the PI3K/Akt/mTOR cascade response. In vivo, through the observation of mice tumor growth and the results of H&E staining, the GNE‑477@DBD group exhibited more positive therapeutic outcomes than the free drug group with almost no adverse effects on other organs. In conclusion, H2O2‑responsive DA‑B‑DEX presents a promising delivery system for hydrophobic anti‑tumor drugs for OS therapy.
    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:三阴性乳腺癌(TNBC)的特征在于其高转移潜力,导致患者生存率低下。癌相关成纤维细胞(CAF)在通过诱导线粒体生物发生促进TNBC转移方面至关重要。然而,如何抑制CAF赋予的线粒体生物合成仍需探索。
    方法:我们使用伤口愈合和细胞侵袭试验研究了转移,3D文化,anoikis检测,和NOD/SCID小鼠。线粒体生物发生通过MitoTracker绿色FM染色检测,线粒体DNA水平的定量,和蓝色天然聚丙烯酰胺凝胶电泳。表达式,转录,通过蛋白质印迹法检测过氧化物酶体增殖物激活受体共激活因子1α(PGC-1α)的磷酸化,染色质免疫沉淀,双荧光素酶报告分析,定量聚合酶链反应,免疫沉淀,和液相色谱-串联质谱。使用Kaplan-Meier绘图仪数据库和临床乳腺癌组织样本评估了PGC-1α在TNBC中的预后作用。
    结果:我们证明PGC-1α提示淋巴结转移,肿瘤血栓形成,TNBC患者的生存率很低,它是由CAFs诱导的,在TNBC中充当线粒体生物发生和转移的诱导剂。紫草素阻碍了CAF诱导的PGC-1α表达,核定位,以及与雌激素相关受体α(ERRα)的相互作用,从而抑制靶向PGC-1α/ERRα的线粒体基因。机械上,PGC-1α的下调是由合酶激酶3β诱导的PGC-1α在Thr295的磷酸化介导的,这与神经前体细胞表达的发育下调的4e1识别以及随后通过泛素蛋白水解的降解有关。PGC-1α在Thr295的突变消除了紫草素对CAF刺激的TNBC线粒体生物发生和转移的体外和体内抑制作用。
    结论:我们的研究结果表明,PGC-1α是通过破坏线粒体生物发生来阻断TNBC转移的可行靶标,紫草素具有通过靶向PGC-1α作为线粒体生物发生抑制剂治疗TNBC转移的潜力。
    BACKGROUND: Triple-negative breast cancer (TNBC) is characterized by its high metastatic potential, which results in poor patient survival. Cancer-associated fibroblasts (CAFs) are crucial in facilitating TNBC metastasis via induction of mitochondrial biogenesis. However, how to inhibit CAF-conferred mitochondrial biogenesis is still needed to explore.
    METHODS: We investigated metastasis using wound healing and cell invasion assays, 3D-culture, anoikis detection, and NOD/SCID mice. Mitochondrial biogenesis was detected by MitoTracker green FM staining, quantification of mitochondrial DNA levels, and blue-native polyacrylamide gel electrophoresis. The expression, transcription, and phosphorylation of peroxisome-proliferator activated receptor coactivator 1α (PGC-1α) were detected by western blotting, chromatin immunoprecipitation, dual-luciferase reporter assay, quantitative polymerase chain reaction, immunoprecipitation, and liquid chromatography-tandem mass spectrometry. The prognostic role of PGC-1α in TNBC was evaluated using the Kaplan-Meier plotter database and clinical breast cancer tissue samples.
    RESULTS: We demonstrated that PGC-1α indicated lymph node metastasis, tumor thrombus formation, and poor survival in TNBC patients, and it was induced by CAFs, which functioned as an inducer of mitochondrial biogenesis and metastasis in TNBC. Shikonin impeded the CAF-induced PGC-1α expression, nuclear localization, and interaction with estrogen-related receptor alpha (ERRα), thereby inhibiting PGC-1α/ERRα-targeted mitochondrial genes. Mechanistically, the downregulation of PGC-1α was mediated by synthase kinase 3β-induced phosphorylation of PGC-1α at Thr295, which associated with neural precursor cell expressed developmentally downregulated 4e1 recognition and subsequent degradation by ubiquitin proteolysis. Mutation of PGC-1α at Thr295 negated the suppressive effects of shikonin on CAF-stimulated TNBC mitochondrial biogenesis and metastasis in vitro and in vivo.
    CONCLUSIONS: Our findings indicate that PGC-1α is a viable target for blocking TNBC metastasis by disrupting mitochondrial biogenesis, and that shikonin merits potential for treatment of TNBC metastasis as an inhibitor of mitochondrial biogenesis through targeting PGC-1α.
    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
    在非小细胞肺癌(NSCLC)中,药物耐受性已成为驱动靶向治疗(TT)抵抗的主要非遗传适应性过程之一。然而,控制这种适应性反应的分子事件的动力学和顺序仍然知之甚少。这里,我们将细胞周期动力学的实时监测和单细胞RNA测序结合在一组广泛的致癌成瘾中,如EGFR-,ALK-,BRAF和KRAS突变型非小细胞肺癌,用相应的TT治疗。我们确定了药物适应的共同路径,它总是涉及肺泡1型(AT1)分化和Rho相关蛋白激酶(ROCK)介导的细胞骨架重塑。我们还分离并描述了罕见的早期逃生者,它们代表了在治疗的最初几个小时内从AT1样群体中出现的最早的抗性起始细胞。表型药物筛选确定法尼基转移酶抑制剂(FTI)如替比法尼是在几种致癌成瘾模型中体外和体内预防TT复发的最有效药物。法尼基转移酶的遗传耗竭证实了这一点。这些发现为结合TT和FTI的治疗方法的发展铺平了道路,以有效预防癌基因成瘾的NSCLC患者的肿瘤复发。
    Drug-tolerance has emerged as one of the major non-genetic adaptive processes driving resistance to targeted therapy (TT) in non-small cell lung cancer (NSCLC). However, the kinetics and sequence of molecular events governing this adaptive response remain poorly understood. Here, we combine real-time monitoring of the cell-cycle dynamics and single-cell RNA sequencing in a broad panel of oncogenic addiction such as EGFR-, ALK-, BRAF- and KRAS-mutant NSCLC, treated with their corresponding TT. We identify a common path of drug adaptation, which invariably involves alveolar type 1 (AT1) differentiation and Rho-associated protein kinase (ROCK)-mediated cytoskeletal remodeling. We also isolate and characterize a rare population of early escapers, which represent the earliest resistance-initiating cells that emerge in the first hours of treatment from the AT1-like population. A phenotypic drug screen identify farnesyltransferase inhibitors (FTI) such as tipifarnib as the most effective drugs in preventing relapse to TT in vitro and in vivo in several models of oncogenic addiction, which is confirmed by genetic depletion of the farnesyltransferase. These findings pave the way for the development of treatments combining TT and FTI to effectively prevent tumor relapse in oncogene-addicted NSCLC patients.
    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
    肺癌是最常见和最致命的癌症之一。临床前模型对于研究考虑肿瘤遗传学的新疗法和组合至关重要。我们已经从具有不同遗传背景的细胞系中建立了表达荧光素酶基因的细胞系,常见于肺腺癌患者。我们通过测试这些品系对多种药物的反应来表征这些品系。因此,我们开发了非小细胞肺癌原位临床前小鼠模型,其移植效率非常高.这些模型可以轻松监测肿瘤的生长,特别是对治疗的反应,以及肿瘤细胞在体内的传播。我们表明,奥希替尼(第三代酪氨酸激酶抑制剂靶向突变的EGFR)和贝伐单抗(抗血管生成靶向VEGF)的联合治疗可以对EGFR突变的肿瘤产生有益的治疗效果。我们还表明,在奥希替尼治疗的肿瘤中加入阿法替尼导致肿瘤生长抑制。使用司美替尼或辛伐他汀没有观察到这种效果。因此,这些临床前小鼠模型可以测试创新的治疗组合,也是研究抗性机制的首选工具。
    Lung cancer is one of the most common and deadliest cancers. Preclinical models are essential to study new therapies and combinations taking tumor genetics into account. We have established cell lines expressing the luciferase gene from lines with varied genetic backgrounds, commonly encountered in patients with pulmonary adenocarcinoma. We have characterized these lines by testing their response to multiple drugs. Thus, we have developed orthotopic preclinical mouse models of NSCLC with very high engraftment efficiency. These models allow the easy monitoring of tumor growth, particularly in response to treatment, and of tumor cells dissemination in the body. We show that concomitant treatment with osimertinib (3rd generation tyrosine kinase inhibitor targeting mutated EGFR) and bevacizumab (anti-angiogenic targeting VEGF) can have a beneficial therapeutic effect on EGFR-mutated tumors. We also show that the addition of afatinib to osimertinib-treated tumors in escape leads to tumor growth inhibition. No such effect is observed with selumetinib or simvastatin. These preclinical mouse models therefore make it possible to test innovative therapeutic combinations and are also a tool of choice for studying resistance mechanisms.
    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
    双特异性抗体(BsAb)可以同时靶向不同抗原靶标的两个表位,为抗体药物设计的多样性带来了可能性,并且是治疗癌症和其他疾病的有希望的工具。T细胞接合bsAb是双特异性抗体的重要应用,可以通过同时靶向肿瘤相关抗原(TAA)和CD3来促进T细胞介导的肿瘤细胞杀伤。
    这项研究包括抗体纯化,用于抗原结合的Elisa测定,细胞毒性试验,体外流式细胞术检测T细胞活化和体内异种肿瘤模型。
    我们提出了一种名为PHE-Ig技术的新型bsAb平台,通过替换不同单克隆抗体的CH1/CL区促进同源重链(HC)-轻链(LC)配对。我们还验证了PHE-Ig技术可以有效地用作平台来合成不同的所需bsAb用于T细胞免疫疗法。尤其是,BCMA×CD3PHE-IgbsAb在体外和体内表现出强大的抗多发性骨髓瘤(MM)活性。
    此外,PHE1结构域进一步缩短D14G和R41S突变,名叫PHE-S,基于PHE-S的BCMA×CD3bsAb在体内和体外也显示出抗BCMA肿瘤作用,为不同bsAb的开发和优化带来更多可能性。总而言之,用于bsAb构建的基于PHE1的IgG样抗体平台为增强的T细胞免疫疗法提供了新策略。
    UNASSIGNED: Bispecific antibodies (BsAbs) can simultaneously target two epitopes of different antigenic targets, bringing possibilities for diversity in antibody drug design and are promising tools for the treatment of cancers and other diseases. T-cell engaging bsAb is an important application of the bispecific antibody, which could promote T cell-mediated tumor cell killing by targeting tumor-associated antigen (TAA) and CD3 at the same time.
    UNASSIGNED: This study comprised antibodies purification, Elisa assay for antigen binding, cytotoxicity assays, T cell activation by flow cytometry in vitro and xenogenic tumor model in vivo.
    UNASSIGNED: We present a novel bsAb platform named PHE-Ig technique to promote cognate heavy chain (HC)-light chain (LC) pairing by replacing the CH1/CL regions of different monoclonal antibodies (mAbs) with the natural A and B chains of PHE1 fragment of Integrin β2 based on the knob-in-hole (KIH) technology. We had also verified that PHE-Ig technology can be effectively used as a platform to synthesize different desired bsAbs for T-cell immunotherapy. Especially, BCMA×CD3 PHE-Ig bsAbs exhibited robust anti-multiple myeloma (MM) activity in vitro and in vivo.
    UNASSIGNED: Moreover, PHE1 domain was further shortened with D14G and R41S mutations, named PHE-S, and the PHE-S-based BCMA×CD3 bsAbs also showed anti BCMA+ tumor effect in vitro and in vivo, bringing more possibilities for the development and optimization of different bsAbs. To sum up, PHE1-based IgG-like antibody platform for bsAb construction provides a novel strategy for enhanced T-cell immunotherapy.
    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
    柴胡皂苷D(SSD),来自于柴胡,具有各种药理特性,包括免疫调节,抗炎,和抗过敏作用。一些研究已经调查了SSD对多器官癌症的抗肿瘤作用。然而,其在结直肠癌(CRC)中的作用尚不清楚.因此,本研究旨在阐明SSD对CRC细胞存活和转移的抑制作用。SSD降低了CRC细胞的存活和集落形成能力。使用流式细胞术测量SSD诱导的CRC细胞自噬和凋亡。SSD处理增加CRC细胞中LC3B和p62自噬因子水平。此外,SSD诱导的细胞凋亡是通过caspase-9,caspase-3和PARP的裂解而发生的,随着Bcl-2家族的下调。在体内实验中,口服SSD后观察到肺部转移性肿瘤结节数量减少.基于这些结果,SSD通过诱导自噬和凋亡抑制CRC细胞向肺转移。总之,SSD抑制CRC细胞的增殖和转移,提示其作为转移性CRC治疗新物质的潜力。
    Saikosaponin D (SSD), derived from Bupleurum falcatum L., has various pharmacological properties, including immunoregulatory, anti-inflammatory, and anti-allergic effects. Several studies have investigated the anti-tumor effects of SSD on cancer in multiple organs. However, its role in colorectal cancer (CRC) remains unclear. Therefore, this study aimed to elucidate the suppressive effects of SSD on CRC cell survival and metastasis. SSD reduced the survival and colony formation ability of CRC cells. SSD-induced autophagy and apoptosis in CRC cells were measured using flow cytometry. SSD treatment increased LC3B and p62 autophagic factor levels in CRC cells. Moreover, SSD-induced apoptosis occurred through the cleavage of caspase-9, caspase-3, and PARP, along with the downregulation of the Bcl-2 family. In the in vivo experiment, a reduction in the number of metastatic tumor nodules in the lungs was observed after the oral administration of SSD. Based on these results, SSD inhibits the metastasis of CRC cells to the lungs by inducing autophagy and apoptosis. In conclusion, SSD suppressed the proliferation and metastasis of CRC cells, suggesting its potential as a novel substance for the metastatic CRC treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    前列腺癌(PC)是全球男性最常见的癌症。目前,去势抵抗前列腺癌(CRPC),对雄激素剥夺疗法有抗性,预后不良,是一个治疗问题。我们研究了抗体中和分泌的整合素和含金属蛋白酶结构域的蛋白9(sADAM9)对PC的抗肿瘤作用,这是一种血液可溶的形式。我们进行了增殖试验,伤口愈合试验,入侵检测,Westernblot(WB),以及一项体内研究,其中将sADAM9中和抗体瘤内给予携带PC的小鼠。在入侵检测中,sADAM9中和抗体显著抑制所有细胞系的侵袭(TRAMP-C2:p=0.00776,LNCaP:p=0.000914,PC-3:p=0.0327,和DU145:p=0.0254)。我们检查了上皮-间质转化(EMT)标志物,转移机制之一,在WB中显示Slug在TRAMP-C2,LNCaP,和DU145以及通过sADAM9中和在TRAMP-C2和PC-3中上调E-钙黏着蛋白。在老鼠实验中,与对照组相比,sADAM9中和抗体显着抑制肿瘤生长(TRAMP-C2为1.68倍,LNCaP为1.89倍,PC-3中的2.67倍)。这些结果表明,sADAM9中和抗体抑制侵袭,迁移,和PC中的肿瘤生长。以前的研究检查了敲低总ADAM9或sADAM9的抗肿瘤作用,但这项研究使用了中和sADAM9抗体的新技术。这可能是新颖的,因为没有使用sADAM9的中和抗体的动物研究来观察ADAM9表达与前列腺癌之间的关系。
    Prostate cancer (PC) is the most common cancer diagnosed in men worldwide. Currently, castration-resistant prostate cancer (CRPC), which is resistant to androgen deprivation therapy, has a poor prognosis and is a therapeutic problem. We investigated the antitumor effects on PC of an antibody neutralizing secreted disintegrin and metalloproteinase domain-containing protein 9 (sADAM9), which is a blood-soluble form. We performed proliferation assays, wound healing assays, invasion assays, Western blot (WB), and an in vivo study in which a sADAM9 neutralizing antibody was administered intratumorally to PC-bearing mice. In invasion assays, the sADAM9 neutralizing antibody significantly inhibited invasion in all cell lines (TRAMP-C2: p = 0.00776, LNCaP: p = 0.000914, PC-3: p = 0.0327, and DU145: p = 0.0254). We examined epithelial-mesenchymal transition (EMT) markers, one of the metastatic mechanisms, in WB and showed downregulation of Slug in TRAMP-C2, LNCaP, and DU145 and upregulation of E-cadherin in TRAMP-C2 and PC-3 by sADAM9 neutralization. In mouse experiments, the sADAM9 neutralizing antibody significantly suppressed tumor growth compared to controls (1.68-fold in TRAMP-C2, 1.89-fold in LNCaP, and 2.67-fold in PC-3). These results suggested that the sADAM9 neutralizing antibody inhibits invasion, migration, and tumor growth in PC. Previous studies examined the anti-tumor effect of knockdown of total ADAM9 or sADAM9, but this study used the new technology of neutralizing antibodies for sADAM9. This may be novel because there was no animal study using a neutralizing antibody for sADAM9 to see the relationship between ADAM9 expression and prostate 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
    靶向癌症治疗旨在破坏调节癌症进展的蛋白质的功能,主要通过使用小分子抑制剂(SMI)。SMI通过调节信号通路发挥作用,细胞器完整性,染色质成分,和几个对细胞分裂和存活至关重要的生物合成过程。与正常细胞相比,抗凋亡蛋白BCL2在许多癌症中高度上调,使其成为癌症治疗的理想靶点。大约75%的原发性乳腺癌过度表达BCL2,为探索BCL2抑制剂作为治疗选择提供了机会。Disarib是一种SMI,已被开发为选择性BCL2抑制剂。Disarib通过破坏BCL2-BAK相互作用并激活白血病细胞中的内在凋亡途径而工作,同时保留正常细胞。我们调查了Disarib的影响,BCL2特异性抑制剂,在乳腺癌细胞和异种移植物上。细胞毒性和荧光分析显示,Disarib通过增加三阴性乳腺癌细胞(MDA-MB-231和MDA-MB-468)中的活性氧和激活内在凋亡途径来诱导细胞死亡。Disarib还影响这些细胞的集落形成特性。MDA-MB-231-和MDA-MB-468衍生的异种移植物在Disarib治疗后显示出肿瘤的显着减少。通过转录组学方法,我们还探讨了BCL2抑制剂对能量代谢的影响,线粒体动力学,和上皮-间质转化(EMT)。线粒体动力学和糖代谢主要调节能量代谢。能量学的变化通过上皮-间质转化调节肿瘤生长,和血管生成。RNA测序(RNAseq)分析显示,BCL2抑制剂ABT-199和Disarib维持MDA-MB-231中的Oxphos水平。然而,关键的糖酵解基因显著下调。在RNAseq数据和Disarib处理的TNBC细胞和异种移植物中,线粒体裂变基因均被下调。最后,Disarib抑制伤口愈合和上皮-间质转化。这项研究表明Disarib会破坏线粒体功能,激活乳腺癌的内在凋亡途径,并在体外和体内抑制上皮-间质转化。这些发现强调了Disarib作为三阴性乳腺癌患者多方面治疗策略的潜力。
    Targeted cancer therapy aims to disrupt the functions of proteins that regulate cancer progression, mainly by using small molecule inhibitors (SMIs). SMIs exert their effect by modulating signalling pathways, organelle integrity, chromatin components, and several biosynthetic processes essential for cell division and survival. Antiapoptotic protein BCL2 is highly upregulated in many cancers compared with normal cells, making it an ideal target for cancer therapy. Around 75% of primary breast cancers overexpress BCL2, providing an opportunity to explore BCL2 inhibitors as a therapeutic option. Disarib is an SMI that has been developed as a selective BCL2 inhibitor. Disarib works by disrupting BCL2-BAK interaction and activating intrinsic apoptotic pathways in leukemic cells while sparing normal cells. We investigated the effects of Disarib, a BCL2 specific inhibitor, on breast cancer cells and xenografts. Cytotoxicity and fluorometric assays revealed that Disarib induced cell death by increasing reactive oxygen species and activating intrinsic apoptotic pathways in Triple-Negative Breast Cancer cells (MDA-MB-231 and MDA-MB-468). Disarib also affected the colony-forming properties of these cells. MDA-MB-231- and MDA-MB-468-derived xenografts showed a significant reduction in tumours upon Disarib treatment. Through the transcriptomics approach, we also explored the influence of BCL2 inhibitors on energy metabolism, mitochondrial dynamics, and epithelial-to-mesenchymal transition (EMT). Mitochondrial dynamics and glucose metabolism mainly regulate energy metabolism. The change in energetics regulates tumour growth through epithelial-mesenchymal transition, and angiogenesis. RNA sequencing (RNAseq) analysis revealed that BCL2 inhibitors ABT-199 and Disarib maintain Oxphos levels in MDA-MB-231. However, key glycolytic genes were significantly downregulated. Mitochondrial fission genes were seen to be downregulated both in RNAseq data and semi quantitative real time polymerase chain reaction (qRTPCR) in Disarib-treated TNBC cells and xenografts. Lastly, Disarib inhibited wound healing and epithelial-to-mesenchymal transition. This study showed that Disarib disrupts mitochondrial function, activates the intrinsic apoptotic pathway in breast cancer, and inhibits epithelial-to-mesenchymal transition both in vitro and in vivo. These findings highlight Disarib\'s potential as a multifaceted therapeutic strategy for patients with Triple-Negative 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
    癌症的化疗耐药性是导致高死亡率的重要因素。肿瘤多药耐药是自噬过程的结果。我们先前的研究发现,化合物1-硝基-2酰基蒽醌-亮氨酸(C2)表现出优异的抗结直肠癌(CRC)活性,涉及自噬和凋亡相关蛋白,而其潜在机制尚不清楚。这项研究的一个值得注意的方面是C2如何克服HCT116/L-OHP的多药敏感性,对体外和体内奥沙利铂(反式-/-二氨基环己烷草酸铂;L-OHP)均具有抗性的结肠癌细胞系。在异种移植肿瘤小鼠模型中,我们发现,C2和L-OHP的混合物逆转了HCT116/L-OHP细胞对L-OHP的抗性,并抑制了肿瘤的生长;此外,C2下调P-gp和BCRP的基因表达水平,降低P-gp的药物外排活性。重要的是要注意,虽然C2使HCT116/L-OHP细胞对L-OHP重新敏感,它还触发了保护性的自噬途径。裂解的caspase-3和Beclin1的表达水平稳步上升。PI3K的表达,磷酸化AKT,mTOR降低了,而p53增加。我们证明蒽醌衍生物C2充当L-OHP敏化剂并逆转HCT116/L-OHP细胞对L-OHP的抗性。提示C2可通过介导p53和PI3K/AKT/mTOR信号通路诱导HCT116/L-OHP细胞自噬。
    Chemotherapy resistance in cancer is an essential factor leading to high mortality rates. Tumor multidrug resistance arises as a result of the autophagy process. Our previous study found that compound 1-nitro-2 acyl anthraquinone-leucine (C2) exhibited excellent anti-colorectal cancer (CRC) activity involving autophagy and apoptosis-related proteins, whereas its underlying mechanism remains unclear. A notable aspect of this study is how C2 overcomes the multidrug susceptibility of HCT116/L-OHP, a colon cancer cell line that is resistant to both in vitro and in vivo oxaliplatin (trans-/-diaminocyclohexane oxalatoplatinum; L-OHP). In a xenograft tumor mouse model, we discovered that the mixture of C2 and L-OHP reversed the resistance of HCT116/L-OHP cells to L-OHP and inhibited tumor growth; furthermore, C2 down-regulated the gene expression levels of P-gp and BCRP and decreased P-gp\'s drug efflux activity. It is important to note that while C2 re-sensitized the HCT116/L-OHP cells to L-OHP for apoptosis, it also triggered a protective autophagic pathway. The expression levels of cleaved caspase-3 and Beclin 1 steadily rose. Expression of PI3K, phosphorylated AKT, and mTOR were decreased, while p53 increased. We demonstrated that the anthraquinone derivative C2 acts as an L-OHP sensitizer and reverses resistance to L-OHP in HCT116/L-OHP cells. It suggests that C2 can induce autophagy in HCT116/L-OHP cells by mediating p53 and the PI3K/AKT/mTOR signaling pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号