MZ1

MZ1
  • 文章类型: Journal Article
    目的:本研究旨在发现三阴性乳腺癌(TNBC)细胞系的增殖和迁移是否可以通过布罗莫结构域和末端外结构域(BET)抑制剂JQ1和BET蛋白靶向嵌合体(PROTACs)ARV-771和MZ1治疗来降低。
    方法:细胞毒性试验,用于癌症相关蛋白蛋白表达的划痕迁移测定和蛋白质印迹蛋白质组分析器阵列用于评估BET抑制剂和两种BET定向PROTACs对细胞活力的影响。迁移和蛋白质表达。
    结果:JQ1和PROTACsMZ1和ARV-771显著抑制KRASG13D突变的MDA-MB-231细胞的生长和迁移。在这个细胞系中,PROTACs抑制了ERBB2/HER2,3和4的残留表达,这对乳腺癌细胞的增殖至关重要,并且该细胞系被证明对HER2抑制剂敏感.相比之下,PROTACs对MDA-MB-436细胞蛋白质表达的影响主要影响细胞因子及其同源受体。
    结论:通过PROTACs降解BET蛋白表现出显著的抗增殖作用。KRAS突变的MDA-MB-231细胞属于低HER2表达肿瘤,其与无HER2患者相比具有较差的预后。由于第一个口服抗肿瘤激素受体的PROTACs正在临床试验中,这种肿瘤治疗模式有望成为未来TNBC治疗的重要治疗策略。
    OBJECTIVE: This study aims to find whether the proliferation and migration of triple negative breast cancer (TNBC) cell lines can be reduced by treatment with bromodomain and extra-terminal domain (BET) inhibitor JQ1 and BET protein targeting chimeras (PROTACs) ARV-771 and MZ1.
    METHODS: Cytotoxicity tests, scratch migration assays and western blot proteome profiler arrays for protein expression of cancer-related proteins were used to evaluate the impact of a BET-inhibitor and two BET-directed PROTACs on cell viability, migration and on protein expression.
    RESULTS: JQ1 and the PROTACs MZ1 and ARV-771 significantly inhibited the growth and migration of the KRAS G13D-mutated MDA-MB-231 cells. In this cell line, the PROTACs suppressed the residual expression of ERBB2/HER2, 3 and 4 that are essential for the proliferation of breast cancer cells and this cell line proved sensitive to HER2 inhibitors. In contrast, the effects of the PROTACs on the protein expression of MDA-MB-436 cells mostly affected cytokines and their cognate receptors.
    CONCLUSIONS: The degradation of BET-protein by PROTACs demonstrated significant anti-proliferative effects. The KRAS-mutated MDA-MB-231 cells belong to the low-HER2 expressing tumors that have a poorer prognosis compared to HER2-null patients. Since first oral PROTACs against tumor hormone receptors are in clinical trials, this mode of tumor therapy is expected to become an important therapeutic strategy in the future treatment of 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
    B细胞淋巴瘤(BCL)是狗中最常见的血液肿瘤。治疗通常包括化疗,使用基于CHOP的协议。然而,结果总体上仍然很差,敦促探索具有针对性的新治疗策略。Myc转录因子在调节细胞过程中起着至关重要的作用,它的失调与许多人类和犬类恶性肿瘤有关,包括犬BCL(cBCL)。本研究旨在评估在两种体外模型(CLBL-1和KLR-1201)中使用BI2536和MZ1化合物间接抑制cBCL中Myc的功效。BI2536和MZ1,单独和组合,以显着的浓度和时间依赖性方式影响细胞活力。Western印迹显示,用BI2536处理后,两种细胞系中PLK1的表达均上调,并伴有c-Myc蛋白水平的降低。相反,MZ1导致其主要目标减少,BRD4,以及c-Myc的减少。此外,与单独的MZ1相比,BI2536单独和与MZ1组合在细胞中诱导更大的转录组变化,主要影响MYC靶基因和参与细胞周期调控的基因。这些数据强调了Myc作为cBCL治疗靶点的潜在作用,提供了一种间接调节该分子的新方法。
    B-cell lymphomas (BCL) is the most frequent hematological cancer in dogs. Treatment typically consists of chemotherapy, with CHOP-based protocols. However, outcome remains generally poor, urging the exploration of new therapeutic strategies with a targeted approach. Myc transcription factor plays a crucial role in regulating cellular processes, and its dysregulation is implicated in numerous human and canine malignancies, including canine BCL (cBCL). This study aims to evaluate the efficacy of indirectly inhibiting Myc in cBCL using BI2536 and MZ1 compounds in two in vitro models (CLBL-1 and KLR-1201). Both BI2536 and MZ1, alone and combined, affected cell viability in a significant concentration- and time-dependent manner. Western Blot revealed an upregulation of PLK1 expression in both cell lines upon treatment with BI2536, in association with a reduction in c-Myc protein levels. Conversely, MZ1 led to a decrease in its primary target, BRD4, along with a reduction in c-Myc. Furthermore, BI2536, both alone and in combination with MZ1, induced larger transcriptomic changes in cells compared to MZ1 alone, primarily affecting MYC target genes and genes involved in cell cycle regulation. These data underscore the potential role of Myc as therapeutic target in cBCL, providing a novel approach to indirectly modulate this molecule.
    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:晚期骨肉瘤预后较差,需要新的靶向疗法,特别是转移性疾病。溴结构域抑制剂(BETi)是表观遗传调节剂,可广泛损害致癌蛋白的表达并发挥抗肿瘤作用。BETi可以与化疗剂组合以增加具有优异效果的治疗反应,以蛋白水解靶向嵌合体(PROTACs)的形式在多个循环中降解感兴趣的蛋白质(POI)。这项工作旨在研究BETi的疗效,例如JQ1,dBET57和MZ1PROTACs与针对骨肉瘤细胞系的细胞毒性药物组合。
    方法:骨肉瘤细胞系HOS的化学敏感性,Saos-2、MG-63和G292单独使用BET定向药物或与包含顺铂的细胞毒性药物组合进行测试。阿霉素,托泊替康,和吉西他滨使用细胞活力测定。
    结果:BET降解物对HOS细胞表现出最高的毒性,并与化学疗法组合显示出协同活性,降解剂-拓扑替康/吉西他滨组合除外。对于更具化学抗性的Saos-2细胞,发现BET试剂和化学治疗剂之间的协同作用最高,并且对于BET试剂-阿霉素组合和MZ1-托泊替康对,MG-63细胞的毒性增强。HOS和Saos-2细胞系的AXL蛋白表达降低,BCL-X,e-cadherin,CAIX,EpCAM,ErbB2和波形蛋白响应JQ1、MZ1和BET57。
    结论:该研究表明,新型BETPROTACs联合化疗药物的应用可能是改善骨肉瘤治疗的一种新的治疗选择。第一个口服可用的PROTACs已经达到临床试验。
    OBJECTIVE: Osteosarcoma at an advanced stage has a poor outcome, and novel targeted therapies are needed, especially for metastatic disease. Bromodomain inhibitors (BETi) are epigenetic modulators that broadly impair the expression of oncogenic proteins and exert antitumor effects. BETi can be combined with chemotherapeutics to increase therapeutic responses with superior effects in the form of proteolysis targeting chimeras (PROTACs) that degrade proteins of interest (POI) in multiple cycles. This work aimed to investigate the efficacy of BETi, such as JQ1, dBET57, and MZ1 PROTACs in combination with cytotoxic drugs against osteosarcoma cell lines.
    METHODS: Chemosensitivity of the osteosarcoma cell lines HOS, Saos-2, MG-63, and G292 were tested with BET-directed agents alone or in combination with cytotoxic drugs comprising cisplatin, doxorubicin, topotecan, and gemcitabine using cell viability assays.
    RESULTS: The BET degraders exhibited highest toxicity to HOS cells and showed synergistic activity in combination with the chemotherapeutics, except for the degrader - topotecan/gemcitabine combinations. Highest synergy between BET agents and chemotherapeutics were found for the more chemoresistant Saos-2 cells and potentiation of toxicity in MG-63 cells for the BET agents - doxorubicin combinations and the MZ1-topotecan pair. HOS and Saos-2 cell lines had reduced protein expression of AXL, BCL-X, e-cadherin, CAIX, EpCAM, ErbB2, and vimentin in response to JQ1, MZ1, and BET57.
    CONCLUSIONS: The study suggests that the application of novel BET PROTACs in combination with chemotherapeutics could represent a new therapeutic option to improve the therapy of osteosarcomas. First orally available PROTACs have reached clinical trials.
    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
    B细胞急性淋巴细胞白血病(B-ALL)是影响儿童的最常见的恶性肿瘤。虽然大多数B-ALL患者(90%)成功康复,早期复发的B-ALL病例继续表现出较高的死亡率.MZ1,一种新的溴结构域和末端外(BET)蛋白抑制剂,已证明对血液恶性肿瘤具有有效的抗肿瘤活性。这项研究的目的是检查MZ1在B-ALL治疗中的作用和治疗潜力。
    为了确定MZ1的基本机制,对B-ALL细胞系进行了一系列体外测定,包括细胞计数试剂盒8(CCK8)测定,碘化丙啶(PI)染色,和膜联蛋白V/PI染色。采用蛋白质印迹和定量实时聚合酶链反应(qRT-PCR)检测蛋白质和mRNA表达水平。转录组RNA测序(RNA-seq)用于筛选MZ1的靶基因,并采用慢病毒转染来建立稳定表达/敲低的细胞系。
    已观察到MZ1诱导含溴结构域4(BRD4)的降解,含溴结构域3(BRD3),B-ALL细胞株中的含溴结构域2(BRD2),导致体外抑制细胞生长,诱导细胞凋亡和周期阻滞。这些发现表明,MZ1对B-ALL的两种不同的分子亚型表现出细胞毒性作用。即697(TCF3/PBX1)和RS4;11(MLL-AF4)B-ALL细胞系。此外,RNA测序分析显示MZ1显著下调细胞周期蛋白D3(CCND3)基因在B-ALL细胞系中的表达,反过来促进细胞凋亡,细胞周期受阻,并引起细胞增殖抑制。
    我们的结果表明MZ1具有潜在的抗B-ALL作用,可能是一个新的治疗靶点。
    UNASSIGNED: B-cell acute lymphoblastic leukemia (B-ALL) is the most prevalent malignant tumor affecting children. While the majority of B-ALL patients (90%) experience successful recovery, early relapse cases of B-ALL continue to exhibit high mortality rates. MZ1, a novel inhibitor of Bromodomains and extra-terminal (BET) proteins, has demonstrated potent antitumor activity against hematological malignancies. The objective of this study was to examine the role and therapeutic potential of MZ1 in the treatment of B-ALL.
    UNASSIGNED: In order to ascertain the fundamental mechanism of MZ1, a sequence of in vitro assays was conducted on B-ALL cell lines, encompassing Cell Counting Kit 8 (CCK8) assay, Propidium iodide (PI) staining, and Annexin V/PI staining. Western blotting and quantitative real-time polymerase chain reaction (qRT-PCR) were employed to examine protein and mRNA expression levels. Transcriptomic RNA sequencing (RNA-seq) was utilized to screen the target genes of MZ1, and lentiviral transfection was employed to establish stably-expressing/knockdown cell lines.
    UNASSIGNED: MZ1 has been observed to induce the degradation of Bromodomain Containing 4 (BRD4), Bromodomain Containing 3 (BRD3), and Bromodomain Containing 2 (BRD2) in B-ALL cell strains, leading to inhibited cell growth and induction of cell apoptosis and cycle arrest in vitro. These findings suggest that MZ1 exhibits cytotoxic effects on two distinct molecular subtypes of B-ALL, namely 697 (TCF3/PBX1) and RS4;11 (MLL-AF4) B-ALL cell lines. Additionally, RNA-sequencing analysis revealed that MZ1 significantly downregulated the expression of Cyclin D3 (CCND3) gene in B-ALL cell lines, which in turn promoted cell apoptosis, blocked cell cycle, and caused cell proliferation inhibition.
    UNASSIGNED: Our results suggest that MZ1 has potential anti-B-ALL effects and might be a novel therapeutic target.
    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
    急性髓性白血病(AML)是一种高度癌性和侵袭性的血液系统疾病,耐药性和复发水平升高,导致高死亡率。最近,溴结构域和末端外(BET)蛋白抑制剂作为潜在的抗癌剂在血液肿瘤中得到了广泛的研究。MZ1是一种新型的BET抑制剂,通过蛋白水解靶向嵌合体(PROTAC)技术介导选择性蛋白质降解和抑制肿瘤生长。因此,本研究旨在探讨MZ1在AML中的作用和治疗潜力.在这项研究中,我们首次发现,BRD4高表达的AML患者的总体生存率低于低表达组.MZ1在体外抑制AML细胞生长并诱导凋亡和周期阻滞。MZ1诱导AML细胞株BRD4、BRD3和BRD2降解。此外,MZ1还启动了聚ADP-核糖聚合酶(PARP)的裂解,对NB4(PML-RARa)显示出细胞毒性作用,K562(BCR-ABL),Kasumi-1(AML1-ETO),和代表AML的不同分子亚型的MV4-11(MLL-AF4)细胞系。在AML小鼠白血病模型中,MZ1显著降低白血病细胞的生长并增加小鼠的存活时间。根据RNA测序分析,MZ1导致c-Myc和ANP32B基因在AML细胞系中显著下调。敲除ANP32B促进AML细胞凋亡并抑制细胞生长。总的来说,我们的数据表明,MZ1对具有不同分子病变的AML细胞系具有广泛的抗癌作用,这可能被用作AML患者的一种新的治疗策略。
    Acute myeloid leukemia (AML) is a highly cancerous and aggressive hematologic disease with elevated levels of drug resistance and relapse resulting in high mortality. Recently, bromodomains and extra-terminal (BET) protein inhibitors have been extensively researched in hematological tumors as potential anticancer agents. MZ1 is a novel BET inhibitor that mediates selective proteins degradation and suppression of tumor growth through proteolysis-targeting chimeras (PROTAC) technology. Accordingly, this study aimed to investigate the role and therapeutic potential of MZ1 in AML. In this study, we first identified that AML patients with high BRD4 expression had poor overall survival than those with low expression group. MZ1 inhibited AML cell growth and induced apoptosis and cycle arrest in vitro. MZ1 induced degradation of BRD4, BRD3 and BRD2 in AML cell strains. Additionally, MZ1 also initiated the cleavage of poly-ADP-ribose polymerase (PARP), which showed cytotoxic effects on NB4 (PML-RARa), K562 (BCR-ABL), Kasumi-1 (AML1-ETO), and MV4-11 (MLL-AF4) cell lines representing different molecular subtypes of AML. In AML mouse leukemia model, MZ1 significantly decreased leukemia cell growth and increased the mouse survival time. According to the RNA-sequencing analysis, MZ1 led to c-Myc and ANP32B genes significant downregulation in AML cell lines. Knockdown of ANP32B promoted AML cell apoptosis and inhibited cell growth. Overall, our data indicated that MZ1 had broad anti-cancer effects on AML cell lines with different molecular lesions, which might be exploited as a novel therapeutic strategy for AML 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
    神经母细胞瘤(NB)是儿童常见的实体瘤,而MYCN基因扩增高危人群中多数患者预后较差。近年来,抑制溴结构域和额外末端(BET)蛋白在研究MYCN驱动的恶性肿瘤中显示出相当大的希望。MZ1是一种新型的BET抑制剂,它采用蛋白水解靶向嵌合体(PROTAC)技术,用于蛋白酶体降解靶蛋白,并在某些肿瘤中显示出优异的效果。但其在神经母细胞瘤中的作用仍知之甚少。在这里,我们观察到MZ1抑制MYC扩增的NB细胞增殖和正常细胞周期,同时促进细胞凋亡。MZ1还在体内提供显著的治疗效果。机械上,MZ1通过抑制N-Myc或C-Myc的表达以及MAPK信号通路在NB细胞中表现出抗肿瘤作用。总的来说,我们的数据暗示MZ1可能被用作NB治疗的一种可能的治疗方法.
    Neuroblastoma(NB) is a common childhood solid tumor, and most patients in the high-risk group with MYCN gene amplification have a poor prognosis. Inhibition of bromodomain and extra terminal (BET) proteins has shown considerable promise in the investigation of MYCN-driven malignancies in recent years. MZ1 is a novel BET inhibitor that employs proteolytic-targeting chimera (PROTAC) technology for proteasomal degradation of target proteins and has shown excellent effects in some tumors, but its role in neuroblastoma remains poorly understood. Herein, we observed that MZ1 suppressed MYC-amplified NB cell proliferation and normal cell cycle, while simultaneously boosting cell apoptosis. MZ1 also provides a significant therapeutic impact in vivo. Mechanistically, MZ1 exhibits anti-tumor effect in NB cells by suppressing the expression of N-Myc or C-Myc as well as the MAPK signaling pathway. Overall, our data imply that MZ1 might be exploited as a possible therapeutic method for NB 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
    蛋白水解靶向嵌合体(PROTACs)代表了小分子疗法的新方向,其中目的蛋白(POI)的异双功能接头通过募集E3连接酶诱导其基于泛素化的蛋白水解。这里,我们显示电荷减少,天然质谱,和气相活化方法相结合,对PROTAC连接的三元络合物进行了深入分析。完整POI-PROTAC-VCB复合物(E3泛素连接酶的三聚体亚基)的电子捕获解离(ECD)促进POI解离。碰撞诱导解离(CID)导致非外周PROTAC的消除,产生完整的VCB-POI复合物,但未在溶液中看到,但与PROTAC诱导的蛋白质-蛋白质相互作用一致。此外,我们使用离子迁移谱(IMS)和碰撞激活来确定这种意外解离的来源。一起,证据表明,这种综合方法可用于筛选三元复合物形成和PROTAC-蛋白质接触,并可能报道PROTAC诱导的蛋白质相互作用,与PROTAC选择性和功效相关的特征。
    Proteolysis-targeting chimeras (PROTACs) represent a new direction in small-molecule therapeutics whereby a heterobifunctional linker to a protein of interest (POI) induces its ubiquitination-based proteolysis by recruiting an E3 ligase. Here, we show that charge reduction, native mass spectrometry, and gas-phase activation methods combine for an in-depth analysis of a PROTAC-linked ternary complex. Electron capture dissociation (ECD) of the intact POI-PROTAC-VCB complex (a trimeric subunit of an E3 ubiquitin ligase) promotes POI dissociation. Collision-induced dissociation (CID) causes elimination of the nonperipheral PROTAC, producing an intact VCB-POI complex not seen in solution but consistent with PROTAC-induced protein-protein interactions. In addition, we used ion mobility spectrometry (IMS) and collisional activation to identify the source of this unexpected dissociation. Together, the evidence shows that this integrated approach can be used to screen for ternary complex formation and PROTAC-protein contacts and may report on PROTAC-induced protein-protein interactions, a characteristic correlated with PROTAC selectivity and efficacy.
    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
    背景:尽管抗HER2抗体曲妥珠单抗可增加HER2+乳腺癌患者的生存率,相关数量的患者进展到这种治疗。在这种情况下,需要新的药物组合来增加其抗肿瘤活性。在这项工作中,我们评估了基于BET抑制剂(BETi)的蛋白水解靶向嵌合体(PROTAC)化合物在HER2+乳腺癌模型中增强曲妥珠单抗活性的功效.
    方法:使用BT474和SKBR3HER2+乳腺癌细胞系。曲妥珠单抗和BET-PROTACMZ1单独或联合使用的效果,使用MTT增殖试验进行评估,三维入侵和粘附培养,流式细胞术,qPCR和Western印迹。在小鼠的异种移植模型中进行体内研究。最后,应用Clariom_S_人类转录组阵列来鉴定处理后失调的基因。
    结果:与BETiJQ1相比,MZ1诱导了更高的抗增殖作用。MZ1和-曲妥珠单抗的组合显着降低细胞增殖,与任何一种单独的药物相比,三维结构的形成和细胞侵袭。细胞凋亡的评估导致联合治疗后细胞死亡的增加,生化研究显示细胞凋亡和DNA损伤成分的改变。单独或联合体内给药药物,小鼠原位异种移植的肿瘤,仅在MZ1-曲妥珠单抗联合治疗后导致肿瘤体积减小.转录组阵列的结果表明一系列新描述的转录因子,包括HOXB7,MEIS2,TCERG1和DNAJC2,这些转录因子与HER2乳腺癌亚型的不良结果相关,并被MZ1-曲妥珠单抗组合下调。
    结论:我们描述了一种新的活性组合,包括BET-PROTACMZ1和曲妥珠单抗,在HER2+肿瘤中。应进行进一步的研究以证实这些发现,并为其未来的临床发展铺平道路。
    BACKGROUND: Although the anti-HER2 antibody trastuzumab augments patient survival in HER2+ breast cancer, a relevant number of patients progress to this treatment. In this context, novel drug combinations are needed to increase its antitumor activity. In this work, we have evaluated the efficacy of proteolysis targeting chimera (PROTAC) compounds based on BET inhibitors (BETi) to augment the activity of trastuzumab in HER2+ breast cancer models.
    METHODS: BT474 and SKBR3 HER2+ breast cancer cell lines were used. The effects of trastuzumab and the BET-PROTAC MZ1 either alone or in combination, were evaluated using MTT proliferation assays, three-dimensional invasion and adhesion cultures, flow cytometry, qPCR and Western blot. In vivo studies were carried out in a xenografted model in mice. Finally, a Clariom_S_Human transcriptomic array was applied to identify deregulated genes after treatments.
    RESULTS: MZ1 induced a higher antiproliferative effect compared to the BETi JQ1. The combination of MZ1 and -trastuzumab significantly decreased cell proliferation, the formation of three-dimensional structures and cellular invasion compared to either of the drugs alone. Evaluation of apoptosis resulted in an increase of cell death following treatment with the combination, and biochemical studies displayed modifications of apoptosis and DNA damage components. In vivo administration of agents alone or combined, to tumors orthotopically xenografted in mice, resulted in a decrease of the tumor volume only after MZ1-Trastuzumab combination treatment. Results from a transcriptomic array indicated a series of newly described transcription factors including HOXB7, MEIS2, TCERG1, and DNAJC2, that were associated to poor outcome in HER2+ breast cancer subtype and downregulated by the MZ1-trastuzumab combination.
    CONCLUSIONS: We describe an active novel combination that includes the BET-PROTAC MZ1 and trastuzumab, in HER2+ tumors. Further studies should be performed to confirm these findings and pave the way for their future clinical development.
    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
    Bromo and extraterminal domain (BET) inhibitors-PROteolysis TArgeting Chimera (BETi-PROTAC) is a new family of compounds that induce proteasomal degradation through the ubiquitination of the tagged to BET inhibitors Bromodomain proteins, BRD2 and BRD. The encapsulation and controlled release of BET-PROTACs through their vectorization with antibodies, like trastuzumab, could facilitate their pharmacokinetic and efficacy profile. Antibody conjugated nanoparticles (ACNPs) using PROTACs have not been designed and evaluated. In this pioneer approach, the commercial MZ1 PROTAC was encapsulated into the FDA-approved polymeric nanoparticles. The nanoparticles were conjugated with trastuzumab to guide the delivery of MZ1 to breast tumoral cells that overexpress HER2. These ACNPs were characterized by means of size, polydispersity index, and Z-potential. Morphology of the nanoparticles, along with stability and release studies, completed the characterization. MZ1-loaded ACNPs showed a significant cytotoxic effect maintaining its mechanism of action and improving its therapeutic properties.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号