vemurafenib

维罗非尼
  • 文章类型: Journal Article
    患有BRAF突变黑色素瘤的患者在使用有效的BRAF抑制剂(BRAFi)如维罗非尼治疗的仅7个月内就有肿瘤复发。已经证明,驱动BRAFi抗性的多种分子途径收敛于黑素瘤中c-Myc的激活。因此,我们通过将10号染色体上缺失的磷酸酶和张力蛋白同源物(PTEN)肿瘤抑制基因缺失和BRD4癌蛋白上调作为耐药黑色素瘤的Myc依赖性脆弱性,确定了一种新的组合治疗策略.作为有希望的治疗目标,我们决定同时递送PTEN质粒和BRD4靶向的Proteopoly-Targeting嵌合体(ARV),用于治疗BRAFi耐药黑色素瘤的“不可药物化”c-Myc.由于PTEN质粒和ARV的理化性质不同,我们制备了载有PTEN质粒的脂质纳米颗粒(PL-NANO)和载有ARV-825的纳米脂质体(AL-NANO),使每种治疗有效负载的平均粒径小于100nm,包封效率大于99%.PL-NANO和AL-NANO的组合在体外二维和三维模型中显示出协同的肿瘤生长抑制和大量凋亡。重要的是,PL-NANO和AL-NANO的同时递送实现了PTEN表达水平的显著上调和BRD4蛋白的降解,最终下调了BRAFi抗性黑色素瘤细胞中的c-Myc水平.总之,提供这种新型致命鸡尾酒的脂质纳米载体是一种针对BRAFi抗性黑色素瘤中不可药用的c-Myc癌基因的唯一基因疗法。
    Patients suffering from BRAF mutant melanoma have tumor recurrence within merely 7 months of treatment with a potent BRAF inhibitor (BRAFi) like vemurafenib. It has been proven that diverse molecular pathways driving BRAFi resistance converge to activation of c-Myc in melanoma. Therefore, we identified a novel combinatorial therapeutic strategy by targeting loss of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) tumor suppressor gene and upregulated BRD4 oncoprotein as Myc-dependent vulnerabilities of drug-resistant melanoma. Being promising therapeutic targets, we decided to concomitantly deliver PTEN plasmid and BRD4 targeted PROteolysis-TArgeting Chimera (ARV) to drug the \"undruggable\" c-Myc in BRAFi-resistant melanoma. Since PTEN plasmid and ARV are distinct in their physicochemical properties, we fabricated PTEN-plasmid loaded lipid nanoparticles (PL-NANO) and ARV-825-loaded nanoliposomes (AL-NANO) to yield a mean particle size of less than 100 nm and greater than 99% encapsulation efficiency for each therapeutic payload. Combination of PL-NANO and AL-NANO displayed synergistic tumor growth inhibition and substantial apoptosis in in vitro two-dimensional and three-dimensional models. Importantly, simultaneous delivery of PL-NANO and AL-NANO achieved significant upregulation of PTEN expression levels and degradation of BRD4 protein to ultimately downregulate c-Myc levels in BRAFi-resistant melanoma cells. Altogether, lipid nanocarriers delivering this novel lethal cocktail stands as one-of-a-kind gene therapy to target undruggable c-Myc oncogene in BRAFi-resistant melanoma.
    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
    基于新抗原的免疫疗法是治疗先前顽固性肿瘤的有吸引力的潜在治疗方法。为了有效地拓宽这种方法的应用,严格的生物标志物对于识别有反应的患者至关重要.ARID1A,SWI/SNF染色质重塑复合物的一个经常突变的亚基,据报道,在一些队列中可以确定肿瘤的免疫原性;然而,ARID1A的突变和缺失并不总是与免疫治疗的临床反应相关.在这项研究中,我们基于ARID1A状态调查了靶向治疗耐药癌症的免疫治疗应答.将有或没有ARID1A表达的小鼠和人BRAFV600E黑色素瘤转化为对vemurafenib的抗性,FDA批准的特异性BRAFV600E抑制剂。抗PD-1抗体治疗增强了对vemurafenib耐药的ARID1A缺陷型肿瘤的抗肿瘤免疫反应,但在ARID1A完整肿瘤或对vemurafenib敏感的ARID1A缺陷型肿瘤中却没有。来自维罗非尼耐药期间累积的体细胞突变的新抗原在ARID1A缺陷型肿瘤中高表达并促进肿瘤免疫原性。此外,新产生的新抗原可作为疫苗的免疫治疗靶点。最后,在缺乏ARID1A的实验性人黑色素瘤细胞中,靶向治疗耐药特异性新抗原被证实可引发T细胞受体应答.总的来说,基于vemurafenib耐药的ARID1A突变肿瘤的分类作为免疫治疗反应的额外指标,将能够更准确地预测以指导癌症治疗.此外,伴随治疗抗性出现的新抗原可能是难治性肿瘤的有希望的治疗靶点.意义:化疗抗性促进ARID1A缺陷型肿瘤中免疫原性新抗原的获得,赋予对免疫检查点阻断的敏感性,并可用于开发抗肿瘤疫苗。提供改善免疫治疗效果的策略。
    Neoantigen-based immunotherapy is an attractive potential treatment for previously intractable tumors. To effectively broaden the application of this approach, stringent biomarkers are crucial to identify responsive patients. ARID1A, a frequently mutated subunit of SWI/SNF chromatin remodeling complex, has been reported to determine tumor immunogenicity in some cohorts; however, mutations and deletions of ARID1A are not always linked to clinical responses to immunotherapy. In this study, we investigated immunotherapeutic responses based on ARID1A status in targeted therapy-resistant cancers. Mouse and human BRAFV600E melanomas with or without ARID1A expression were transformed into resistant to vemurafenib, an FDA-approved specific BRAFV600E inhibitor. Anti-PD-1 antibody treatment enhanced antitumor immune responses in vemurafenib-resistant ARID1A-deficient tumors but not in ARID1A-intact tumors or vemurafenib-sensitive ARID1A-deficient tumors. Neoantigens derived from accumulated somatic mutations during vemurafenib resistance were highly expressed in ARID1A-deficient tumors and promoted tumor immunogenicity. Furthermore, the newly generated neoantigens could be utilized as immunotherapeutic targets by vaccines. Finally, targeted therapy resistance-specific neoantigen in experimental human melanoma cells lacking ARID1A were validated to elicit T-cell receptor responses. Collectively, the classification of ARID1A-mutated tumors based on vemurafenib resistance as an additional indicator of immunotherapy response will enable a more accurate prediction to guide cancer treatment. Furthermore, the neoantigens that emerge with therapy resistance can be promising therapeutic targets for refractory tumors. Significance: Chemotherapy resistance promotes the acquisition of immunogenic neoantigens in ARID1A-deficient tumors that confer sensitivity to immune checkpoint blockade and can be utilized for developing antitumor vaccines, providing strategies to improve immunotherapy efficacy.
    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:朗格汉斯细胞组织细胞增生症(LCH)是一种具有炎性成分的克隆性髓样肿瘤。难治性疾病是一个挑战,但是vemurafenib已经成为一种治疗选择。我们将描述两名巴西儿童患有难治性LCH的病例,对vemurafenib反应积极。
    方法:两例病例均诊断为多系统疾病,受累于危险器官,对标准和二线治疗无反应。在对经典治疗方案无效后,对BRAF突变进行了调查,发现这两名患者均为阳性,并寻求vemurafenib的靶向治疗。第一个案例已经使用vemurafenib大约2年,第二个案例已经使用了大约3年,在合并使用维持治疗后尝试暂停药物治疗。然而,停药4个月后病情复发。幸运的是,再次使用药物后,疾病恢复到缓解状态.
    结论:这些病例是巴西首次报道的使用非标签威罗菲尼治疗LCH的案例,并且两名患者均表现出对药物的良好反应。然而,儿童的长期副作用是未知的,和前瞻性研究是必要的。此外,巴西缺乏有关组织细胞增生症的流行病学数据,并且有研究评估了将BRAF突变研究和使用vemurafenib纳入公共卫生系统的预算影响.这些报告可能是一个起点。
    BACKGROUND: Langerhans cell histiocytosis (LCH) is a clonal myeloid neoplasm with inflammatory component. Refractory disease is a challenge, but vemurafenib has emerged as a therapeutic option. We will delineate the cases of two Brazilian children suffering from refractory LCH with a positive response to vemurafenib.
    METHODS: Both cases had a diagnosis of multisystem disease with involvement of organs at risk and had not responded to standard and second-line treatment. After refractoriness to classic treatment regimens, the BRAF mutation was investigated and found to be positive in both patients, and target therapy with vemurafenib was sought. The first case has been using vemurafenib for about 2 years and the second case has been using it for about 3 years, having had an attempt to suspend the medication after concomitant use with maintenance therapy. However, the disease returned 4 months after stopping the medication. Fortunately, the disease returned to remission status after the medication was reintroduced.
    CONCLUSIONS: These cases represent the first reported instances of off-label vemurafenib use in Brazil for the treatment of LCH and both patients have demonstrated excellent responses to the medication. However, the long-term side effects are unknown in children, and prospective studies are needed. In addition, there is a lack of epidemiological data on histiocytosis in Brazil and studies evaluating the budgetary impact of incorporating BRAF mutation research and the use of vemurafenib into the public health system. These reports could be a starting point.
    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:BRAF(v-raf鼠肉瘤病毒癌基因同源物B1)/MEK(丝裂原活化蛋白激酶激酶)抑制剂用于黑素瘤治疗。不幸的是,用这种联合疗法治疗的患者很快就会对治疗产生抗药性,但是这种现象背后的机制尚未完全理解。这里,我们报告并鉴定了两种对BRAF(vemurafenib)和MEK(cobimetinib)抑制剂耐药的黑色素瘤细胞系(WM9和Hs294T).
    方法:通过XTT测试评估细胞活力。使用蛋白质印迹评估所选蛋白质的水平以及信号传导途径的激活。通过RT-PCR评估所选基因的表达。通过流式细胞术分析细胞周期阶段的分布,共聚焦显微镜用于拍摄球体的照片。使用人细胞因子阵列测定细胞分泌的细胞因子的组成。
    结果:在BRAF/MEK抑制剂存在下,耐药细胞的存活和ERK激酶的激活增加。这些细胞的IC50值比对照高1000倍以上。抗性细胞也表现出升高的AKT活化,p38和JNK信号通路与EGFR表达增加,ErbB2MET,和PDGFRβ受体以及ErbB3受体的表达降低。此外,这些细胞显示出编码参与药物转运和代谢的蛋白质的基因表达增加。抗性细胞还表现出上皮-间质转化和癌症干细胞的特征以及降低的增殖率和升高的细胞因子分泌。
    结论:总之,这项工作描述了BRAF/MEK抑制剂抗性黑色素瘤细胞,以便更好地理解抵抗的潜在机制。因此,结果可能有助于开发新的,更有效的治疗策略。
    BACKGROUND: BRAF (v-raf murine sarcoma viral oncogene homolog B1)/MEK (mitogen-activated protein kinase kinase) inhibitors are used for melanoma treatment. Unfortunately, patients treated with this combined therapy develop resistance to treatment quite quickly, but the mechanisms underlying this phenomenon are not yet fully understood. Here, we report and characterize two melanoma cell lines (WM9 and Hs294T) resistant to BRAF (vemurafenib) and MEK (cobimetinib) inhibitors.
    METHODS: Cell viability was assessed via the XTT test. The level of selected proteins as well as activation of signaling pathways were evaluated using Western blotting. The expression of the chosen genes was assessed by RT-PCR. The distribution of cell cycle phases was analyzed by flow cytometry, and confocal microscopy was used to take photos of spheroids. The composition of cytokines secreted by cells was determined using a human cytokine array.
    RESULTS: The resistant cells had increased survival and activation of ERK kinase in the presence of BRAF/MEK inhibitors. The IC50 values for these cells were over 1000 times higher than for controls. Resistant cells also exhibited elevated activation of AKT, p38, and JNK signaling pathways with increased expression of EGFR, ErbB2, MET, and PDGFRβ receptors as well as reduced expression of ErbB3 receptor. Furthermore, these cells demonstrated increased expression of genes encoding proteins involved in drug transport and metabolism. Resistant cells also exhibited features of epithelial-mesenchymal transition and cancer stem cells as well as reduced proliferation rate and elevated cytokine secretion.
    CONCLUSIONS: In summary, this work describes BRAF/MEK-inhibitor-resistant melanoma cells, allowing for better understanding the underlying mechanisms of resistance. The results may thus contribute to the development of new, more effective therapeutic strategies.
    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
    转移性黑色素瘤,一种致命的皮肤癌,经常对BRAF抑制剂药物vemurafenib产生耐药性,强调需要了解潜在的耐药机制,并探索针对整合素和TGF-β信号传导的潜在治疗策略。在这项研究中,研究了整合素和TGF-β信号在黑色素瘤中vemurafenib耐药中的作用,并研究了维罗非尼与西伦吉肽联合作为治疗策略的潜力。在这项研究中,发现PAI1和p21的转录是由获得性维罗非尼抗性诱导的,和ITGA5水平由于这种抗性而增加。在vemurafenib抗性的发展中,ITGA5的转录由TGF-β途径介导。维罗非尼和西仑吉肽的联合治疗观察到对维罗非尼耐药的黑色素瘤细胞增殖的协同作用。此外,这种联合疗法显著降低了这些耐药细胞的侵袭和集落形成.总之,建议靶向整合素和TGF-β信号,特别是ITGA5、ITGB3、PAI1和P21,可能提供有希望的方法来克服维罗非尼耐药性,从而改善转移性黑色素瘤患者的预后。
    Metastatic melanoma, a deadly form of skin cancer, often develops resistance to the BRAF inhibitor drug vemurafenib, highlighting the need for understanding the underlying mechanisms of resistance and exploring potential therapeutic strategies targeting integrins and TGF-β signalling. In this study, the role of integrins and TGF-β signalling in vemurafenib resistance in melanoma was investigated, and the potential of combining vemurafenib with cilengitide as a therapeutic strategy was investigated. In this study, it was found that the transcription of PAI1 and p21 was induced by acquired vemurafenib resistance, and ITGA5 levels were increased as a result of this resistance. The transcription of ITGA5 was mediated by the TGF-β pathway in the development of vemurafenib resistance. A synergistic effect on the proliferation of vemurafenib-resistant melanoma cells was observed with the combination therapy of vemurafenib and cilengitide. Additionally, this combination therapy significantly decreased invasion and colony formation in these resistant cells. In conclusion, it is suggested that targeting integrins and TGF-β signalling, specifically ITGA5, ITGB3, PAI1, and p21, may offer promising approaches to overcoming vemurafenib resistance, thereby improving outcomes for metastatic melanoma 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
    恶性黑色素瘤,侵袭性皮肤癌预后差,通常特征是BRAFV600E突变导致MAPK通路的激活以及黑素细胞的增殖和存活。BRAFV600E抑制剂,如vemurafenib和dabrafenib提高了患者的生存率,然而,耐药性仍然是一个重大挑战。我们研究了ERK5途径在BRAFV600E黑色素瘤细胞和对PLX4720(vemurafenib)和dabrafenib具有获得性抗性的细胞中的作用。在BRAFV600E黑色素瘤中,与ERK1/2抑制相比,ERK5抑制最低限度地影响生存力。在维罗非尼耐药细胞中,单独抑制ERK5不会影响生存力或恢复对维罗非尼的药物敏感性。然而,在dabrafenib抗性细胞中,ERK5抑制降低了活力并增强了MEK1/2抑制的抗增殖作用。靶向ERK5途径可能代表达拉非尼耐药黑色素瘤的治疗机会。
    Malignant melanoma, an aggressive skin cancer with a poor prognosis, frequently features BRAFV600E mutation resulting in activation of the MAPK pathway and melanocyte proliferation and survival. BRAFV600E inhibitors like vemurafenib and dabrafenib have enhanced patient survival, yet drug resistance remains a significant challenge. We investigated the role of the ERK5 pathway in BRAFV600E melanoma cells and cells with acquired resistance to PLX4720 (vemurafenib) and dabrafenib. In BRAFV600E melanoma, ERK5 inhibition minimally affected viability compared to ERK1/2 inhibition. In vemurafenib-resistant cells, ERK5 inhibition alone didn\'t impact viability or restore drug sensitivity to vemurafenib. However, in dabrafenib-resistant cells, ERK5 inhibition reduced viability and enhanced the anti-proliferative effect of MEK1/2 inhibition. Targeting the ERK5 pathway may represent a therapeutic opportunity in dabrafenib-resistant melanoma.
    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:BRAF抑制剂广泛用于治疗具有BRAFV600E突变的黑素瘤。然而,耐药性的发展损害了它们的治疗功效。在BRAF抑制剂抗性黑色素瘤中发现了不同的基因组和转录组改变,迫切需要收敛,具有不同耐药机制的逆转治疗耐药肿瘤的药物靶标。
    方法:进行CRISPR-Cas9筛选以鉴定新的靶基因,其抑制选择性靶向A375VR,对vemurafenib具有获得性抗性的BRAFV600E突变细胞系。各种体外和体内试验,包括细胞竞争试验,水溶性四唑(WST)测定,进行活死试验和异种移植试验以确认协同细胞死亡.液相色谱-质谱分析定量多胺生物合成和维罗非尼耐药黑色素瘤蛋白质组的变化。EIF5A催眠依赖的蛋白质翻译和随后的线粒体生物发生和活性的变化通过O-炔丙基-嘌呤霉素标记测定,mitotracker,mitoSOX标记和海马测定。生物信息学分析用于确定多胺生物合成与BRAF抑制剂抗性和黑色素瘤患者队列中不良预后的关联。
    结果:我们阐明了多胺生物合成及其调节机制在促进BRAF抑制剂抗性中的作用。利用CRISPR-Cas9屏幕,我们鉴定了AMD1(S-腺苷甲硫氨酸脱羧酶1),多胺生物合成的关键酶,作为一种药物靶标,其抑制作用降低了维罗非尼的耐药性。代谢组学和蛋白质组学分析显示,维罗非尼耐药癌症中多胺生物合成上调,导致增强的EIF5A催眠,线粒体蛋白的翻译和氧化磷酸化。我们还确定维罗非尼耐药癌症中持续的c-Myc水平是多胺生物合成升高的原因。在体外细胞系模型和在异种移植模型中体内抑制多胺生物合成或c-Myc逆转维罗非尼抗性。多胺生物合成特征与黑色素瘤患者BRAF/MAPK抑制剂治疗后的不良预后和较短的无进展生存期相关,强调我们的研究结果的临床相关性。
    结论:我们的发现描述了在黑色素瘤中涉及多胺-EIF5A催眠-线粒体呼吸途径赋予BRAF抑制剂抗性的分子机制。这些靶标将作为有效的治疗靶标,其可以最大化现有BRAF抑制剂的治疗功效。
    BACKGROUND: BRAF inhibitors are widely employed in the treatment of melanoma with the BRAF V600E mutation. However, the development of resistance compromises their therapeutic efficacy. Diverse genomic and transcriptomic alterations are found in BRAF inhibitor resistant melanoma, posing a pressing need for convergent, druggable target that reverse therapy resistant tumor with different resistance mechanisms.
    METHODS: CRISPR-Cas9 screens were performed to identify novel target gene whose inhibition selectively targets A375VR, a BRAF V600E mutant cell line with acquired resistance to vemurafenib. Various in vitro and in vivo assays, including cell competition assay, water soluble tetrazolium (WST) assay, live-dead assay and xenograft assay were performed to confirm synergistic cell death. Liquid Chromatography-Mass Spectrometry analyses quantified polyamine biosynthesis and changes in proteome in vemurafenib resistant melanoma. EIF5A hypusination dependent protein translation and subsequent changes in mitochondrial biogenesis and activity were assayed by O-propargyl-puromycin labeling assay, mitotracker, mitoSOX labeling and seahorse assay. Bioinformatics analyses were used to identify the association of polyamine biosynthesis with BRAF inhibitor resistance and poor prognosis in melanoma patient cohorts.
    RESULTS: We elucidate the role of polyamine biosynthesis and its regulatory mechanisms in promoting BRAF inhibitor resistance. Leveraging CRISPR-Cas9 screens, we identify AMD1 (S-adenosylmethionine decarboxylase 1), a critical enzyme for polyamine biosynthesis, as a druggable target whose inhibition reduces vemurafenib resistance. Metabolomic and proteomic analyses reveal that polyamine biosynthesis is upregulated in vemurafenib-resistant cancer, resulting in enhanced EIF5A hypusination, translation of mitochondrial proteins and oxidative phosphorylation. We also identify that sustained c-Myc levels in vemurafenib-resistant cancer are responsible for elevated polyamine biosynthesis. Inhibition of polyamine biosynthesis or c-Myc reversed vemurafenib resistance both in vitro cell line models and in vivo in a xenograft model. Polyamine biosynthesis signature is associated with poor prognosis and shorter progression free survival after BRAF/MAPK inhibitor treatment in melanoma cohorts, highlighting the clinical relevance of our findings.
    CONCLUSIONS: Our findings delineate the molecular mechanisms involving polyamine-EIF5A hypusination-mitochondrial respiration pathway conferring BRAF inhibitor resistance in melanoma. These targets will serve as effective therapeutic targets that can maximize the therapeutic efficacy of existing BRAF inhibitors.
    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
    源自邻近的v-raf鼠肉瘤病毒癌基因同源物B1抑制剂(BRAFi)抗性黑色素瘤细胞的外泌体介导对BRAFi敏感的黑色素瘤细胞中耐药性的形成。外泌体miRNA在黑色素瘤BRAFi耐药中的功能和分子机制尚未研究。我们发明miR-19a在BRAFi抗性黑色素瘤细胞中的表达显著高于敏感细胞,miR-19a通过靶向免疫球蛋白样结构域蛋白1(LRIG1)促进黑色素瘤细胞对BRAFi的抗性。miR-19a在BRAFi抗性黑色素瘤细胞分泌的外泌体中高度富集,而这些外泌体miR-19a促进BRAFi抗性的传播。蛋白激酶B(AKT)和丝裂原活化蛋白激酶(MAPK)通路的再激活是黑色素瘤细胞BRAFi抵抗的主要原因。我们证明,来自黑色素瘤细胞的外泌体miR-19a通过靶向LRIG1重新激活AKT和MAPK通路促进黑色素瘤中BRAFi抗性的形成和传播。因此,miR-19a可作为获得性耐药黑色素瘤患者的潜在治疗靶点。
    Exosomes derived from neighboring v-raf murine sarcoma viral oncogene homolog B1 inhibitor (BRAFi)-resistant melanoma cells mediate the formation of resistance in melanoma cells sensitive to BRAFi. The function and molecular mechanisms of exosomal miRNA in BRAFi resistance of melanoma have not been studied. We found that the expression of miR-19a in BRAFi resistant melanoma cells was significantly higher than that in sensitive cells, and miR-19a contributes to the resistance of melanoma cells to BRAFi by targeting immunoglobulin-like domains protein 1 (LRIG1). miR-19a was highly enriched in exosomes secreted from BRAFi resistant melanoma cells, and these exosomal miR-19a promote the spread of BRAFi resistant. The reactivation of Protein kinase B (AKT) and mitogen-activated protein kinase (MAPK) pathways is the main reason for the BRAFi resistant of melanoma cells. We demonstrated that exosomal miR-19a derived from melanoma cell promotes the formation and spread of BRAFi resistant in melanoma through targeting LRIG1 to reactivate AKT and MAPK pathway. Therefore, miR-19a may serve as a potential therapeutic target in melanoma patients with acquired 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
    Raf激酶在调节细胞增殖的信号转导中起关键作用,分化,和生存。尽管对Raf激酶相对于其他胞质蛋白的功能和动力学进行了数十年的研究,理解Raf激酶受限于其在原子分辨率下缺乏全长结构。这里,我们介绍了从人工智能/机器学习(AI/ML)算法获得的全长CRaf激酶的第一个模型,该模型具有通过大规模温度复制交换模拟模拟的结构的收敛集合。通过将模拟结构与最新的Cryo-EM结构进行比较来验证我们的模型,该结构详细说明了CRaf的三个关键域和区域之间的密切接触。我们的模拟揭示了CRaf分子内相互作用的潜在新表位,并揭示了CRaf激酶的动力学性质。其中三个域可以相对于彼此来回移动,以实现监管动态。然后将动态构象用于对接算法中,以与悖论破坏者PLX7904相比,深入了解Vemurafenib引起的悖论效应。我们基于全长CRaf的动力学,提出了Raf-异二聚体/KRas-二聚体的模型作为信号体。
    Raf kinases play key roles in signal transduction in cells for regulating proliferation, differentiation, and survival. Despite decades of research into functions and dynamics of Raf kinases with respect to other cytosolic proteins, understanding Raf kinases is limited by the lack of their full-length structures at the atomic resolution. Here, we present the first model of the full-length CRaf kinase obtained from artificial intelligence/machine learning algorithms with a converging ensemble of structures simulated by large-scale temperature replica exchange simulations. Our model is validated by comparing simulated structures with the latest cryo-EM structure detailing close contacts among three key domains and regions of the CRaf. Our simulations identify potentially new epitopes of intramolecule interactions within the CRaf and reveal a dynamical nature of CRaf kinases, in which the three domains can move back and forth relative to each other for regulatory dynamics. The dynamic conformations are then used in a docking algorithm to shed insight into the paradoxical effect caused by vemurafenib in comparison with a paradox breaker PLX7904. We propose a model of Raf-heterodimer/KRas-dimer as a signalosome based on the dynamics of the full-length CRaf.
    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
    黑色素瘤细胞表达高水平的CD73,其产生细胞外免疫抑制性腺苷。CD73表达的变化是响应肿瘤环境因素而发生的,有助于肿瘤表型可塑性和治疗抗性。以前,我们观察到,CD73在黑素瘤细胞表面的表达可在营养应激下上调.这里,我们探讨了黑素瘤细胞在低营养可获得性下释放可溶性CD73的机制,以及这是否可能受到靶向原癌基因B-Raf(BRAF)的药物的影响.我们发现饥饿的黑色素瘤细胞可以释放高水平的CD73,能够将AMP转化为腺苷,这种活性被选择性CD73抑制剂废除,APCP或PSB-12489。CD73从黑素瘤细胞的释放由基质金属蛋白酶MMP-9介导。的确,MMP-9抑制剂显着降低从细胞释放的CD73的水平,而其表面水平增加。相关的,黑色素瘤细胞,带有激活的BRAF突变,用达布拉非尼或维罗非尼治疗后,显示CD73细胞表达的强烈减少和释放到细胞外空间中的CD73的水平降低。相反,对dabrafenib具有抗性的黑色素瘤细胞显示出膜结合的CD73和释放到培养基中的可溶性CD73的高表达。总之,我们的数据表明CD73从黑色素瘤细胞中释放.CD73的表达与对BRAF抑制剂的反应相关。发展对dabrafenib抗性的黑色素瘤细胞显示CD73表达增加,包括从细胞释放的可溶性CD73,表明CD73参与获得对治疗的抵抗。
    Melanoma cells express high levels of CD73 that produce extracellular immunosuppressive adenosine. Changes in the CD73 expression occur in response to tumor environmental factors, contributing to tumor phenotype plasticity and therapeutic resistance. Previously, we have observed that CD73 expression can be up-regulated on the surface of melanoma cells in response to nutritional stress. Here, we explore the mechanism by which melanoma cells release soluble CD73 under low nutrient availability and whether this might be affected by agents targeting the proto-oncogene B-Raf (BRAF). We found that starved melanoma cells can release high levels of CD73, able to convert AMP into adenosine, and this activity is abrogated by selective CD73 inhibitors, APCP or PSB-12489. The release of CD73 from melanoma cells is mediated by the matrix metalloproteinase MMP-9. Indeed, MMP-9 inhibitors significantly reduce the levels of CD73 released from the cells, while its surface levels increase. Of relevance, melanoma cells, harboring an activating BRAF mutation, upon treatment with dabrafenib or vemurafenib, show a strong reduction of CD73 cell expression and reduced levels of CD73 released into the extracellular space. Conversely, melanoma cells resistant to dabrafenib show high expression of membrane-bound CD73 and soluble CD73 released into the culture medium. In summary, our data indicate that CD73 is released from melanoma cells. The expression of CD73 is associated with response to BRAF inhibitors. Melanoma cells developing resistance to dabrafenib show increased expression of CD73, including soluble CD73 released from cells, suggesting that CD73 is involved in acquiring resistance to treatment.
    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