cancer mutations

癌症突变
  • 文章类型: Journal Article
    背景:确定体细胞突变的影响需要了解基因获得突变的功能关系;但是,功能相关基因的突变是如何相互影响的,这在很大程度上是未知的。
    方法:我们采用非同义对同义或dNdS比率来评估基因对的进化依赖性(ED),假设一个基因对的一个基因中的突变会影响其伴侣基因中突变作为突变背景的进化适应性。我们使用PanCancer和肿瘤类型特异性突变谱来推断基因对的ED,并评估了它们在基因依赖性和药物敏感性方面的生物学相关性。
    结果:我们建议基因对的dNdS比率及其衍生的cdNS(环境依赖性dNdS)评分作为ED区分基因对的测量,无论是协同(SYN)还是拮抗(ANT)。突变背景可以诱导配对基因中突变的进化适应性发生实质性变化,例如,IDH1和IDH2突变背景导致ATRXindel和IDH1错义突变的dNdS比率大幅增加和减少,对应于SYN和ANT与cdNS评分的正负关系,分别。基因沉默或敲除对细胞活力(遗传依赖性)的影响通常取决于ED,这表明ED可以指导合成致死性候选基因的选择,如TCF7L2-KRAS突变。使用基于细胞系的药物敏感性数据,靶向药物对细胞系的影响通常与具有靶基因的ED基因的突变有关,告知靶向抑制剂的药物致敏或耐药突变,例如,PRSS1和CTCF突变作为肺腺癌和黑色素瘤的EGFR和BRAF抑制剂的耐药突变,分别。
    结论:我们建议通过dNdS比率评估基因对的ED可以促进我们对具有潜在生物学和临床意义的基因功能关系的理解。
    BACKGROUND: Determining the impact of somatic mutations requires understanding the functional relationship of genes acquiring mutations; however, it is largely unknown how mutations in functionally related genes influence each other.
    METHODS: We employed non-synonymous-to-synonymous or dNdS ratios to evaluate the evolutionary dependency (ED) of gene pairs, assuming a mutation in one gene of a gene pair can affect the evolutionary fitness of mutations in its partner genes as mutation context. We employed PanCancer- and tumor type-specific mutational profiles to infer the ED of gene pairs and evaluated their biological relevance with respect to gene dependency and drug sensitivity.
    RESULTS: We propose that dNdS ratios of gene pairs and their derived cdNS (context-dependent dNdS) scores as measure of ED distinguishing gene pairs either as synergistic (SYN) or antagonistic (ANT). Mutation contexts can induce substantial changes in the evolutionary fitness of mutations in the paired genes, e.g., IDH1 and IDH2 mutation contexts lead to substantial increase and decrease of dNdS ratios of ATRX indels and IDH1 missense mutations corresponding to SYN and ANT relationship with positive and negative cdNS scores, respectively. The impact of gene silencing or knock-outs on cell viability (genetic dependencies) often depends on ED, suggesting that ED can guide the selection of candidates for synthetic lethality such as TCF7L2-KRAS mutations. Using cell line-based drug sensitivity data, the effects of targeted agents on cell lines are often associated with mutations of genes exhibiting ED with the target genes, informing drug sensitizing or resistant mutations for targeted inhibitors, e.g., PRSS1 and CTCF mutations as resistant mutations to EGFR and BRAF inhibitors for lung adenocarcinomas and melanomas, respectively.
    CONCLUSIONS: We propose that the ED of gene pairs evaluated by dNdS ratios can advance our understanding of the functional relationship of genes with potential biological and clinical implications.
    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
    癌症的发展涉及几种基本生物学途径中体细胞突变的积累。描绘肿瘤发生过程中通路突变的时间顺序对于理解癌症发展的生物学机制和确定治疗干预的潜在靶标至关重要。已经引入了几种计算和统计方法来基于来自患者队列的突变谱数据来估计体细胞突变的顺序。然而,当前方法的一个主要问题是它们没有考虑肿瘤内异质性(ITH),这限制了他们准确辨别通路突变顺序的能力。为了解决这个问题,我们提议PATOPAI,一种概率方法,通过整合ITH信息以及突变的途径和功能注释信息,在途径水平上估计突变的时间顺序。PATOPAI使用最大似然方法来估计特定序列中发生的通路突变事件的概率,其中它侧重于与肿瘤的系统发育结构一致的顺序。癌症基因组图谱(TCGA)对整个外显子组测序数据的应用说明了我们的方法能够恢复几种癌症类型中通路突变的时间顺序。
    The development of cancer involves the accumulation of somatic mutations in several essential biological pathways. Delineating the temporal order of pathway mutations during tumorigenesis is crucial for comprehending the biological mechanisms underlying cancer development and identifying potential targets for therapeutic intervention. Several computational and statistical methods have been introduced for estimating the order of somatic mutations based on mutation profile data from a cohort of patients. However, one major issue of current methods is that they do not take into account intra-tumor heterogeneity (ITH), which limits their ability to accurately discern the order of pathway mutations. To address this problem, we propose PATOPAI, a probabilistic approach to estimate the temporal order of mutations at the pathway level by incorporating ITH information as well as pathway and functional annotation information of mutations. PATOPAI uses a maximum likelihood approach to estimate the probability of pathway mutational events occurring in a specific sequence, wherein it focuses on the orders that are consistent with the phylogenetic structure of the tumors. Applications to whole exome sequencing data from The Cancer Genome Atlas (TCGA) illustrate our method\'s ability to recover the temporal order of pathway mutations in several cancer types.
    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
    κB-Ras(NF-κB抑制剂相互作用的Ras样蛋白)GTP酶是小的Ras样GTP酶,但在重要的序列基序中具有有趣的差异。它们以抑制肿瘤的方式作为Ral(Ras样)GTP酶和NF-κB信号的负调节因子,但对它们的功能模式知之甚少。这里,我们证明,与基于一级结构的预测相反,κB-RasGTP酶具有水解活性。结合低核苷酸亲和力,这使得它们在细胞中主要结合GTP的快速循环GTP酶。我们表征了在肿瘤中发生的κB-Ras突变的影响,并证明了核苷酸结合会影响κB-Ras的稳定性,但不是RalGAP(RalGTP酶激活蛋白)结合所必需的。这表明RalGAP/Ral信号传导的κB-Ras控制以核苷酸结合和开关非依赖性方式发生。
    κB-Ras (NF-κB inhibitor-interacting Ras-like protein) GTPases are small Ras-like GTPases but harbor interesting differences in important sequence motifs. They act in a tumor-suppressive manner as negative regulators of Ral (Ras-like) GTPase and NF-κB signaling, but little is known about their mode of function. Here, we demonstrate that, in contrast to predictions based on primary structure, κB-Ras GTPases possess hydrolytic activity. Combined with low nucleotide affinity, this renders them fast-cycling GTPases that are predominantly GTP-bound in cells. We characterize the impact of κB-Ras mutations occurring in tumors and demonstrate that nucleotide binding affects κB-Ras stability but is not strictly required for RalGAP (Ral GTPase-activating protein) binding. This demonstrates that κB-Ras control of RalGAP/Ral signaling occurs in a nucleotide-binding- and switch-independent fashion.
    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
    含有过早终止密码子的mRNA负责各种遗传疾病以及癌症。由于无义介导的mRNA衰变(NMD)途径,从这些异常mRNA合成的截短蛋白很少被检测到。这样的监测机制检测大多数这些异常的mRNA,并从mRNA池中迅速破坏它们。这里,我们实施了化学交联质谱(CLMS)技术来追踪NMD机制中由蛋白质-蛋白质相互作用(PPI)组成的新生物学.UPF2(无义转录物2的调节剂)之间的一组新颖的复杂网络,SMG1(丝氨酸/苏氨酸蛋白激酶SMG1),和来自NMD途径的SMG7被鉴定出来,其中UPF2被发现是SMG1和SMG7之间的连接桥。UPF2N端与SMG7形成了最多的相互作用,一组残基从MIF4G-I中出现,II,和III域与SMG1或SMG7对接。SMG1介导与UPF2初始残基的相互作用,而SMG7在该区域形成的相互作用很少。建模结构突出表明,UPF2和SMG1的PPI从定义明确的二级结构中出现,而SMG7从连接环中出现。比较癌症来源的突变对不同CLMS位点的影响揭示UPF2或SMG1的PPI中的变体具有显著的结构稳定性效应。我们的数据突出了SMG1,UPF2和SMG7基因的蛋白质-蛋白质界面,可用于潜在的治疗方法。阻断NMD途径可以增强新抗原或内部癌症疫苗的产生,这可以为设计潜在的基于肽的疫苗提供平台。
    mRNAs containing premature stop codons are responsible for various genetic diseases as well as cancers. The truncated proteins synthesized from these aberrant mRNAs are seldom detected due to the nonsense-mediated mRNA decay (NMD) pathway. Such a surveillance mechanism detects most of these aberrant mRNAs and rapidly destroys them from the pool of mRNAs. Here, we implemented chemical cross-linking mass spectrometry (CLMS) techniques to trace novel biology consisting of protein-protein interactions (PPIs) within the NMD machinery. A set of novel complex networks between UPF2 (Regulator of nonsense transcripts 2), SMG1 (Serine/threonine-protein kinase SMG1), and SMG7 from the NMD pathway were identified, among which UPF2 was found as a connection bridge between SMG1 and SMG7. The UPF2 N-terminal formed most interactions with SMG7, and a set of residues emerged from the MIF4G-I, II, and III domains docked with SMG1 or SMG7. SMG1 mediated interactions with initial residues of UPF2, whereas SMG7 formed very few interactions in this region. Modelled structures highlighted that PPIs for UPF2 and SMG1 emerged from the well-defined secondary structures, whereas SMG7 appeared from the connecting loops. Comparing the influence of cancer-derived mutations over different CLMS sites revealed that variants in the PPIs for UPF2 or SMG1 have significant structural stability effects. Our data highlights the protein-protein interface of the SMG1, UPF2, and SMG7 genes that can be used for potential therapeutic approaches. Blocking the NMD pathway could enhance the production of neoantigens or internal cancer vaccines, which could provide a platform to design potential peptide-based vaccines.
    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
    多溴,BRG1相关因子(PBAF)染色质重塑复合物亚基聚溴-1(PBRM1)包含六个溴结构域,可识别并结合组蛋白尾巴和其他核蛋白上的乙酰化赖氨酸残基。PBRM1溴结构域因此提供表观遗传翻译后修饰与PBAF调节染色质可及性和转录之间的联系。作为几种癌症的推定肿瘤抑制剂,PBRM1蛋白表达通常通过截短和缺失来消除。然而,癌症中33%的PBRM1突变是错义的,并聚集在其溴结构域内。此类突变可产生具有未确定的结构和功能特征的全长PBRM1变体蛋白。这里,我们采用了计算,生物物理,和细胞测定以询问PBRM1溴结构域错义变体对溴结构域稳定性和功能的影响。由于PBRM1的第四个溴结构域(PBRM1-BD4)中的突变占所有与癌症相关的PBRM1错义突变的近20%,我们的分析重点是PBRM1-BD4错义蛋白变体。根据高残基突变频率和/或保守性,选择16种潜在有害的PBRM1-BD4错义蛋白变体进行进一步研究。我们表明,与癌症相关的PBRM1-BD4错义变体表现出不同的溴结构域稳定性和结合乙酰化组蛋白的能力.我们的结果证明了识别单个PBRM1-BD4错义变体对蛋白质结构和功能的独特影响的有效性,基于溴结构域内受影响的残基位置。这些知识为绘制特定癌症相关的PBRM1错义变异与PBRM1功能的不同改变之间的相关性奠定了基础。告知未来癌症个性化医疗方法。
    The polybromo, brahma-related gene 1-associated factors (PBAF) chromatin remodeling complex subunit polybromo-1 (PBRM1) contains six bromodomains that recognize and bind acetylated lysine residues on histone tails and other nuclear proteins. PBRM1 bromodomains thus provide a link between epigenetic posttranslational modifications and PBAF modulation of chromatin accessibility and transcription. As a putative tumor suppressor in several cancers, PBRM1 protein expression is often abrogated by truncations and deletions. However, ∼33% of PBRM1 mutations in cancer are missense and cluster within its bromodomains. Such mutations may generate full-length PBRM1 variant proteins with undetermined structural and functional characteristics. Here, we employed computational, biophysical, and cellular assays to interrogate the effects of PBRM1 bromodomain missense variants on bromodomain stability and function. Since mutations in the fourth bromodomain of PBRM1 (PBRM1-BD4) comprise nearly 20% of all cancer-associated PBRM1 missense mutations, we focused our analysis on PBRM1-BD4 missense protein variants. Selecting 16 potentially deleterious PBRM1-BD4 missense protein variants for further study based on high residue mutational frequency and/or conservation, we show that cancer-associated PBRM1-BD4 missense variants exhibit varied bromodomain stability and ability to bind acetylated histones. Our results demonstrate the effectiveness of identifying the unique impacts of individual PBRM1-BD4 missense variants on protein structure and function, based on affected residue location within the bromodomain. This knowledge provides a foundation for drawing correlations between specific cancer-associated PBRM1 missense variants and distinct alterations in PBRM1 function, informing future cancer personalized medicine approaches.
    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
    TYK2是一种非受体酪氨酸激酶,Janus激酶(JAK)的成员,在几种疾病中起着核心作用,包括癌症.JAK催化结构域(KD)高度保守,然而,分离的TYK2-KD表现出独特的特异性。在以前的工作中,使用催化受损的TYK2-KD变体(P1104A)的分子动力学(MD)模拟,我们发现其JAK特征插入物(αFG)的这种氨基酸变化,在动力学层面采取行动。鉴于结构动力学是蛋白激酶变构激活的关键,在这项研究中,我们应用了一个长期的MD模拟和研究了活性TYK2-KD形式在腺苷5'-三磷酸和一个镁离子的存在,代表了催化循环的动态和关键步骤,在其他蛋白激酶中。MD轨迹的社区分析揭示了,第一次,关于激活过程中TYK2-KD内的动态轮廓和动态驱动的变构通信,并特别揭示了αFG和氨基酸P1104,P1105和I1112,发挥关键作用,并与提供KD内信号的氨基酸动态偶联通讯网络协同作用,以调节TYK2活性。证实我们的发现,大多数已鉴定的氨基酸与Tyk2基因的癌症相关错义/剪接位点突变有关.我们建议在催化循环的这一步的构象动力学,由αFG协调,作为TYK2独特底物识别的基础,并解释了其独特的特异性。总的来说,这项工作增加了对TYK2激活的深入理解的知识,并且可能对变构TYK2特异性抑制剂的合理设计有价值.
    TYK2 is a nonreceptor tyrosine kinase, member of the Janus kinases (JAK), with a central role in several diseases, including cancer. The JAKs\' catalytic domains (KD) are highly conserved, yet the isolated TYK2-KD exhibits unique specificities. In a previous work, using molecular dynamics (MD) simulations of a catalytically impaired TYK2-KD variant (P1104A) we found that this amino acid change of its JAK-characteristic insert (αFG), acts at the dynamics level. Given that structural dynamics is key to the allosteric activation of protein kinases, in this study we applied a long-scale MD simulation and investigated an active TYK2-KD form in the presence of adenosine 5\'-triphosphate and one magnesium ion that represents a dynamic and crucial step of the catalytic cycle, in other protein kinases. Community analysis of the MD trajectory shed light, for the first time, on the dynamic profile and dynamics-driven allosteric communications within the TYK2-KD during activation and revealed that αFG and amino acids P1104, P1105, and I1112 in particular, hold a pivotal role and act synergistically with a dynamically coupled communication network of amino acids serving intra-KD signaling for allosteric regulation of TYK2 activity. Corroborating our findings, most of the identified amino acids are associated with cancer-related missense/splice-site mutations of the Tyk2 gene. We propose that the conformational dynamics at this step of the catalytic cycle, coordinated by αFG, underlie TYK2-unique substrate recognition and account for its distinct specificity. In total, this work adds to knowledge towards an in-depth understanding of TYK2 activation and may be valuable towards a rational design of allosteric TYK2-specific inhibitors.
    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
    ROS1是人类基因组中最年夜的受体酪氨酸激酶。ROS1基因的重排导致致癌ROS1激酶融合蛋白,这是目前唯一有效的用于患者ROS1TKI靶向治疗的生物标志物。虽然癌症基因组中存在许多ROS1的体细胞错义突变,它们对催化活性和致病潜力的影响是未知的。我们询问了AACRGenie数据库并鉴定了ROS1酪氨酸激酶结构域中的34个错义突变用于进一步分析。我们的实验表明,这些突变对ROS1激酶功能有不同的影响,从完全损失到显著增加的催化活性。值得注意的是,在基于细胞的模型系统中,发现ROS1激酶结构域中Asp2113处的Asn和Gly取代是TKI敏感的致癌变体。体内实验表明,ROS1D2113N诱导的肿瘤形成对克唑替尼和氯拉替尼敏感,FDA批准的ROS1-TKIs。总的来说,这些发现强调了ROS1激酶结构域内特定点突变的致瘤潜力,以及它们作为FDA批准的ROS1-TKIs治疗靶点的潜力.
    ROS1 is the largest receptor tyrosine kinase in the human genome. Rearrangements of the ROS1 gene result in oncogenic ROS1 kinase fusion proteins that are currently the only validated biomarkers for targeted therapy with ROS1 TKIs in patients. While numerous somatic missense mutations in ROS1 exist in the cancer genome, their impact on catalytic activity and pathogenic potential is unknown. We interrogated the AACR Genie database and identified 34 missense mutations in the ROS1 tyrosine kinase domain for further analysis. Our experiments revealed that these mutations have varying effects on ROS1 kinase function, ranging from complete loss to significantly increased catalytic activity. Notably, Asn and Gly substitutions at Asp2113 in the ROS1 kinase domain were found to be TKI-sensitive oncogenic variants in cell-based model systems. In vivo experiments showed that ROS1 D2113N induced tumor formation that was sensitive to crizotinib and lorlatinib, FDA-approved ROS1-TKIs. Collectively, these findings highlight the tumorigenic potential of specific point mutations within the ROS1 kinase domain and their potential as therapeutic targets with FDA-approved ROS1-TKIs.
    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
    pre-mRNAs的剪接对真核生物中的基因调控和蛋白质组扩增至关重要。但是我们对剪接体组装过程中剪接位点的识别和配对的理解缺乏细节。这里,我们将多域RNA结合蛋白FUBP1鉴定为与迄今未知的顺式调节基序结合的关键剪接因子.通过收集核磁共振,结构,和体内相互作用数据,我们证明FUBP1稳定U2AF2和SF1,在3'剪接位点的关键组件,通过位于其无序区域内的多价结合界面。转录分析和动力学模型表明,FUBP1是长内含子有效剪接所必需的,在携带FUBP1突变的癌症患者中受损。值得注意的是,FUBP1与许多U1snRNP相关蛋白相互作用,提示FUBP1在长内含子剪接位点桥接中的独特作用。我们提出了一个令人信服的模型,用于长内含子的3'剪接位点识别,占所有人类内含子的80%。
    Splicing of pre-mRNAs critically contributes to gene regulation and proteome expansion in eukaryotes, but our understanding of the recognition and pairing of splice sites during spliceosome assembly lacks detail. Here, we identify the multidomain RNA-binding protein FUBP1 as a key splicing factor that binds to a hitherto unknown cis-regulatory motif. By collecting NMR, structural, and in vivo interaction data, we demonstrate that FUBP1 stabilizes U2AF2 and SF1, key components at the 3\' splice site, through multivalent binding interfaces located within its disordered regions. Transcriptional profiling and kinetic modeling reveal that FUBP1 is required for efficient splicing of long introns, which is impaired in cancer patients harboring FUBP1 mutations. Notably, FUBP1 interacts with numerous U1 snRNP-associated proteins, suggesting a unique role for FUBP1 in splice site bridging for long introns. We propose a compelling model for 3\' splice site recognition of long introns, which represent 80% of all human introns.
    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
    肿瘤抑制因子p53在癌症的发展中起着关键作用,因此近几十年来得到了广泛的研究。虽然众所周知p53作为四聚体具有生物活性,四聚机制仍未完全理解。p53在近50%的癌症中突变,突变可以改变蛋白质的寡聚状态,对蛋白质的生物学功能和细胞命运决定有影响。这里,我们描述了许多代表性的癌症相关突变对四聚结构域(TD)寡聚化的影响,定义了允许具有折叠和结构化结构域的肽长度,从而避免了侧翼区和N-和C-末端的净电荷的影响。已经在不同的实验条件下研究了这些肽。我们应用了各种技术,包括圆二色性(CD),天然质谱(MS)和高场溶液核磁共振。天然MS使我们能够检测复合物的天然状态,使肽复合物在气相中保持完整;通过NMR在溶液中分析二级和四级结构,低聚形式通过扩散NMR实验确定。对于所有研究的突变体,观察到显著的去稳定作用和可变的单体群体。
    Tumour suppressor p53 plays a key role in the development of cancer and has therefore been widely studied in recent decades. While it is well known that p53 is biologically active as a tetramer, the tetramerisation mechanism is still not completely understood. p53 is mutated in nearly 50% of cancers, and mutations can alter the oligomeric state of the protein, having an impact on the biological function of the protein and on cell fate decisions. Here, we describe the effects of a number of representative cancer-related mutations on tetramerisation domain (TD) oligomerisation defining a peptide length that permits having a folded and structured domain, thus avoiding the effect of the flanking regions and the net charges at the N- and C-terminus. These peptides have been studied under different experimental conditions. We have applied a variety of techniques, including circular dichroism (CD), native mass spectrometry (MS) and high-field solution NMR. Native MS allows us to detect the native state of complexes maintaining the peptide complexes intact in the gas phase; the secondary and quaternary structures were analysed in solution by NMR, and the oligomeric forms were assigned by diffusion NMR experiments. A significant destabilising effect and a variable monomer population were observed for all the mutants studied.
    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
    在这篇自传文章中,我回顾了我的瑞典背景。然后,我讨论了内源性DNA改变和碱基切除修复途径以及一些不寻常的DNA损伤的替代修复策略。内源性DNA损伤,如嘌呤碱基和胞嘧啶脱氨基的损失,被提议作为致癌突变的主要来源。
    In this autobiographical article, I reflect on my Swedish background. Then I discuss endogenous DNA alterations and the base excision repair pathway and alternative repair strategies for some unusual DNA lesions. Endogenous DNA damage, such as loss of purine bases and cytosine deamination, is proposed as a major source of cancer-causing mutations.
    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