Cancer biology

癌症生物学
  • 文章类型: Journal Article
    上皮-间质转化(EMT)对化疗耐药有显著影响,仍然是治疗晚期乳腺癌的关键挑战。EMT的复杂性,涉及冗余的pro-EMT信号通路及其悖论逆转过程,间充质到上皮转化(MET),阻碍了有效治疗方法的发展。在这项研究中,我们利用小鼠Tri-PyMTEMT谱系追踪模型和单细胞RNA测序(scRNA-seq)全面分析肿瘤细胞的EMT状态.我们的发现表明,在EMT和MET过程的过渡阶段,核糖体生物发生(RiBi)升高。由ERK和mTOR信号介导的RiBi及其随后的新生蛋白质合成对于EMT/MET完成至关重要。重要的是,抑制过量的RiBi在遗传或药理学上损害了肿瘤细胞的EMT/MET能力。RiBi抑制与化疗药物联合协同降低化疗下上皮和间质肿瘤细胞的转移生长。我们的研究表明,靶向RiBi途径是治疗晚期乳腺癌患者的有希望的策略。
    尽管多年来在乳腺癌治疗方面取得了相当大的进步,仍然有许多患者的癌细胞对治疗产生抗药性,包括化疗.几种不同的因素可能导致化疗耐药,但一个重要的变化是上皮-间质转化(或简称EMT)。在这个转变过程中,乳腺癌细胞变得更具侵略性,更能够转移并扩散到身体的其他部位。细胞也可以经历称为间充质到上皮转化(或简称MET)的反向过程。一起,EMT和MET帮助乳腺癌细胞变得适应治疗。然而,目前尚不清楚这些转变是否有共同的机制或途径,可以作为一种使癌症治疗更有效的方法。为了调查,Ban,Zou等人。研究了用荧光蛋白标记的小鼠的乳腺癌细胞,这些荧光蛋白表明细胞是否曾经在上皮和间质状态之间转变。各种基因实验表明,处于EMT或MET阶段的乳腺癌细胞产生了更多的核糖体,对生产新蛋白质至关重要的分子。Ban,周等人。发现阻断核糖体的产生(使用药物或遗传工具)阻止了细胞经历EMT和MET。进一步的实验表明,当患有乳腺癌的小鼠接受标准化疗治疗以及抗核糖体药物治疗时,这减少了转移到肺部的肿瘤的数量和大小.这表明阻断核糖体的产生使得经历EMT和/或MET的乳腺癌细胞对化疗的抗性较小。未来的研究将必须确定这些发现是否也适用于乳腺癌患者。特别是,这项研究中用于阻断核糖体产生的药物之一处于早期临床试验阶段,因此,未来的试验可能能够评估药物联合化疗的效果。
    Epithelial-to-mesenchymal transition (EMT) contributes significantly to chemotherapy resistance and remains a critical challenge in treating advanced breast cancer. The complexity of EMT, involving redundant pro-EMT signaling pathways and its paradox reversal process, mesenchymal-to-epithelial transition (MET), has hindered the development of effective treatments. In this study, we utilized a Tri-PyMT EMT lineage-tracing model in mice and single-cell RNA sequencing (scRNA-seq) to comprehensively analyze the EMT status of tumor cells. Our findings revealed elevated ribosome biogenesis (RiBi) during the transitioning phases of both EMT and MET processes. RiBi and its subsequent nascent protein synthesis mediated by ERK and mTOR signalings are essential for EMT/MET completion. Importantly, inhibiting excessive RiBi genetically or pharmacologically impaired the EMT/MET capability of tumor cells. Combining RiBi inhibition with chemotherapy drugs synergistically reduced metastatic outgrowth of epithelial and mesenchymal tumor cells under chemotherapies. Our study suggests that targeting the RiBi pathway presents a promising strategy for treating patients with advanced breast cancer.
    Although there have been considerable improvements in breast cancer treatments over the years, there are still many patients whose cancerous cells become resistant to treatments, including chemotherapy. Several different factors can contribute to resistance to chemotherapy, but one important change is the epithelial-to-mesenchymal transition (or EMT for short). During this transition, breast cancer cells become more aggressive, and more able to metastasize and spread to other parts of the body. Cells can also go through the reverse process called the mesenchymal-to-epithelial transition (or MET for short). Together, EMT and MET help breast cancer cells become resilient to treatment. However, it was not clear if these transitions shared a mechanism or pathway that could be targeted as a way to make cancer treatments more effective. To investigate, Ban, Zou et al. studied breast cancer cells from mice which had been labelled with fluorescent proteins that indicated whether a cell had ever transitioned between an epithelial and mesenchymal state. Various genetic experiments revealed that breast cancer cells in the EMT or MET phase made a lot more ribosomes, molecules that are vital for producing new proteins. Ban, Zhou et al. found that blocking the production of ribosomes (using drugs or genetic tools) prevented the cells from undergoing both EMT and MET. Further experiments showed that when mice with breast cancer were treated with a standard chemotherapy treatment plus an anti-ribosome drug, this reduced the number and size of tumors that had metastasized to the lung. This suggests that blocking ribosome production makes breast cancer cells undergoing EMT and/or MET less resistant to chemotherapy. Future studies will have to ascertain whether these findings also apply to patients with breast cancer. In particular, one of the drugs used to block ribosome production in this study is in early-phase clinical trials, so future trials may be able to assess the drug’s effect in combination with chemotherapies.
    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T细胞急性淋巴细胞白血病(T-ALL)中,超过50%的病例显示Notch1信号自动激活,导致致癌转化。我们先前已经鉴定了大麻的特定化学变体,其通过阻止白血病细胞中的Notch1成熟来诱导细胞凋亡。这里,我们从这种化学制剂中分离出三种大麻素,它们协同模拟了整个提取物的作用。两个以前是已知的,大麻二酚(CBD)和大麻二酚(CBDV),而第三种大麻素,我们称之为331-18A,在这项研究中得到了鉴定和充分表征。我们证明了这些大麻素通过2型大麻素受体和TRPV1起作用,通过消耗细胞内Ca2来激活整合的应激反应途径。其次是ATF4、CHOP、和CHAC1,其通过抑制上游起始因子eIF2α而受阻。CHAC1丰度的增加阻止了Notch1成熟,从而降低活性Notch1胞内结构域的水平,并因此降低细胞活力和增加细胞凋亡。在小鼠模型中,用三种分离的分子治疗导致体内肿瘤大小和重量减小,并减缓白血病进展。总之,这项研究阐明了三种不同的大麻素在调节Notch1途径中的作用机制,并且构成了建立治疗NOTCH1突变疾病和癌症如T-ALL的新疗法的重要步骤。
    In T-cell acute lymphoblastic leukemia (T-ALL), more than 50% of cases display autoactivation of Notch1 signaling, leading to oncogenic transformation. We have previously identified a specific chemovar of Cannabis that induces apoptosis by preventing Notch1 maturation in leukemia cells. Here, we isolated three cannabinoids from this chemovar that synergistically mimic the effects of the whole extract. Two were previously known, cannabidiol (CBD) and cannabidivarin (CBDV), whereas the third cannabinoid, which we termed 331-18A, was identified and fully characterized in this study. We demonstrated that these cannabinoids act through cannabinoid receptor type 2 and TRPV1 to activate the integrated stress response pathway by depleting intracellular Ca2+. This is followed by increased mRNA and protein expression of ATF4, CHOP, and CHAC1, which is hindered by inhibiting the upstream initiation factor eIF2α. The increased abundance of CHAC1 prevents Notch1 maturation, thereby reducing the levels of the active Notch1 intracellular domain, and consequently decreasing cell viability and increasing apoptosis. Treatment with the three isolated molecules resulted in reduced tumor size and weight in vivo and slowed leukemia progression in mice models. Altogether, this study elucidated the mechanism of action of three distinct cannabinoids in modulating the Notch1 pathway, and constitutes an important step in the establishment of a new therapy for treating NOTCH1-mutated diseases and cancers such as T-ALL.
    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
    骨髓增生异常综合征(MDS)的突变谱已经确定,相对少量的遗传畸变,包括SF3B1和SRSF2剪接体突变,导致特定的表型和预后亚群。我们对两个已发表的骨髓单核细胞(BMMNCs)和CD34+细胞的MDS队列进行了多组学因子分析(MOFA),具有三种数据模式(临床,基因型,和转录组学)。七种不同的观点,包括免疫特征,炎症/衰老,反转录转座子(RTE)表达,和细胞类型组成,从这些方法中得出,以确定对MDS预后有重大影响的潜在因素。SF3B1是BMMNC队列中13个突变中唯一的突变,表明与高炎症显著相关。在CD34+组群中也观察到较小程度的这种趋势。有趣的是,代表炎症的MOFA因子对高度炎症的MDS患者显示良好的预后。相比之下,SRSF2突变病例显示粒细胞-单核细胞祖细胞(GMP)模式和高水平的衰老,免疫衰老,和恶性骨髓细胞,与他们的不良预后一致。此外,MOFA将RTE表达确定为MDS的危险因素。这项工作阐明了我们评估MDS风险的综合方法的有效性,该方法超出了迄今为止针对MDS描述的所有评分系统。
    Mutational profiles of myelodysplastic syndromes (MDS) have established that a relatively small number of genetic aberrations, including SF3B1 and SRSF2 spliceosome mutations, lead to specific phenotypes and prognostic subgrouping. We performed a multi-omics factor analysis (MOFA) on two published MDS cohorts of bone marrow mononuclear cells (BMMNCs) and CD34 + cells with three data modalities (clinical, genotype, and transcriptomics). Seven different views, including immune profile, inflammation/aging, retrotransposon (RTE) expression, and cell-type composition, were derived from these modalities to identify the latent factors with significant impact on MDS prognosis. SF3B1 was the only mutation among 13 mutations in the BMMNC cohort, indicating a significant association with high inflammation. This trend was also observed to a lesser extent in the CD34 + cohort. Interestingly, the MOFA factor representing the inflammation shows a good prognosis for MDS patients with high inflammation. In contrast, SRSF2 mutant cases show a granulocyte-monocyte progenitor (GMP) pattern and high levels of senescence, immunosenescence, and malignant myeloid cells, consistent with their poor prognosis. Furthermore, MOFA identified RTE expression as a risk factor for MDS. This work elucidates the efficacy of our integrative approach to assess the MDS risk that goes beyond all the scoring systems described thus far for MDS.
    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
    由于新型药物在一线治疗中的整合主要影响了晚期经典霍奇金淋巴瘤(cHL)的前期治疗,这篇综述将概述一线晚期cHL以及进展和复发时的当前管理,专注于生物学,临床特征,和治疗方法。由于S1826,HD21和ECHELON-1,晚期cHL的一线治疗发生了巨大变化,新的药物是标准一线治疗的一部分。BV-AVD,Bracadd,Nivo-AVD现在是III-IV期cHL患者的标准一线治疗方案,与cHL的历史数据相比,结果有所改善。在复发/难治性(r/r)cHL化疗方案中添加BV和PD-1抑制剂可改善该人群的预后。现在,PD-1进入一线治疗有一个范式转变,因此,需要进行新的研究来评估这些新型药剂在救助中的作用,以确定r/rcHL的最佳救助方法。
    As the integration of novel agents in the frontline therapy has primarily impacted upfront therapy of advanced stage classic Hodgkin lymphoma (cHL), this review will outline current management of advanced stage cHL at first line and at progression and relapse, focusing on the biology, clinical features, and therapeutic approaches. Due to S1826, HD21, and ECHELON-1, the first-line treatment of advanced cHL has dramatically changed, with novel agents part of standard frontline therapy. BV-AVD, BrECADD, and Nivo-AVD are now standard first-line regimens for patients with stage III-IV cHL, with improved outcomes compared to historical data in cHL. The addition of BV and PD-1 inhibitors to relapsed/refractory (r/r) cHL chemotherapy regimens improved outcomes in this population. Now, there is a paradigm shift with PD-1 moving into frontline therapy, so new studies to evaluate the role of these novel agents in salvage will be required to determine the optimal salvage approach in r/r cHL.
    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
    KRAS的突变激活通常发生在肺癌发生中,随着最近美国食品和药物管理局批准的KRASG12C的共价抑制剂,如索托拉西或阿达格拉西布,KRAS癌蛋白是非小细胞肺癌(NSCLC)的重要药理靶点。然而,并非所有KRASG12C驱动的NSCLC都对这些抑制剂有反应,在那些有反应的患者中出现耐药性可以是快速和多效性的。因此,基于KRASG12C的共价抑制骨架,正在努力开发有效的联合疗法。这里,我们报道,抑制KRASG12C信号可增加表达KRASG12C的肺癌细胞的自噬.此外,选择性ULK1/2抑制剂DCC-3116的组合,加索托拉西布显示协同/协同抑制体外人KRASG12C驱动的肺癌细胞增殖和体内优异的肿瘤控制。此外,在KRASG12C驱动的非小细胞肺癌的基因工程小鼠模型中,抑制KRASG12C或ULK1/2可降低肿瘤负荷并增加小鼠存活率.因此,这些数据提示ULK1/2介导的自噬是肺癌中对KRASG12C抑制的一种药理学上可行的细胞保护应激反应.
    Mutational activation of KRAS occurs commonly in lung carcinogenesis and, with the recent U.S. Food and Drug Administration approval of covalent inhibitors of KRASG12C such as sotorasib or adagrasib, KRAS oncoproteins are important pharmacological targets in non-small cell lung cancer (NSCLC). However, not all KRASG12C-driven NSCLCs respond to these inhibitors, and the emergence of drug resistance in those patients who do respond can be rapid and pleiotropic. Hence, based on a backbone of covalent inhibition of KRASG12C, efforts are underway to develop effective combination therapies. Here, we report that the inhibition of KRASG12C signaling increases autophagy in KRASG12C-expressing lung cancer cells. Moreover, the combination of DCC-3116, a selective ULK1/2 inhibitor, plus sotorasib displays cooperative/synergistic suppression of human KRASG12C-driven lung cancer cell proliferation in vitro and superior tumor control in vivo. Additionally, in genetically engineered mouse models of KRASG12C-driven NSCLC, inhibition of either KRASG12C or ULK1/2 decreases tumor burden and increases mouse survival. Consequently, these data suggest that ULK1/2-mediated autophagy is a pharmacologically actionable cytoprotective stress response to inhibition of KRASG12C in lung 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
    免疫逃避是了解肿瘤复发的关键现象,转移,以及肿瘤进展中的其他关键步骤。肿瘤微环境(TME)由于肿瘤释放影响它的信号的能力而处于不断变化的状态,而其中的免疫细胞可以影响癌细胞的行为。癌细胞经历了一些变化,改变不同免疫细胞的富集,调节肿瘤微环境中现有免疫细胞的活性。癌细胞可以通过下调抗原呈递或表达免疫检查点分子来逃避免疫监视。高水平的肿瘤浸润淋巴细胞(TIL)与更好的结果相关,强大的免疫反应可以控制肿瘤的生长。相反,Tregs的富集增加,骨髓来源的抑制细胞,M2样抗炎巨噬细胞可阻碍有效的免疫监视并预测不良预后。总的来说,了解这些免疫逃避机制指导治疗策略。研究人员旨在调节TME以增强免疫监视并改善患者预后。在这篇评论文章中,我们努力总结肿瘤免疫微环境的组成,影响肿瘤免疫微环境(TIME)的因素,以及针对免疫细胞的不同治疗方式。这篇综述是了解免疫监视和免疫逃避基础知识的第一手参考。
    Immune evasion is a key phenomenon in understanding tumor recurrence, metastasis, and other critical steps in tumor progression. The tumor microenvironment (TME) is in constant flux due to the tumor\'s ability to release signals that affect it, while immune cells within it can impact cancer cell behavior. Cancer cells undergo several changes, which can change the enrichment of different immune cells and modulate the activity of existing immune cells in the tumor microenvironment. Cancer cells can evade immune surveillance by downregulating antigen presentation or expressing immune checkpoint molecules. High levels of tumor-infiltrating lymphocytes (TILs) correlate with better outcomes, and robust immune responses can control tumor growth. On the contrary, increased enrichment of Tregs, myeloid-derived suppressor cells, and M2-like anti-inflammatory macrophages can hinder effective immune surveillance and predict poor prognosis. Overall, understanding these immune evasion mechanisms guides therapeutic strategies. Researchers aim to modulate the TME to enhance immune surveillance and improve patient outcomes. In this review article, we strive to summarize the composition of the tumor immune microenvironment, factors affecting the tumor immune microenvironment (TIME), and different therapeutic modalities targeting the immune cells. This review is a first-hand reference to understand the basics of immune surveillance and immune evasion.
    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
    肿瘤代谢的异质性是重要的,但对肿瘤生物学的了解仍然很少。目前的工作重点是使用氧化还原辅因子NAD(P)H的荧光寿命成像(FLIM)对大肠癌的细胞代谢异质性进行可视化和定量。在四种癌细胞系(HT29,HCT116,CaCo2和CT26)中进行了NAD(P)H的FLIM显微镜检查,在小鼠四种类型的结肠直肠肿瘤中的体内和在患者体内的肿瘤样本中的离体。评估衰变参数的分散性和双峰性,以量化细胞间代谢异质性。我们的结果表明,与培养细胞和肿瘤异种移植物相比,结直肠肿瘤患者的能量代谢异质性明显更高,其显示为样品中NAD(P)H的游离(糖酵解)部分的贡献的更广泛且频繁的双峰分布。在肿瘤患者中,高等级和早期阶段的分散性更大,没有,然而,任何与双峰的联系。这些结果表明,从NAD(P)HFLIM评估的细胞水平代谢异质性有可能成为临床预后因素。
    Heterogeneity of tumor metabolism is an important, but still poorly understood aspect of tumor biology. Present work is focused on the visualization and quantification of cellular metabolic heterogeneity of colorectal cancer using fluorescence lifetime imaging (FLIM) of redox cofactor NAD(P)H. FLIM-microscopy of NAD(P)H was performed in vitro in four cancer cell lines (HT29, HCT116, CaCo2 and CT26), in vivo in the four types of colorectal tumors in mice and ex vivo in patients\' tumor samples. The dispersion and bimodality of the decay parameters were evaluated to quantify the intercellular metabolic heterogeneity. Our results demonstrate that patients\' colorectal tumors have significantly higher heterogeneity of energy metabolism compared with cultured cells and tumor xenografts, which was displayed as a wider and frequently bimodal distribution of a contribution of a free (glycolytic) fraction of NAD(P)H within a sample. Among patients\' tumors, the dispersion was larger in the high-grade and early stage ones, without, however, any association with bimodality. These results indicate that cell-level metabolic heterogeneity assessed from NAD(P)H FLIM has a potential to become a clinical prognostic factor.
    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
    癌症被认为是COVID-19死亡率的危险因素,然而,一些国家报告说,在COVID-19大流行期间,患有主要癌症的死亡人数仍处于历史水平。这里,我们进一步阐明了美国人群中癌症死亡率与COVID-19之间的关系.我们比较了6种癌症的基础和多因(MC)死亡数据中与大流行相关的死亡率模式,糖尿病,和老年痴呆症。编码实践中与大流行相关的任何变化都应通过研究MC数据来消除。在2020年全国范围内,MC癌症死亡率仅比大流行前的基线上升了3%,相当于约13,600例超额死亡。死亡率升高在致命性较低的癌症(乳腺癌,结直肠,和血液学,2-7%)比生存率低的癌症(肺癌和胰腺癌,0-1%)。相比之下,糖尿病(37%)和阿尔茨海默病(19%)导致的MC死亡显著升高。为了理解这些差异,我们使用COVID-19发作率模拟了每种情况的预期超额死亡率,预期寿命,人口规模,以及生活在每种情况下的个体的平均年龄。我们发现,观察到的死亡率差异主要是由预期寿命的差异来解释的,与致命癌症死亡的风险超过COVID-19死亡的风险。
    确定像COVID-19这样的大流行的真实死亡人数是困难的,因为实验室测试通常太有限,无法直接计算可归因于特定病原体的死亡人数。为了克服这一点,研究人员分析了超额死亡率-也就是说,他们根据前几年的趋势将观察到的死亡人数与预期水平进行了比较.这些技术已经使用了100多年来估计大流行性流感的负担,并成为估计COVID-19大流行死亡的流行方法。超额死亡率也可以揭示COVID-19对患有慢性病的亚人群的影响。例如,以前的研究表明,糖尿病死亡,被列为主要死亡原因的心脏病和阿尔茨海默氏病在COVID-19浪潮期间增加。癌症死亡没有表现出这样的模式,然而,尽管一些流行病学研究确定癌症是COVID-19死亡的危险因素。为了理解为什么会这样,汉森等人。在大流行的第一年,审查了来自美国不同州的死亡证明。他们对多原因死亡记录的分析(在死亡证明上列出癌症,不仅仅是死亡的主要原因)表明,大流行期间的死亡证明编码实践并不能解释癌症死亡率过高的原因。虽然预期寿命较长的癌症(乳腺癌,例如),预期寿命较低的癌症未观察到升高,比如胰腺癌。分析表明,特别致命的癌症缺乏超额死亡率可以通过竞争风险来解释-换句话说,死于癌症本身的高风险远远超过了COVID-19带来的额外风险。这些发现揭示了竞争性死亡风险如何掩盖COVID-19对癌症死亡率的真正影响,并解释了队列研究和超额死亡率研究之间的明显差异。为了充分理解COVID-19对癌症患者的影响,未来的研究应该着眼于大流行封锁期间晚期诊断导致癌症死亡率长期增加的可能性,和严重疾病的风险增加。
    Cancer is considered a risk factor for COVID-19 mortality, yet several countries have reported that deaths with a primary code of cancer remained within historic levels during the COVID-19 pandemic. Here, we further elucidate the relationship between cancer mortality and COVID-19 on a population level in the US. We compared pandemic-related mortality patterns from underlying and multiple cause (MC) death data for six types of cancer, diabetes, and Alzheimer\'s. Any pandemic-related changes in coding practices should be eliminated by study of MC data. Nationally in 2020, MC cancer mortality rose by only 3% over a pre-pandemic baseline, corresponding to ~13,600 excess deaths. Mortality elevation was measurably higher for less deadly cancers (breast, colorectal, and hematological, 2-7%) than cancers with a poor survival rate (lung and pancreatic, 0-1%). In comparison, there was substantial elevation in MC deaths from diabetes (37%) and Alzheimer\'s (19%). To understand these differences, we simulated the expected excess mortality for each condition using COVID-19 attack rates, life expectancy, population size, and mean age of individuals living with each condition. We find that the observed mortality differences are primarily explained by differences in life expectancy, with the risk of death from deadly cancers outcompeting the risk of death from COVID-19.
    Establishing the true death toll of a pandemic like COVID-19 is difficult, as laboratory testing is generally too limited to directly count the number of deaths that can be attributed to a particular pathogen. To overcome this, researchers analyse excess mortality – that is, they compare the observed number of deaths with the expected level based on trends in prior years. These techniques have been used for over 100 years to estimate the burden of pandemic influenza and became a popular way to estimate deaths due to the COVID-19 pandemic. Excess mortality can also reveal the impact of COVID-19 on sub-populations with chronic conditions. For example, previous studies showed that deaths with diabetes, heart disease and Alzheimer’s disease listed as the primary cause of death increased during waves of COVID-19. Cancer deaths did not show such a pattern, however, despite some epidemiological studies identifying cancer as a risk factor for COVID-19 mortality. To understand why this may be the case, Hansen et al. reviewed death certificates from different states in the United States during the first year of the pandemic. Their analyses of multiple-cause death records (listing cancer anywhere on the death certificate, not just as the primary cause of death) showed that death certificate coding practices during the pandemic did not explain the absence of excess cancer mortality. While a low level of excess mortality was detectable for cancers with longer life expectancy (breast cancer, for example), no elevation was observed for cancers with lower life expectancy, such as pancreatic cancer. The analyses demonstrate that the lack of excess mortality for especially deadly cancers can be explained through competing risks – in other words, the high risk of dying from the cancer itself vastly outweighs the additional risk posed by COVID-19. These findings shed light on how competing mortality risks might mask the true impact of COVID-19 on cancer mortality and explain the apparent discrepancy between cohort studies and excess mortality studies. To fully comprehend the impact of COVID-19 on patients living with cancers, future research should look at the possibility of longer-term increases in cancer mortality due to late diagnosis during pandemic lockdowns, and an elevated risk of severe illness.
    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
    胞嘧啶甲基化有助于调节哺乳动物的基因表达和正常造血。它由DNA甲基转移酶家族催化,包括DNMT1,DNMT3A,DNMT3B。外周T细胞淋巴瘤(PTCL)代表侵袭性成熟T细胞恶性肿瘤,表现出广泛的临床特征,预后差,分子病理学理解不充分。为了更好地了解分子景观并确定与疾病维持有关的候选基因,我们分析了PTCL的DNA甲基化和基因表达。我们发现PTCL中的甲基化模式是失调和异质的,但在所有样品中共享767个低甲基化和567个高甲基化差异甲基化区域(DMRs)以及231个基因上调和91个基因下调。提示与肿瘤发展的潜在关联。我们进一步鉴定了在大多数PTCL中与基因表达增加相关的39个低甲基化启动子。这种推定的致癌特征包括TRIP13(甲状腺激素受体相互作用子13)基因,其遗传和药理失活通过诱导G2-M阻滞和凋亡来抑制T细胞系的增殖。因此,我们的数据表明,人类PTCL具有大量的复发性甲基化改变,这些甲基化改变可能会影响对增殖至关重要的基因的表达,这些基因的靶向可能在抗淋巴瘤治疗中有益。
    Cytosine methylation contributes to the regulation of gene expression and normal hematopoiesis in mammals. It is catalyzed by the family of DNA methyltransferases that include DNMT1, DNMT3A, and DNMT3B. Peripheral T-cell lymphomas (PTCLs) represent aggressive mature T-cell malignancies exhibiting a broad spectrum of clinical features with poor prognosis and inadequately understood molecular pathobiology. To better understand the molecular landscape and identify candidate genes involved in disease maintenance, we profiled DNA methylation and gene expression of PTCLs. We found that the methylation patterns in PTCLs are deregulated and heterogeneous but share 767 hypo- and 567 hypermethylated differentially methylated regions (DMRs) along with 231 genes up- and 91 genes downregulated in all samples, suggesting a potential association with tumor development. We further identified 39 hypomethylated promoters associated with increased gene expression in the majority of PTCLs. This putative oncogenic signature included the TRIP13 (thyroid hormone receptor interactor 13) gene whose genetic and pharmacologic inactivation inhibited the proliferation of T-cell lines by inducing G2-M arrest and apoptosis. Our data thus show that human PTCLs have a significant number of recurrent methylation alterations that may affect the expression of genes critical for proliferation whose targeting might be beneficial in anti-lymphoma treatments.
    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
    MYC家族癌蛋白通过RNA聚合酶II(RNAPII)调节大量基因的表达并广泛刺激延伸。虽然控制MYC蛋白的染色质关联的因素是众所周知的,关于相互作用蛋白如何介导MYC对转录的影响,人们知之甚少。这里,我们展示了TFIIC,一种结构蛋白复合物,控制靶位点的三维染色质组织,直接结合到MYCN的氨基末端转录调节域。令人惊讶的是,TFIIC在MYCN依赖性基因表达和转录延伸中没有明显的作用。相反,MYCN和TFIIIC优先与具有暂停的RNAPII的启动子结合,并且总体上限制非磷酸化的RNAPII在启动子处的积累。与其在转录中无处不在的作用一致,MYCN广泛参与活跃启动子中心。TFIIIC的耗尽进一步增加了MYCN对这些枢纽的本地化。这种增加与核外泌体和BRCA1的失败相关,两者都参与新生RNA降解,定位到有活性的启动子。我们的数据表明,MYCN和TFIIIC在早期转录中发挥审查功能,限制了无活性RNAPII的启动子积累,并促进了新生RNA的启动子近端降解。
    MYC family oncoproteins regulate the expression of a large number of genes and broadly stimulate elongation by RNA polymerase II (RNAPII). While the factors that control the chromatin association of MYC proteins are well understood, much less is known about how interacting proteins mediate MYC\'s effects on transcription. Here, we show that TFIIIC, an architectural protein complex that controls the three-dimensional chromatin organisation at its target sites, binds directly to the amino-terminal transcriptional regulatory domain of MYCN. Surprisingly, TFIIIC has no discernible role in MYCN-dependent gene expression and transcription elongation. Instead, MYCN and TFIIIC preferentially bind to promoters with paused RNAPII and globally limit the accumulation of non-phosphorylated RNAPII at promoters. Consistent with its ubiquitous role in transcription, MYCN broadly participates in hubs of active promoters. Depletion of TFIIIC further increases MYCN localisation to these hubs. This increase correlates with a failure of the nuclear exosome and BRCA1, both of which are involved in nascent RNA degradation, to localise to active promoters. Our data suggest that MYCN and TFIIIC exert an censoring function in early transcription that limits promoter accumulation of inactive RNAPII and facilitates promoter-proximal degradation of nascent RNA.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号