CP: Cancer biology

  • 文章类型: Journal Article
    通过多组学分析癌症患者的数据的可用性正在迅速增加。然而,对这些数据进行综合分析以进行个性化目标识别并不是微不足道的。Multiomics2Targets是一个平台,使用户能够上传转录组学,蛋白质组学,和从同一癌症患者队列中收集的磷酸化蛋白质组学数据矩阵。上传数据后,Multiomics2Targets产生的报告类似于研究出版物。处理上传的矩阵,分析,并使用Enrichr工具进行可视化,KEA3,ChEA3,Expression2激酶,和TargetRanger来识别和优先考虑蛋白质,基因,和成绩单作为潜在的目标。数字和表格,以及方法和结果的描述,是自动生成的。报告包括摘要,介绍,方法,结果,讨论,结论,和引用,并可导出为可引用的PDF和Jupyter笔记本。Multiomics2Targets用于分析临床蛋白质组学肿瘤分析联盟(CPTAC3)泛癌症队列的第3版,确定每种CPTAC3癌症亚型的潜在靶标。Multiomics2Targets可从https://multiomics2targets获得。Maayanlab.云/。
    The availability of data from profiling of cancer patients with multiomics is rapidly increasing. However, integrative analysis of such data for personalized target identification is not trivial. Multiomics2Targets is a platform that enables users to upload transcriptomics, proteomics, and phosphoproteomics data matrices collected from the same cohort of cancer patients. After uploading the data, Multiomics2Targets produces a report that resembles a research publication. The uploaded matrices are processed, analyzed, and visualized using the tools Enrichr, KEA3, ChEA3, Expression2Kinases, and TargetRanger to identify and prioritize proteins, genes, and transcripts as potential targets. Figures and tables, as well as descriptions of the methods and results, are automatically generated. Reports include an abstract, introduction, methods, results, discussion, conclusions, and references and are exportable as citable PDFs and Jupyter Notebooks. Multiomics2Targets is applied to analyze version 3 of the Clinical Proteomic Tumor Analysis Consortium (CPTAC3) pan-cancer cohort, identifying potential targets for each CPTAC3 cancer subtype. Multiomics2Targets is available from https://multiomics2targets.maayanlab.cloud/.
    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
    从多组学数据中提取预后因素的深度学习工具最近有助于对生存结果进行个性化预测。然而,集成组学-成像-临床数据集的有限规模带来了挑战.这里,我们提出了两种生物学可解释和强大的深度学习架构,用于非小细胞肺癌(NSCLC)患者的生存预测,同时从计算机断层扫描(CT)扫描图像中学习,基因表达数据,和临床信息。拟议的模型集成了患者特定的临床,转录组,和成像数据,并纳入京都基因和基因组百科全书(KEGG)和反应组途径信息,在学习过程中增加生物学知识,以提取预后基因生物标志物和分子通路。虽然在仅130名患者的数据集上进行训练时,这两种模型都可以准确地对高风险和低风险组的患者进行分层,在稀疏自动编码器中引入交叉注意机制显着提高了性能,突出肿瘤区域和NSCLC相关基因作为潜在的生物标志物,因此在从小型成像组学临床样本中学习时提供了显着的方法学进步。
    Deep-learning tools that extract prognostic factors derived from multi-omics data have recently contributed to individualized predictions of survival outcomes. However, the limited size of integrated omics-imaging-clinical datasets poses challenges. Here, we propose two biologically interpretable and robust deep-learning architectures for survival prediction of non-small cell lung cancer (NSCLC) patients, learning simultaneously from computed tomography (CT) scan images, gene expression data, and clinical information. The proposed models integrate patient-specific clinical, transcriptomic, and imaging data and incorporate Kyoto Encyclopedia of Genes and Genomes (KEGG) and Reactome pathway information, adding biological knowledge within the learning process to extract prognostic gene biomarkers and molecular pathways. While both models accurately stratify patients in high- and low-risk groups when trained on a dataset of only 130 patients, introducing a cross-attention mechanism in a sparse autoencoder significantly improves the performance, highlighting tumor regions and NSCLC-related genes as potential biomarkers and thus offering a significant methodological advancement when learning from small imaging-omics-clinical samples.
    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
    PAX3/7融合阴性横纹肌肉瘤(FN-RMS)是儿童中胚层系恶性肿瘤,对转移性或复发性病例预后不良。对高级FN-RMS的有限理解部分归因于缺乏连续的侵袭和传播事件以及研究细胞行为的挑战。使用,例如,非侵入性活体显微镜(IVM),在目前使用的异种移植模型中。这里,我们开发了FN-RMS的原位异种舌移植模型,以使用IVM研究细胞行为和侵袭和转移的分子基础。FN-RMS细胞保留在舌中,并局部侵入肌肌间隙和血管腔,有血行播散到肺和淋巴播散到淋巴结的证据。使用舌异种移植物的IVM揭示了细胞表型的变化,迁移到血液和淋巴管,和淋巴渗透.从这个模型对肿瘤侵袭和转移的组织的洞察力,细胞,和亚细胞水平可以指导晚期FN-RMS的新治疗途径。
    PAX3/7 fusion-negative rhabdomyosarcoma (FN-RMS) is a childhood mesodermal lineage malignancy with a poor prognosis for metastatic or relapsed cases. Limited understanding of advanced FN-RMS is partially attributed to the absence of sequential invasion and dissemination events and the challenge in studying cell behavior, using, for example, non-invasive intravital microscopy (IVM), in currently used xenograft models. Here, we developed an orthotopic tongue xenograft model of FN-RMS to study cell behavior and the molecular basis of invasion and metastasis using IVM. FN-RMS cells are retained in the tongue and invade locally into muscle mysial spaces and vascular lumen, with evidence of hematogenous dissemination to the lungs and lymphatic dissemination to lymph nodes. Using IVM of tongue xenografts reveals shifts in cellular phenotype, migration to blood and lymphatic vessels, and lymphatic intravasation. Insight from this model into tumor invasion and metastasis at the tissue, cellular, and subcellular level can guide new therapeutic avenues for advanced FN-RMS.
    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
    肿瘤微环境含有多种不同的细胞类型,对肿瘤生物学产生不同的影响。在本期的细胞报告方法中,Raffo-Romero等人。标准化和优化的3D肿瘤类器官,以体外模拟肿瘤相关巨噬细胞和肿瘤细胞之间的相互作用。
    The tumor microenvironment harbors a variety of different cell types that differentially impact tumor biology. In this issue of Cell Reports Methods, Raffo-Romero et al. standardized and optimized 3D tumor organoids to model the interactions between tumor-associated macrophages and tumor cells in vitro.
    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
    肿瘤的细胞成分及其微环境在肿瘤的进展中起着关键作用,患者生存,以及对癌症治疗的反应。通过单细胞RNA测序(scRNA-seq)数据在大量肿瘤中揭示全面的细胞特征至关重要,因为它揭示了固有的肿瘤细胞特征,这些特征无法通过传统的癌症亚型方法进行识别。我们的贡献,scBeacon,是一种工具,通过整合和聚类多个scRNA-seq数据集来提取用于在批量样本上对不相关的肿瘤数据集进行去卷积的签名,从而得出细胞类型签名。通过在癌症基因组图谱(TCGA)队列中使用scBeacon,我们发现特定肿瘤类别中的细胞和分子属性,许多与患者结果相关。我们开发了肿瘤细胞类型图,以基于细胞类型推断直观地描绘TCGA样品之间的关系。
    The cellular components of tumors and their microenvironment play pivotal roles in tumor progression, patient survival, and the response to cancer treatments. Unveiling a comprehensive cellular profile within bulk tumors via single-cell RNA sequencing (scRNA-seq) data is crucial, as it unveils intrinsic tumor cellular traits that elude identification through conventional cancer subtyping methods. Our contribution, scBeacon, is a tool that derives cell-type signatures by integrating and clustering multiple scRNA-seq datasets to extract signatures for deconvolving unrelated tumor datasets on bulk samples. Through the employment of scBeacon on the The Cancer Genome Atlas (TCGA) cohort, we find cellular and molecular attributes within specific tumor categories, many with patient outcome relevance. We developed a tumor cell-type map to visually depict the relationships among TCGA samples based on the cell-type inferences.
    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
    未知原发癌(CUP)代表转移性癌症,尽管有标准的诊断程序,原发部位仍未被识别。为了确定这种情况下的肿瘤起源,我们开发了BPformer,一种深度学习方法,将变压器模型与生物路径的先验知识相结合。对来自32种癌症类型的10,410种原发性肿瘤的转录组进行了培训,BPformer取得了94%的显著准确率,92%,89%在原发肿瘤和转移性肿瘤的原发和转移部位,分别,超越现有方法。此外,BPformer在一项回顾性研究中得到了验证,与通过免疫组织化学和组织病理学诊断的肿瘤部位一致。此外,BPformer能够根据它们对肿瘤起源鉴定的贡献对通路进行排序,这有助于将致癌信号传导途径分类为在不同癌症中高度保守的那些,而不是根据其起源高度可变的那些。
    Cancer of unknown primary (CUP) represents metastatic cancer where the primary site remains unidentified despite standard diagnostic procedures. To determine the tumor origin in such cases, we developed BPformer, a deep learning method integrating the transformer model with prior knowledge of biological pathways. Trained on transcriptomes from 10,410 primary tumors across 32 cancer types, BPformer achieved remarkable accuracy rates of 94%, 92%, and 89% in primary tumors and primary and metastatic sites of metastatic tumors, respectively, surpassing existing methods. Additionally, BPformer was validated in a retrospective study, demonstrating consistency with tumor sites diagnosed through immunohistochemistry and histopathology. Furthermore, BPformer was able to rank pathways based on their contribution to tumor origin identification, which helped to classify oncogenic signaling pathways into those that are highly conservative among different cancers versus those that are highly variable depending on their origins.
    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
    3D肿瘤通过概括肿瘤的复杂多样性,彻底改变了体外/离体癌症生物学。虽然肿瘤样为癌症发展和治疗反应提供了新的见解,仍然存在一些限制。作为肿瘤的微环境,尤其是免疫系统,强烈影响肿瘤的发展,肿瘤中缺乏免疫细胞可能导致不恰当的结论.巨噬细胞,肿瘤进展的关键角色,整合到肿瘤中特别具有挑战性。在这项研究中,我们建立了三种优化和标准化的共培养人巨噬细胞和乳腺癌肿瘤样的方法:一种半液体模型和两种针对特定应用定制的基质嵌入模型.然后,我们使用流式细胞术和光片显微镜跟踪这些系统中的相互作用和巨噬细胞浸润,并显示巨噬细胞不仅影响肿瘤分子谱,而且影响化疗反应。这强调了增加3D模型的复杂性以更准确地反映体内状况的重要性。
    3D tumoroids have revolutionized in vitro/ex vivo cancer biology by recapitulating the complex diversity of tumors. While tumoroids provide new insights into cancer development and treatment response, several limitations remain. As the tumor microenvironment, especially the immune system, strongly influences tumor development, the absence of immune cells in tumoroids may lead to inappropriate conclusions. Macrophages, key players in tumor progression, are particularly challenging to integrate into the tumoroids. In this study, we established three optimized and standardized methods for co-culturing human macrophages with breast cancer tumoroids: a semi-liquid model and two matrix-embedded models tailored for specific applications. We then tracked interactions and macrophage infiltration in these systems using flow cytometry and light sheet microscopy and showed that macrophages influenced not only tumoroid molecular profiles but also chemotherapy response. This underscores the importance of increasing the complexity of 3D models to more accurately reflect in vivo conditions.
    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
    我们提出了一种整合全基因组多组数据的创新策略,它通过利用多任务编码器从高维组学数据中导出的隐藏层特征来促进自适应合并。对八个基准癌症数据集的经验评估证实,我们提出的框架超过了癌症亚型的比较算法,提供优越的亚型结果。在这些子类型结果的基础上,我们建立了一个强大的管道来识别全基因组生物标志物,发掘195个重要的生物标志物。此外,我们进行了详尽的分析,以评估在癌症亚型分型过程中,在全基因组水平上每个组学和非编码区特征的重要性.我们的研究表明,组学和非编码区特征都会对癌症的发展和生存预后产生重大影响。这项研究强调了整合全基因组数据在癌症研究中的潜在和实际意义。证明了全面基因组表征的效力。此外,我们的发现为采用深度学习方法的多组学分析提供了有见地的观点.
    We present an innovative strategy for integrating whole-genome-wide multi-omics data, which facilitates adaptive amalgamation by leveraging hidden layer features derived from high-dimensional omics data through a multi-task encoder. Empirical evaluations on eight benchmark cancer datasets substantiated that our proposed framework outstripped the comparative algorithms in cancer subtyping, delivering superior subtyping outcomes. Building upon these subtyping results, we establish a robust pipeline for identifying whole-genome-wide biomarkers, unearthing 195 significant biomarkers. Furthermore, we conduct an exhaustive analysis to assess the importance of each omic and non-coding region features at the whole-genome-wide level during cancer subtyping. Our investigation shows that both omics and non-coding region features substantially impact cancer development and survival prognosis. This study emphasizes the potential and practical implications of integrating genome-wide data in cancer research, demonstrating the potency of comprehensive genomic characterization. Additionally, our findings offer insightful perspectives for multi-omics analysis employing deep learning methodologies.
    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
    局部皮肤神经纤维瘤(cNFs)是良性肿瘤,出现在受1型神经纤维瘤病综合征影响的患者的真皮中。cNFs是良性病变:它们不经历恶性转化或转移。然而,它们可以覆盖身体的很大一部分,有些人会发展成百上千的病变。cNF可以引起疼痛,瘙痒,和毁容导致重大的社会情感影响。目前,手术和激光干燥是唯一的治疗选择,但可能会导致疤痕和潜在的再生不完全切除。为了确定有效的全身疗法,我们介绍了一种建立和筛选cNF类器官的方法。我们优化了条件以支持基因组多样的cNF的离体生长。通过免疫组织病理学测量,患者来源的cNF类器官密切概括了亲本肿瘤的细胞和分子特征,甲基化,RNA测序,和流式细胞术。我们的cNF类器官平台能够以患者和肿瘤特异性方式快速筛选数百种化合物。
    Localized cutaneous neurofibromas (cNFs) are benign tumors that arise in the dermis of patients affected by neurofibromatosis type 1 syndrome. cNFs are benign lesions: they do not undergo malignant transformation or metastasize. Nevertheless, they can cover a significant proportion of the body, with some individuals developing hundreds to thousands of lesions. cNFs can cause pain, itching, and disfigurement resulting in substantial socio-emotional repercussions. Currently, surgery and laser desiccation are the sole treatment options but may result in scarring and potential regrowth from incomplete removal. To identify effective systemic therapies, we introduce an approach to establish and screen cNF organoids. We optimized conditions to support the ex vivo growth of genomically diverse cNFs. Patient-derived cNF organoids closely recapitulate cellular and molecular features of parental tumors as measured by immunohistopathology, methylation, RNA sequencing, and flow cytometry. Our cNF organoid platform enables rapid screening of hundreds of compounds in a patient- and tumor-specific manner.
    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
    先前尚未研究蛋白质周转在胰腺导管腺癌(PDA)转移中的作用。我们介绍了类器官的动态稳定同位素标记(dSILO):一种动态SILAC衍生物,将同位素标记的氨基酸脉冲与同量异位串联质量标签(TMT)标记相结合,以测量类器官中蛋白质组范围的蛋白质转换率。我们将其应用于PDA模型,发现与原发性肿瘤器官相比,转移性器官表现出加速的整体蛋白质组更新。全球范围内,大多数营业额的变化没有反映在蛋白质丰度水平上。有趣的是,与肿瘤相比,转移性PDA中显示最高周转增加的一组蛋白质与线粒体呼吸有关。这表明转移性PDA可以采用替代的呼吸链功能,该功能受蛋白质翻转速率的控制。总的来说,我们对PDA类器官中蛋白质组周转的分析提供了对PDA转移的潜在机制的见解.
    The role of protein turnover in pancreatic ductal adenocarcinoma (PDA) metastasis has not been previously investigated. We introduce dynamic stable-isotope labeling of organoids (dSILO): a dynamic SILAC derivative that combines a pulse of isotopically labeled amino acids with isobaric tandem mass-tag (TMT) labeling to measure proteome-wide protein turnover rates in organoids. We applied it to a PDA model and discovered that metastatic organoids exhibit an accelerated global proteome turnover compared to primary tumor organoids. Globally, most turnover changes are not reflected at the level of protein abundance. Interestingly, the group of proteins that show the highest turnover increase in metastatic PDA compared to tumor is involved in mitochondrial respiration. This indicates that metastatic PDA may adopt alternative respiratory chain functionality that is controlled by the rate at which proteins are turned over. Collectively, our analysis of proteome turnover in PDA organoids offers insights into the mechanisms underlying PDA metastasis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号