Cell of origin

起源细胞
  • 文章类型: Journal Article
    来自血液和其他体液的无细胞核酸(例如,无细胞DNA或cfDNA)的临床基因组谱分析已经开启了针对健康状况和诸如癌症的疾病的非侵入性诊断和治疗监测策略的新时代。cfDNA的基因组分析不仅可以识别疾病相关的突变,但新发现表明,结构,拓扑,cfDNA的碎片特征揭示了有关源组织位置的关键信息,他们的表观基因组,和其他临床相关特征,导致碎片组学的新兴领域。该领域已经看到了计算和基因组学方法的快速发展,用于对健康状况和疾病进行大规模研究-这导致了基础,机械发现以及翻译应用。最近的几项研究表明,cfDNA片段组学技术的临床应用可能对早期疾病诊断有效,确定治疗结果,和无风险的连续病人监测在一个非侵入性的方式。在这篇文章中,我们概述了计算基因组方法和分析策略的最新进展,以及来自cfNA碎片组学的新兴见解。最后,我们强调当前的挑战和机遇。
    Clinical genomic profiling of cell-free nucleic acids (e.g. cell-free DNA or cfDNA) from blood and other body fluids has ushered in a new era in non-invasive diagnostics and treatment monitoring strategies for health conditions and diseases such as cancer. Genomic analysis of cfDNAs not only identifies disease-associated mutations, but emerging findings suggest that structural, topological, and fragmentation characteristics of cfDNAs reveal crucial information about the location of source tissues, their epigenomes, and other clinically relevant characteristics, leading to the burgeoning field of fragmentomics. The field has seen rapid developments in computational and genomics methodologies for conducting large-scale studies on health conditions and diseases - that have led to fundamental, mechanistic discoveries as well as translational applications. Several recent studies have shown the clinical utilities of the cfDNA fragmentomics technique which has the potential to be effective for early disease diagnosis, determining treatment outcomes, and risk-free continuous patient monitoring in a non-invasive manner. In this article, we outline recent developments in computational genomic methodologies and analysis strategies, as well as the emerging insights from cfNA fragmentomics. We conclude by highlighting the current challenges and opportunities.
    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
    尽管患者预后有所改善,儿科癌症仍然是儿童非意外死亡的主要原因.最近对儿科癌症患者的遗传分析表明,种系遗传易感性和癌症特异性体细胞驱动突变都具有重要作用。越来越多,有证据表明,起源癌细胞转化的发育时间点对于肿瘤身份和治疗反应至关重要.因此,未来的治疗发展将通过使用忠实地概括遗传背景的疾病模型来支持,细胞起源,和儿童癌症的脆弱性发展窗口。人类干细胞有可能将所有这些特征整合到儿科癌症模型中,同时作为快速遗传和药理测试的平台。在这次审查中,我们描述了人类干细胞是如何用于儿科癌症模型的,以及这些模型与其他儿科癌症模型模式的比较.
    今天,儿童癌症是儿童非意外死亡的主要原因.为了进一步改善结果,对于研究人员和临床医生来说,认识到小儿癌症与成人癌症的区别非常重要。遗传的癌症风险可能在儿科癌症风险中发挥更大的作用,和随后的肿瘤特异性获得性驱动突变启动肿瘤形成。然而,遗传和获得性突变之间存在实质性的相互作用,这支持同时考虑两者。生物技术的最新进展,改善了早期发育细胞和儿科癌细胞之间的匹配,尽管某些儿童中枢神经系统肿瘤的细胞来源仍然难以捉摸。越来越多,证据,特别是在小儿髓母细胞瘤中,证明了癌细胞起源转化的发育时间点对于肿瘤身份和治疗反应至关重要。因此,未来的治疗发展将通过使用忠实地概括遗传背景的疾病模型来支持,细胞起源,和儿科癌症的发育窗口。人类干细胞有可能将所有这些特征整合到儿科癌症模型中,同时作为快速遗传和药理测试的平台。在这次审查中,我们描述了如何使用人类干细胞来模拟儿科癌症,这些模型与其他儿科癌症模型相比,以及未来如何改进这些模型。
    Despite improvements in patient outcomes, pediatric cancer remains a leading cause of non-accidental death in children. Recent genetic analysis of patients with pediatric cancers indicates an important role for both germline genetic predisposition and cancer-specific somatic driver mutations. Increasingly, evidence demonstrates that the developmental timepoint at which the cancer cell-of-origin transforms is critical to tumor identity and therapeutic response. Therefore, future therapeutic development would be bolstered by the use of disease models that faithfully recapitulate the genetic context, cell-of-origin, and developmental window of vulnerability in pediatric cancers. Human stem cells have the potential to incorporate all of these characteristics into a pediatric cancer model, while serving as a platform for rapid genetic and pharmacological testing. In this review, we describe how human stem cells have been used to model pediatric cancers and how these models compare to other pediatric cancer model modalities.
    Today, pediatric cancer is a leading cause of non-accidental death in children. In order to further improve outcomes, it is important for researchers and clinicians alike to recognize how pediatric cancers are distinct from adult cancers. Inherited risk of cancer may play a greater role in pediatric cancer risk, and subsequent tumor-specific acquired driver mutations initiate tumor formation. However, there is substantial interaction between inherited and acquired mutations, which supports consideration of both simultaneously. Recent advancements in biotechnology, have improved matching between early cells of development and pediatric cancer cells, although cell-of-origin for certain pediatric central nervous system tumors remain elusive. Increasingly, evidence, particularly in pediatric medulloblastoma, demonstrates that the developmental timepoint at which the cancer cell-of-origin transforms is critical to tumor identity and therapeutic response. Therefore, future therapeutic development would be bolstered by the use of disease models that faithfully recapitulate the genetic context, cell-of-origin, and developmental window of pediatric cancers. Human stem cells have the potential to incorporate all of these characteristics into a pediatric cancer model, while serving as a platform for rapid genetic and pharmacological testing. In this review, we describe how human stem cells have been used to model pediatric cancers, how human these models compare to other pediatric cancer model modalities, and how these models can be improved in the future.
    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
    本研究旨在使用Hans算法分析弥漫性大B细胞淋巴瘤(DLBCLs)中起源细胞(COO)的异质性或变化,其中包括156例具有多个DLBCL标本的患者。通过CD10、BCL6和MUM1的免疫组织化学染色检测COO。在50(32%)和106(68%)患者中,初始诊断时主要肿瘤的COO是生发中心B细胞(GCB)和非GCB类型,分别。126例患者(81%)没有改变。然而,30名患者(19%)发生了变化,从GCB到非GCB在12例患者中,反之亦然在18例患者中。在初始诊断期间,在其他站点的同时样本中有14%的COO是异质或变化的,在7%的主要难愈部位,以及在复发阶段获得的除原发部位以外的20%的样本。CD10,BCL6和MUM1表达的变化在15%,23%,和24%的样本,分别。在具有CD10+/BCL6+/MUM1-的DLBCL中观察到COO变化的低发生率(4%),CD10-/BCL6-/MUM1+(3%),和CD10-/BCL6-/MUM1-(0%)模式,而其他模式的DLBCL显示COO变化的比率为20-37%。总之,在19%的DLBCL病例中,COO是异质的或改变的。应在其他活检样本中重新检查COO,以确定最佳治疗方法。
    This study aimed to analyze the heterogeneity or change in cell of origin (COO) in diffuse large B-cell lymphoma (DLBCLs) using the Hans algorithm including 156 patients with multiple DLBCL specimens. COO was detected via immunohistochemical staining for CD10, BCL6, and MUM1. The COO of the main tumor at initial diagnosis was germinal center B-cell (GCB) and non-GCB type in 50 (32%) and 106 (68%) patients, respectively. It did not change in 126 patients (81%). However, it changed in 30 patients (19%), from GCB to non-GCB in 12 patients and vice versa in 18 patients. The COO was heterogeneous or changed in 14% of simultaneous samples at other sites during the initial diagnosis, in 7% of primary refractory sites, and in 20% of samples obtained in the relapse phase other than the primary site. Changes in CD10, BCL6, and MUM1 expression were observed in 15%, 23%, and 24% samples, respectively. A low incidence of change in COO was observed in DLBCL with CD10+/BCL6+/MUM1- (4%), CD10-/BCL6-/MUM1+ (3%), and CD10-/BCL6-/MUM1- (0%) patterns, whereas DLBCL with other patterns showed COO changes at rates of 20-37%. In conclusion, COO was heterogeneous or changed in 19% of DLBCL cases. The COO should be re-examined in other biopsy samples to determine the optimal treatment.
    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
    目的:PTEN-AKT通路在肝外胆管癌(eCCA)中经常发生改变。我们的目的是评估PTEN在eCCA发病机理中的作用,并找到针对该疾病的新疗法。
    方法:使用Cre-loxp系统将胆管上皮细胞中的Pten基因进行基因缺失。对病理进行了宏观和组织学评估。通过免疫组织化学(IHC)进一步分析特征,RT-PCR,细胞培养,和RNAseq。将一些特征与人eCCA样品中的特征进行比较。进一步的机制研究利用Trp53和Aurora激酶A(Aurka)基因的条件性敲除。使用Aurka抑制剂测试实验疗法。
    结果:我们观察到肝外胆管上皮和导管周围腺中Pten基因的遗传缺失引发小鼠硬化性胆管炎样病变,导致小鼠肝外胆管扩大和扭曲早在一个月大。组织学上,这些病变表现出上皮增殖增加,炎性细胞浸润,和纤维化。随着年龄的增长,病变由低度发育不良发展为浸润性癌.Trp53失活进一步加速了疾病进展,可能通过下调衰老。进一步的机理研究表明,人和小鼠eCCA均显示出AURKA的高表达。值得注意的是,Aurka的基因缺失完全消除了Pten缺乏引起的肝外胆管病变。此外,药物抑制Aurka缓解疾病进展。
    结论:肝外胆管细胞和胆管周围腺体的Pten缺乏通过Aurka依赖性方式导致胆管炎到胆管癌的连续性。这些发现为肝外CCA的预防和治疗干预提供了新的见解。
    异常的PTEN-PI3K-AKT信号通路通常在人肝外胆管癌(eCCA)中观察到,预后不良的疾病。在我们的研究中,我们通过在肝外胆管上皮细胞和胆管周围腺体中通过Pdx1-Cre有条件地删除Pten基因,开发了一种模拟胆管炎对eCCA进展的小鼠模型。这些细胞中有条件的Pten缺失导致胆管炎,逐渐发展为发育不良,最终导致eCCA。Pten的丢失增强了Akt信号,细胞增殖,炎症,纤维化,DNA损伤,表观遗传信号,上皮-间质转化(EMT),细胞发育不良,和细胞衰老。Aurka的遗传缺失或药理学抑制成功地阻止了疾病进展。该模型对于测试新疗法和揭示eCCA肿瘤发生的机制将是有价值的。
    OBJECTIVE: The PTEN-AKT pathway is frequently altered in extrahepatic cholangiocarcinoma (eCCA). We aimed to evaluate the role of PTEN in the pathogenesis of eCCA and identify novel therapeutic targets for this disease.
    METHODS: The Pten gene was genetically deleted using the Cre-loxp system in biliary epithelial cells. The pathologies were evaluated both macroscopically and histologically. The characteristics were further analyzed by immunohistochemistry, reverse-transcription PCR, cell culture, and RNA sequencing. Some features were compared to those in human eCCA samples. Further mechanistic studies utilized the conditional knockout of Trp53 and Aurora kinase A (Aurka) genes. We also tested the effectiveness of an Aurka inhibitor.
    RESULTS: We observed that genetic deletion of the Pten gene in the extrahepatic biliary epithelium and peri-ductal glands initiated sclerosing cholangitis-like lesions in mice, resulting in enlarged and distorted extrahepatic bile ducts in mice as early as 1 month after birth. Histologically, these lesions exhibited increased epithelial proliferation, inflammatory cell infiltration, and fibrosis. With aging, the lesions progressed from low-grade dysplasia to invasive carcinoma. Trp53 inactivation further accelerated disease progression, potentially by downregulating senescence. Further mechanistic studies showed that both human and mouse eCCA showed high expression of AURKA. Notably, the genetic deletion of Aurka completely eliminated Pten deficiency-induced extrahepatic bile duct lesions. Furthermore, pharmacological inhibition of Aurka alleviated disease progression.
    CONCLUSIONS: Pten deficiency in extrahepatic cholangiocytes and peribiliary glands led to a cholangitis-to-cholangiocarcinoma continuum that was dependent on Aurka. These findings offer new insights into preventive and therapeutic interventions for extrahepatic CCA.
    UNASSIGNED: The aberrant PTEN-PI3K-AKT signaling pathway is commonly observed in human extrahepatic cholangiocarcinoma (eCCA), a disease with a poor prognosis. In our study, we developed a mouse model mimicking cholangitis to eCCA progression by conditionally deleting the Pten gene via Pdx1-Cre in epithelial cells and peribiliary glands of the extrahepatic biliary duct. The conditional Pten deletion in these cells led to cholangitis, which gradually advanced to dysplasia, ultimately resulting in eCCA. The loss of Pten heightened Akt signaling, cell proliferation, inflammation, fibrosis, DNA damage, epigenetic signaling, epithelial-mesenchymal transition, cell dysplasia, and cellular senescence. Genetic deletion or pharmacological inhibition of Aurka successfully halted disease progression. This model will be valuable for testing novel therapies and unraveling the mechanisms of eCCA tumorigenesis.
    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:外周T细胞淋巴瘤(PTCL)是指治疗反应和存活时间较差的异质性T细胞肿瘤组。犬PTCL临床和免疫表型类似于最常见的人类亚型,PTCL-未另作规定(PTCL-NOS),引起了人们对这种犬病作为人类PTCL自然发生模型的兴趣。人类PTCL-NOS中的基因表达谱有助于将这种模糊的诊断描述为不同的亚型,但在犬PTCL中缺乏类似的基因表达谱。
    方法:对来自33只具有CD4(26/33)的狗的肿瘤样本进行大量RNA测序,CD8+(4/33),或通过流式细胞术诊断的CD4-CD8-(3/33)PTCL,并从健康对照犬中分选CD4+和CD8+淋巴细胞。在RNA-seq数据归一化后,我们进行了差异基因表达和无监督聚类方法。进行基因集富集分析以确定犬CD4PTCL对人PTCL-NOS的富集。致癌途径,以及不同阶段的T细胞发育基因特征。我们利用基因集变异分析来评估单个犬CD4PTCL的各种人和鼠T细胞和胸腺细胞基因特征。用pan-PI3K抑制剂处理培养的犬PTCL细胞,将细胞存活和增殖与DMSO处理的对照进行比较。通过免疫组织化学验证GATA3和磷酸化AKT的表达。
    结果:虽然犬CD4+PTCL表型表现出一致的基因表达谱,CD8+和CD4-CD8-犬PTCL的表达谱更为异质性。犬CD4+PTCL的GATA3表达上调,其靶基因表达上调,富集PI3K/AKT/mTOR信号,和PTEN的下调,特征与人PTCL-NOS的更具侵袭性的GATA3-PTCL亚型一致。体外测定验证了犬CD4+PTCL细胞对PI3K/AKT/mTOR信号传导的存活和增殖的依赖性。犬CD4+PTCL富集胸腺前体基因特征,表现出不成熟标志物的表达增加(CD34,KIT,DNTT,和CCR9),与T细胞受体相关的基因下调,MHCII类相关基因(DLA-DQA1,DLA-DRA,HLA-DQB1和HLA-DQB2),CD25
    结论:犬CD4+PTCL最接近PTCL-NOS的GATA3-PTCL亚型,可能起源于T细胞发育的早期阶段,而不是更传统的成熟T辅助细胞起源。
    BACKGROUND: Peripheral T-cell lymphoma (PTCL) refers to a heterogenous group of T-cell neoplasms with poor treatment responses and survival times. Canine PTCL clinically and immunophenotypically resembles the most common human subtype, PTCL-not otherwise specified (PTCL-NOS), leading to interest in this canine disease as a naturally occurring model for human PTCL. Gene expression profiling in human PTCL-NOS has helped characterize this ambiguous diagnosis into distinct subtypes, but similar gene expression profiling in canine PTCL is lacking.
    METHODS: Bulk RNA-sequencing was performed on tumor samples from 33 dogs with either CD4+ (26/33), CD8+ (4/33), or CD4-CD8- (3/33) PTCL as diagnosed by flow cytometry, and sorted CD4+ and CD8+ lymphocytes from healthy control dogs. Following normalization of RNA-seq data, we performed differential gene expression and unsupervised clustering methods. Gene set enrichment analysis was performed to determine the enrichment of canine CD4+ PTCL for human PTCL-NOS, oncogenic pathways, and various stages of T-cell development gene signatures. We utilized gene set variation analysis to evaluate individual canine CD4+ PTCLs for various human and murine T-cell and thymocyte gene signatures. Cultured canine PTCL cells were treated with a pan-PI3K inhibitor, and cell survival and proliferation were compared to DMSO-treated controls. Expression of GATA3 and phosphorylated AKT was validated by immunohistochemistry.
    RESULTS: While the canine CD4+ PTCL phenotype exhibited a consistent gene expression profile, the expression profiles of CD8+ and CD4-CD8- canine PTCLs were more heterogeneous. Canine CD4+ PTCL had increased expression of GATA3, upregulation of its target genes, enrichment for PI3K/AKT/mTOR signaling, and downregulation of PTEN, features consistent with the more aggressive GATA3-PTCL subtype of human PTCL-NOS. In vitro assays validated the reliance of canine CD4+ PTCL cells on PI3K/AKT/mTOR signaling for survival and proliferation. Canine CD4+ PTCL was enriched for thymic precursor gene signatures, exhibited increased expression of markers of immaturity (CD34, KIT, DNTT, and CCR9), and downregulated genes associated with the T-cell receptor, MHC class II associated genes (DLA-DQA1, DLA-DRA, HLA-DQB1, and HLA-DQB2), and CD25.
    CONCLUSIONS: Canine CD4+ PTCL most closely resembled the GATA3-PTCL subtype of PTCL-NOS and may originate from an earlier stage of T-cell development than the more conventionally posited mature T-helper cell origin.
    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:弥漫性大B细胞淋巴瘤(DLBCL)是一种异质性实体。最近,已经发表了几种实现治疗和预后相关的DLBCL亚分类的算法.
    方法:74例常规DLBCL病例的队列通过免疫组织化学(IHC)进行了广泛的表征,BCL2,BCL6和MYC基因座的荧光原位杂交(FISH),和全面的高通量测序(HTS)。根据发现的基因改变,病例使用两个概率工具重新分类-LymphGen和两步分类器,允许比较这两个模型。
    结果:Hans和Tally基于IHC的总体亚分类成功率分别为96%和82%,分别。HTS和FISH数据允许LymphGen算法成功分类11/55例,(1-BN2,7-EZB,1-MCD,和2-基因复合EZB/N1)。总的亚分类率为20%。另一方面,两步分类器分类36/55例,成功率为65.5%(9-BN2,12-EZB,9-MCD,2-N1和4-ST2)。临床相关性强调MCD是与较高的复发和死亡率相关的侵袭性亚型。
    结论:两步算法在基于遗传差异对DLBCL病例进行亚分类时具有更好的成功率。需要进一步改进分类器,以增加可分类病例的数量,从而证明其在常规诊断中的适用性。
    BACKGROUND: Diffuse large B-cell lymphoma (DLBCL) is a heterogeneous entity. Lately, several algorithms achieving therapeutically and prognostically relevant DLBCL subclassification have been published.
    METHODS: A cohort of 74 routine DLBCL cases was broadly characterized by immunohistochemistry (IHC), fluorescence in situ hybridization (FISH) of the BCL2, BCL6, and MYC loci, and comprehensive high-throughput sequencing (HTS). Based on the genetic alterations found, cases were reclassified using two probabilistic tools - LymphGen and Two-step classifier, allowing for comparison of the two models.
    RESULTS: Hans and Tally\'s overall IHC-based subclassification success rate was 96% and 82%, respectively. HTS and FISH data allowed the LymphGen algorithm to successfully classify 11/55 cases (1 - BN2, 7 - EZB, 1 - MCD, and 2 - genetically composite EZB/N1). The total subclassification rate was 20%. On the other hand, the Two-step classifier categorized 36/55 cases, with 65.5% success (9 - BN2, 12 - EZB, 9 - MCD, 2 - N1, and 4 - ST2). Clinical correlations highlighted MCD as an aggressive subtype associated with higher relapse and mortality.
    CONCLUSIONS: The Two-step algorithm has a better success rate at subclassifying DLBCL cases based on genetic differences. Further improvement of the classifiers is required to increase the number of classifiable cases and thus prove their applicability in routine diagnostics.
    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
    In this review focus article, we highlight the main modifications introduced in the latest 2022 International Consensus Classification and World Health Organization classification (ICC and WHO-HAEM5) of mature T (and NK) cell neoplasms (PTCLs) and consequent implications for diagnostic practice. The changes result from recent advances in the genomic and molecular characterization of PTCLs and enhanced understanding of their pathobiology. Specifically, consideration is given to the following groups of diseases: Epstein-Barr virus (EBV)-associated neoplasms; follicular helper T cell lymphoma; anaplastic large cell lymphomas; primary intestinal T and NK cell lymphomas and lymphoproliferative disorders; and PTCL, not otherwise specified.
    UNASSIGNED: In dem vorliegenden Übersichtsbeitrag liegt der Schwerpunkt auf den wesentlichen Modifikationen der aktuellen, 2022 eingeführten Klassifikation reifer T‑ (und NK-)Zell-Neoplasien, also peripherer T‑Zell-Lymphome (PTCL) gemäß International Consensus Classification (ICC) und der Weltgesundheitsorganisation-Klassifikation (5. Auflage, WHO-HAEM5) sowie auf den daraus folgenden Auswirkungen auf die praktische Diagnostik. Die Veränderungen beruhen auf aktuellen Fortschritten in der genomischen und molekularen Charakterisierung der PTCL und einem vertieften Verständnis ihrer Pathobiologie. Insbesondere werden die folgenden Krankheitsgruppen berücksichtigt: Epstein-Barr-Virus(EBV)-assoziierte Neoplasien, follikuläres T‑Helfer-Zell-Lymphom, anaplastische großzellige Lymphome, primäre intestinale T- und NK-Zell-Lymphome sowie lymphoproliferative Erkrankungen, außerdem nicht anderweitig spezifizierte PTCL.
    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
    肺腺癌(LUAD)是肺癌最常见的亚型,在临床上表现出高度的生物学异质性和独特的临床结果。LUAD病因的当前范式将肺泡上皮II型(AT2)细胞作为起源的原代细胞,而AT1细胞在LUAD肿瘤发生中的作用尚不清楚。这里,我们通过致癌KRASG12D检查了含有2(Gramd2)AT1细胞的小鼠革兰氏结构域的致癌转化。AT1细胞中KRASG12D的激活诱导多灶性LUAD,主要是乳头状组织学。此外,在AT2和AT1衍生的LUAD中均观察到KRT8中间细胞状态,但是SCGB3A2+,另一个中间细胞标记,主要与AT1细胞有关,提示肿瘤进化的不同机制。总的来说,我们的研究表明,Gramd2+AT1细胞可以作为LUAD的起源细胞,并提示在治疗方法的开发中考虑基于起源细胞的LUAD的不同亚型.
    Lung adenocarcinoma (LUAD) is the most prevalent subtype of lung cancer and presents clinically with a high degree of biological heterogeneity and distinct clinical outcomes. The current paradigm of LUAD etiology posits alveolar epithelial type II (AT2) cells as the primary cell of origin, while the role of AT1 cells in LUAD oncogenesis remains unknown. Here, we examine oncogenic transformation in mouse Gram-domain containing 2 (Gramd2)+ AT1 cells via oncogenic KRASG12D. Activation of KRASG12D in AT1 cells induces multifocal LUAD, primarily of papillary histology. Furthermore, KRT8+ intermediate cell states were observed in both AT2- and AT1-derived LUAD, but SCGB3A2+, another intermediate cell marker, was primarily associated with AT1 cells, suggesting different mechanisms of tumor evolution. Collectively, our study reveals that Gramd2+ AT1 cells can serve as a cell of origin for LUAD and suggests that distinct subtypes of LUAD based on cell of origin be considered in the development of therapeutics.
    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
    临床和分子证据表明,高级别浆液性卵巢癌(HGSOC)主要起源于输卵管,不是卵巢表面。然而,这种偏好的原因尚不清楚。我们的研究强调了输卵管上皮(FTE)和卵巢表面上皮(OSE)细胞之间的显着差异,为FTE作为HGSOC起源位点提供分子基础。FTE,与OSE不同,表现出增强的复制应力(RS),损坏的叉子修复受损,无效的G2/M检查点,和增加的致瘤性。BRCA1杂合性加剧了这些缺陷,导致RS抑制单倍体不足和侵袭性肿瘤表型。人和小鼠切片的检查显示RS标记53BP1主要在输卵管中积聚,特别是在纤维的末端。此外,绝经状态影响RS水平。我们的研究为FTE作为HGSOC的起源提供了机械原理,调查BRCA1杂合性的影响,并为瞄准早期HGSOC驾驶员奠定了基础。
    Clinical and molecular evidence indicates that high-grade serous ovarian cancer (HGSOC) primarily originates from the fallopian tube, not the ovarian surface. However, the reasons for this preference remain unclear. Our study highlights significant differences between fallopian tube epithelial (FTE) and ovarian surface epithelial (OSE) cells, providing the molecular basis for FTEs as site of origin of HGSOC. FTEs, unlike OSEs, exhibit heightened replication stress (RS), impaired repair of stalled forks, ineffective G2/M checkpoint, and increased tumorigenicity. BRCA1 heterozygosity exacerbates these defects, resulting in RS suppression haploinsufficiency and an aggressive tumor phenotype. Examination of human and mouse sections reveals buildup of the RS marker 53BP1 primarily in the fallopian tubes, particularly at the fimbrial ends. Furthermore, menopausal status influences RS levels. Our study provides a mechanistic rationale for FTE as the site of origin for HGSOC, investigates the impact of BRCA1 heterozygosity, and lays the groundwork for targeting early HGSOC drivers.
    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
    恶性脑肿瘤是高度异质性的肿瘤,在儿童和成人中预后差,发病率和死亡率都很高。癌症干细胞(CSC,也称为肿瘤起始细胞)模型指出肿瘤生长是由CSC子集驱动的。这个模型解释了脑肿瘤的一些临床观察,包括最初成功化疗和/或放疗后几乎不可避免的肿瘤复发和治疗抵抗。在过去的二十年里,脑CSCs的鉴定和表征策略有了显著改善,支持脑肿瘤新诊断和治疗策略的设计。相关研究揭示了CSCs在大脑中的新特性,包括它们的异质性和独特的免疫生物学,这为新的研究方向和潜在的治疗方法提供了机会。在这次审查中,我们总结了脑肿瘤中CSCs标志物和干性调节因子的最新知识。我们还全面描述了CSCs生态位和肿瘤微环境对脑肿瘤干性的影响,包括CSC和免疫系统之间的相互作用,并讨论CSCs在脑肿瘤治疗中的潜在应用。
    Malignant brain tumors are highly heterogeneous tumors with a poor prognosis and a high morbidity and mortality rate in both children and adults. The cancer stem cell (CSC, also named tumor-initiating cell) model states that tumor growth is driven by a subset of CSCs. This model explains some of the clinical observations of brain tumors, including the almost unavoidable tumor recurrence after initial successful chemotherapy and/or radiotherapy and treatment resistance. Over the past two decades, strategies for the identification and characterization of brain CSCs have improved significantly, supporting the design of new diagnostic and therapeutic strategies for brain tumors. Relevant studies have unveiled novel characteristics of CSCs in the brain, including their heterogeneity and distinctive immunobiology, which have provided opportunities for new research directions and potential therapeutic approaches. In this review, we summarize the current knowledge of CSCs markers and stemness regulators in brain tumors. We also comprehensively describe the influence of the CSCs niche and tumor microenvironment on brain tumor stemness, including interactions between CSCs and the immune system, and discuss the potential application of CSCs in brain-based therapies for the treatment of brain tumors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号