Lineage plasticity

  • 文章类型: Journal Article
    骨肉瘤(OS)是最常见的儿童原发性骨恶性肿瘤。一个有希望的新治疗靶点是SKP2,编码SCFE3泛素连接酶的底物识别因子,负责底物p27的泛素化和蛋白酶体降解,从而驱动细胞增殖。我们之前已经证明,在OS的免疫活性转基因小鼠模型中敲除Skp2改善了存活率,驱使细胞凋亡,并诱导肿瘤炎症。这里,我们应用单细胞RNA测序(scRNA-seq)来研究源自Osx-Cre驱动的Rb1和Trp53条件性敲除的原发性OS肿瘤。我们表明,鼠OS模型概括了在患者肿瘤中观察到的肿瘤异质性和微环境复杂性。我们进一步比较了该模型与具有Skp2功能破坏的OS模型:一个具有Skp2敲除,另一个具有Skp2-p27相互作用破坏(导致p27过表达)。我们发现T细胞耗竭的减少和干扰素激活的上调,随着复制和内质网相关的应激在Skp2破坏模型的证据,并显示干扰素诱导与OS患者生存率改善相关。此外,我们的scRNA-seq分析揭示了Skp2破坏的OS中转移相关基因特征的活性降低,我们通过在Skp2基因敲除小鼠中观察到肺转移明显减少来验证。最后,我们报道了几种在OS中逃避靶向Skp2的潜在机制,包括Myc目标的上调,DNA拷贝数扩增和过表达替代性E3连接酶基因,和潜在的替代谱系激活。这些对OS肿瘤生物学和Skp2功能的机制见解为新的,协同疗法,而数据和我们的综合分析可以作为进一步的大数据驱动的操作系统研究的公共资源。
    Osteosarcoma (OS) is the most common primary pediatric bone malignancy. One promising new therapeutic target is SKP2, encoding a substrate recognition factor of the SCF E3 ubiquitin ligase responsible for ubiquitination and proteasome degradation of substrate p27, thus driving cellular proliferation. We have shown previously that knockout of Skp2 in an immunocompetent transgenic mouse model of OS improved survival, drove apoptosis, and induced tumor inflammation. Here, we applied single-cell RNA-sequencing (scRNA-seq) to study primary OS tumors derived from Osx-Cre driven conditional knockout of Rb1 and Trp53. We showed that murine OS models recapitulate the tumor heterogeneity and microenvironment complexity observed in patient tumors. We further compared this model with OS models with functional disruption of Skp2: one with Skp2 knockout and the other with the Skp2-p27 interaction disrupted (resulting in p27 overexpression). We found reduction of T cell exhaustion and upregulation of interferon activation, along with evidence of replicative and endoplasmic reticulum-related stress in the Skp2 disruption models, and showed that interferon induction was correlated with improved survival in OS patients. Additionally, our scRNA-seq analysis uncovered decreased activities of metastasis-related gene signatures in the Skp2-disrupted OS, which we validated by observation of a strong reduction in lung metastasis in the Skp2 knockout mice. Finally, we report several potential mechanisms of escape from targeting Skp2 in OS, including upregulation of Myc targets, DNA copy number amplification and overexpression of alternative E3 ligase genes, and potential alternative lineage activation. These mechanistic insights into OS tumor biology and Skp2 function suggest novel targets for new, synergistic therapies, while the data and our comprehensive analysis may serve as a public resource for further big data-driven OS research.
    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:神经内分泌前列腺癌(NEPC)是前列腺癌的致死性亚群,其特征在于神经内分泌分化和雄激素受体(AR)信号传导丧失。越来越多的证据表明,细胞谱系可塑性对于NEPC疗法的失败至关重要。尽管研究表明神经转录因子PAX6参与了耐药性,其在NEPC中的具体作用尚不清楚。
    方法:通过生物信息学和免疫组织化学鉴定PAX6在NEPC中的表达。CCK8测定,集落形成试验,肿瘤球形成试验和细胞凋亡试验用于说明PAX6在体外进展中的关键作用。进行ChIP和双荧光素酶报告基因测定以确认PAX6启动子区域中AR的结合序列,以及STAT5A和MET启动子区域中PAX6的结合序列。对于体内验证,对代表NEPC亚型的异种移植模型进行病理分析,以验证PAX6在疾病进展中的重要作用.通过公共临床数据集和特定细胞系的转录组测序建立补充诊断。ATAC-seq用于检测特定细胞系的染色质可及性。
    结果:PAX6表达在NEPC中显著升高,并受AR信号的负调控。PAX6在非NEPC细胞中的激活导致NE转分化,而PAX6在NEPC细胞中的敲除抑制NEPC的发生和发展。重要的是,AR的缺失导致PAX6的表达增强,这通过MET/STAT5A信号通路重新编程前列腺癌细胞的谱系可塑性以发展NE表型.通过ATAC-seq,我们发现PAX6的高表达水平引起染色质可及性增强,主要通过H4K20me3的衰减,这通常会导致癌细胞染色质沉默。
    结论:这项研究揭示了一种新的神经转录因子PAX6可以驱动NEPC进展,并表明它可能作为NEPC治疗的潜在治疗靶点。
    BACKGROUND: Neuroendocrine prostate cancer (NEPC) is a lethal subset of prostate cancer which is characterized by neuroendocrine differentiation and loss of androgen receptor (AR) signaling. Growing evidence reveals that cell lineage plasticity is crucial in the failure of NEPC therapies. Although studies suggest the involvement of the neural transcription factor PAX6 in drug resistance, its specific role in NEPC remains unclear.
    METHODS: The expression of PAX6 in NEPC was identified via bioinformatics and immunohistochemistry. CCK8 assay, colony formation assay, tumorsphere formation assay and apoptosis assay were used to illustrate the key role of PAX6 in the progression of in vitro. ChIP and Dual-luciferase reporter assays were conducted to confirm the binding sequences of AR in the promoter region of PAX6, as well as the binding sequences of PAX6 in the promoter regions of STAT5A and MET. For in vivo validation, the xenograft model representing NEPC subtype underwent pathological analysis to verify the significant role of PAX6 in disease progression. Complementary diagnoses were established through public clinical datasets and transcriptome sequencing of specific cell lines. ATAC-seq was used to detect the chromatin accessibility of specific cell lines.
    RESULTS: PAX6 expression was significantly elevated in NEPC and negatively regulated by AR signaling. Activation of PAX6 in non-NEPC cells led to NE trans-differentiation, while knock-down of PAX6 in NEPC cells inhibited the development and progression of NEPC. Importantly, loss of AR resulted in an enhanced expression of PAX6, which reprogramed the lineage plasticity of prostate cancer cells to develop NE phenotypes through the MET/STAT5A signaling pathway. Through ATAC-seq, we found that a high expression level of PAX6 elicited enhanced chromatin accessibility, mainly through attenuation of H4K20me3, which typically causes chromatin silence in cancer cells.
    CONCLUSIONS: This study reveals a novel neural transcription factor PAX6 could drive NEPC progression and suggest that it might serve as a potential therapeutic target for the management of NEPC.
    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
    前列腺癌是世界上男性中第二常见的恶性肿瘤,对男性的健康和生命构成严重威胁。RB1是第一个被描述的人类肿瘤抑制基因,它与发展密切相关,programming,抑制多种肿瘤。研究发现,RB1丢失是前列腺癌发生的早期事件,与前列腺癌的发生密切相关。进展和治疗抵抗。本文从RB1与前列腺细胞谱系可塑性、生物学行为、治疗抗性三个方面综述了RB1与前列腺癌关系的研究现状。为开发RB1丢失前列腺癌的新治疗策略提供了新的视角。
    Prostate cancer is the second most common malignancy among men in the world, posing a serious threat to men\'s health and lives. RB1 is the first human tumor suppressor gene to be described, and it is closely associated with the development, progression, and suppression of a variety of tumors. It was found that the loss of RB1 is an early event in prostate cancer development and is closely related to prostate cancer development, progression and treatment resistance. This paper reviews the current status of research on the relationship between RB1 and prostate cancer from three aspects: RB1 and prostate cell lineage plasticity; biological behavior; and therapeutic resistance. Providing a novel perspective for developing new therapeutic strategies for RB1-loss prostate cancer.
    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
    代谢重编程是癌症发展的关键,尽管丙酮酸激酶M2(PKM2)缺乏和肿瘤糖酵解,但维持三阴性乳腺癌(TNBC)细胞生长的机制仍有待确定.这里,我们发现肿瘤糖酵解的缺乏会激活从糖酵解到脂肪酸β-氧化(FAO)的代谢转换,从而促进TNBC的生长。我们证明,在TNBC细胞中,PKM2直接与组蛋白甲基转移酶EZH2相互作用,协调介导肉碱转运蛋白的表观遗传沉默,SLC16A9.抑制PKM2会导致SLC16A9的EZH2募集受损,进而抑制SLC16A9表达以增加细胞内肉碱流入,将TNBC细胞编程为FAO依赖性和管腔样细胞状态。一起,这些发现揭示了一种新的代谢开关,将TNBC从代谢异质性谱系塑料细胞状态驱动到FAO依赖的谱系定型细胞状态,其中EZH2和FAO的双重靶向在TNBC中诱导有效的合成致死性。
    Metabolic reprogramming is key for cancer development, yet the mechanism that sustains triple-negative breast cancer (TNBC) cell growth despite deficient pyruvate kinase M2 (PKM2) and tumor glycolysis remains to be determined. Here, we find that deficiency in tumor glycolysis activates a metabolic switch from glycolysis to fatty acid β-oxidation (FAO) to fuel TNBC growth. We show that, in TNBC cells, PKM2 directly interacts with histone methyltransferase EZH2 to coordinately mediate epigenetic silencing of a carnitine transporter, SLC16A9. Inhibition of PKM2 leads to impaired EZH2 recruitment to SLC16A9, and in turn de-represses SLC16A9 expression to increase intracellular carnitine influx, programming TNBC cells to an FAO-dependent and luminal-like cell state. Together, these findings reveal a new metabolic switch that drives TNBC from a metabolically heterogeneous-lineage plastic cell state to an FAO-dependent-lineage committed cell state, where dual targeting of EZH2 and FAO induces potent synthetic lethality in TNBC.
    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
    从腺癌到小细胞神经内分泌状态的跨分化与多种癌症类型的治疗抗性相关。为了深入了解转分化的潜在分子事件,我们对泛小细胞神经内分泌癌模型(称为PARCB)进行了多组学时程分析,使用人类前列腺基底细胞进行正向遗传转化,并确定共同的发育,弧形,和所有转换模型复制中的高熵轨迹。使用单细胞分辨率的进一步映射揭示了由ASCL1或ASCL2的互斥表达定义的两个不同的谱系。跨发育阶段的转录因子组的时间调节表明,细胞重编程先于神经元程序的诱导。TFAP4和ASCL1/2反馈被认为是ASCL1和ASCL2表达的潜在调节因子。我们的研究提供了时间转录模式,并揭示了前列腺癌和肺癌之间的泛组织相似性,以及与正常神经内分泌细胞状态的联系。
    Trans-differentiation from an adenocarcinoma to a small cell neuroendocrine state is associated with therapy resistance in multiple cancer types. To gain insight into the underlying molecular events of the trans-differentiation, we perform a multi-omics time course analysis of a pan-small cell neuroendocrine cancer model (termed PARCB), a forward genetic transformation using human prostate basal cells and identify a shared developmental, arc-like, and entropy-high trajectory among all transformation model replicates. Further mapping with single cell resolution reveals two distinct lineages defined by mutually exclusive expression of ASCL1 or ASCL2. Temporal regulation by groups of transcription factors across developmental stages reveals that cellular reprogramming precedes the induction of neuronal programs. TFAP4 and ASCL1/2 feedback are identified as potential regulators of ASCL1 and ASCL2 expression. Our study provides temporal transcriptional patterns and uncovers pan-tissue parallels between prostate and lung cancers, as well as connections to normal neuroendocrine cell states.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    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
    表皮生长因子受体(EGFR)突变的非小细胞肺癌(NSCLC)在临床和遗传上是异质性的,同时发生RB1/TP53突变,表明转化为小细胞肺癌(SCLC)的风险增加。当肿瘤细胞转化为不同的组织学亚型时,他们失去了对原始致癌驱动因素的依赖,导致治疗抗性。然而,与这种转化相关的分子细节仍不清楚.由于缺乏转化前和转化后的临床样品,很难定义肺癌中神经内分泌(NE)转化的分子机制。在这项研究中,我们建立了同时有RB1/TP53突变的NSCLC细胞系,并建立了相应的患者源性异种移植(PDX)模型,以研究转化为SCLC的机制.研究这些PDX模型,我们证明,EGFR缺失促进了由TP53和RB1双等位基因突变引发的肺腺癌的谱系可塑性.这些EGFR敲除肿瘤的基因表达分析显示神经内分泌突触相关谱系基因的表达改变。在这些EGFR敲除肿瘤中,表观遗传重编程因子如Sox2和与神经发育相关的基因如NTRK的表达增加。这些发现揭示了EGFR在可塑性获得中的作用,这是细胞大幅改变其身份并呈现新表型的能力,并定义了肺癌中NE转化的潜在驱动因素的新图景。
    Epidermal growth factor receptor (EGFR)-mutant non-small-cell lung cancer (NSCLC) is clinically and genetically heterogeneous, with concurrent RB1/TP53 mutations, indicating an increased risk of transformation into small cell lung cancer (SCLC). When tumor cells convert into a different histological subtype, they lose their dependence on the original oncogenic driver, resulting in therapeutic resistance. However, the molecular details associated with this transformation remain unclear. It has been difficult to define molecular mechanisms of neuroendocrine (NE) transformation in lung cancer due to a lack of pre- and post-transformation clinical samples. In this study, we established a NSCLC cell line with concurrent RB1/TP53 mutations and built corresponding patient-derived xenograft (PDX) models to investigate the mechanisms underlying transformation to SCLC. Studying these PDX models, we demonstrate that EGFR loss facilitates lineage plasticity of lung adenocarcinoma initiated by biallelic mutations of TP53 and RB1. Gene expression analysis of these EGFR knockout tumors revealed altered expression of neuroendocrine synapse-associated lineage genes. There is an increased expression of epigenetic reprogramming factors like Sox2 and gene associated with neural development like NTRK in these EGFR knockout tumors. These findings uncovered the role of EGFR in the acquisition of plasticity, which is the ability of a cell to substantially modify its identity and take on a new phenotype, and defined a novel landscape of potential drivers of NE transformation in lung cancer.
    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
    胎儿肝脏造血干细胞(HSC)中的PTEN丢失导致骨髓改变,T-,和B谱系潜能和T谱系急性淋巴细胞白血病(T-ALL)的发展。为了探索PTEN调节的造血谱系选择的潜在机制,我们使用测序(ATAC-seq)对转座酶可接近的染色质进行整合测定,单细胞RNA-seq,使用体内分离的小鼠白血病前HSC和祖细胞进行体外培养分析。我们发现PTEN缺失会改变前B阶段关键谱系转录因子(TF)结合位点的染色质可及性,对应于增加的骨髓和T谱系电位和降低的B谱系电位。重要的是,我们发现PU.1是PTEN下游的重要TF,改变PU.1水平可以重新编程染色质可及性景观和骨髓,T-,和Ptennullprepro-B细胞中的B谱系电位。我们的研究发现prepro-B是PTEN调节的造血谱系选择的关键发育阶段,并表明PU.1在调节prepro-B祖细胞的表观遗传状态和谱系可塑性中的关键作用。
    PTEN loss in fetal liver hematopoietic stem cells (HSCs) leads to alterations in myeloid, T-, and B-lineage potentials and T-lineage acute lymphoblastic leukemia (T-ALL) development. To explore the mechanism underlying PTEN-regulated hematopoietic lineage choices, we carry out integrated assay for transposase-accessible chromatin using sequencing (ATAC-seq), single-cell RNA-seq, and in vitro culture analyses using in vivo-isolated mouse pre-leukemic HSCs and progenitors. We find that PTEN loss alters chromatin accessibility of key lineage transcription factor (TF) binding sites at the prepro-B stage, corresponding to increased myeloid and T-lineage potentials and reduced B-lineage potential. Importantly, we find that PU.1 is an essential TF downstream of PTEN and that altering PU.1 levels can reprogram the chromatin accessibility landscape and myeloid, T-, and B-lineage potentials in Ptennull prepro-B cells. Our study discovers prepro-B as the key developmental stage underlying PTEN-regulated hematopoietic lineage choices and suggests a critical role of PU.1 in modulating the epigenetic state and lineage plasticity of prepro-B progenitors.
    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
    基于顺铂的化疗仍然是不可切除和转移性肌肉浸润性膀胱癌(MIBCs)的主要治疗方法。然而,肿瘤经常发展化疗耐药。这里,我们用基因编辑的类器官建立了原发性和原位MIBC小鼠模型,以概括患者化疗的整个过程.我们发现部分鳞状分化,称为半空间化,与小鼠和人类MIBCs的获得性化学抗性有关。多组学分析表明,组织蛋白酶H(CTSH)与化学抗性和半污损化相关。E64治疗抑制组织蛋白酶诱导完全鳞状细胞分化和焦亡,并因此特别抑制化学抗性MIBCs。机械上,E64治疗激活肿瘤坏死因子途径,这对于化学抗性MIBC细胞的终末分化和焦亡是必需的。我们的研究表明,半菌化是一种与化学抗性相关的谱系可塑性,提示通过靶向CTSH进行分化是治疗化疗耐药MIBCs的潜在治疗策略。
    Cisplatin-based chemotherapy remains the primary treatment for unresectable and metastatic muscle-invasive bladder cancers (MIBCs). However, tumors frequently develop chemoresistance. Here, we established a primary and orthotopic MIBC mouse model with gene-edited organoids to recapitulate the full course of chemotherapy in patients. We found that partial squamous differentiation, called semi-squamatization, is associated with acquired chemoresistance in both mice and human MIBCs. Multi-omics analyses showed that cathepsin H (CTSH) is correlated with chemoresistance and semi-squamatization. Cathepsin inhibition by E64 treatment induces full squamous differentiation and pyroptosis, and thus specifically restrains chemoresistant MIBCs. Mechanistically, E64 treatment activates the tumor necrosis factor pathway, which is required for the terminal differentiation and pyroptosis of chemoresistant MIBC cells. Our study revealed that semi-squamatization is a type of lineage plasticity associated with chemoresistance, suggesting that differentiation via targeting of CTSH is a potential therapeutic strategy for the treatment of chemoresistant MIBCs.
    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
    肿瘤微环境(TME)是由肿瘤和间充质细胞以及细胞外基质组成的微生态。TME在肿瘤增殖中起着重要的调节作用,入侵,转移,和差异化。神经内分泌分化(NED)是晚期前列腺癌(PCa)中去势抵抗发展的机制。雄激素剥夺治疗后诱发NED,最终建立神经内分泌前列腺癌(NEPC)。NEPC预后差,总生存期短,是PCa患者死亡的主要原因。TME的细胞和非细胞组分都通过各种途径调节和诱导NEPC形成。洞察TME在NEPC演变中的作用,增长,在过去的几年里,进展有所增加。这些新颖的见解将有助于完善NEPC形成模型,并为发现针对TME的新NEPC疗法奠定基础。
    The tumor microenvironment (TME) is a microecology consisting of tumor and mesenchymal cells and extracellular matrices. The TME plays important regulatory roles in tumor proliferation, invasion, metastasis, and differentiation. Neuroendocrine differentiation (NED) is a mechanism by which castration resistance develops in advanced prostate cancer (PCa). NED is induced after androgen deprivation therapy and neuroendocrine prostate cancer (NEPC) is established finally. NEPC has poor prognosis and short overall survival and is a major cause of death in patients with PCa. Both the cellular and non-cellular components of the TME regulate and induce NEPC formation through various pathways. Insights into the roles of the TME in NEPC evolution, growth, and progression have increased over the past few years. These novel insights will help refine the NEPC formation model and lay the foundation for the discovery of new NEPC therapies targeting the TME.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号