Lineage plasticity

  • 文章类型: Journal Article
    使用放射性配体和基于抗体的疗法靶向细胞表面分子已经在癌症中取得了相当大的成功。然而,目前尚不清楚推定谱系标记的表达,特别是细胞表面分子,在谱系可塑性的过程中变化,其中肿瘤细胞改变其身份并获得新的致癌特性。谱系可塑性的一个值得注意的例子是前列腺腺癌(PRAD)向神经内分泌前列腺癌(NEPC)的转化,这是一种不断增长的抗性机制,导致对雄激素阻断的反应性丧失,并预示着患者生存率低下。为了了解谱系标记在前列腺癌谱系可塑性的演变过程中如何变化,我们将单细胞分析应用于21个人类前列腺肿瘤活检和两个基因工程小鼠模型,连同131个肿瘤样本的组织微阵列分析。我们不仅在去势抗性PRAD和NEPC中观察到比以前预期的更高程度的表型异质性,而且还发现,即PSMA,STEAP1、STEAP2、TROP2、CEACAM5和DLL3在基因调节网络(GRN)的一个子集内变化。我们还注意到NEPC和小细胞肺癌亚型共享一组GRN,表明保守的生物学途径可以在不同类型的肿瘤中进行治疗。虽然这种极端水平的转录异质性,特别是在细胞表面标记表达中,可能会减轻对当前和未来抗原导向疗法的临床反应的持久性,其轮廓可能会在临床试验中产生患者选择的特征,可能跨越不同的癌症类型。
    Targeting cell surface molecules using radioligand and antibody-based therapies has yielded considerable success across cancers. However, it remains unclear how the expression of putative lineage markers, particularly cell surface molecules, varies in the process of lineage plasticity, wherein tumor cells alter their identity and acquire new oncogenic properties. A notable example of lineage plasticity is the transformation of prostate adenocarcinoma (PRAD) to neuroendocrine prostate cancer (NEPC)-a growing resistance mechanism that results in the loss of responsiveness to androgen blockade and portends dismal patient survival. To understand how lineage markers vary across the evolution of lineage plasticity in prostate cancer, we applied single-cell analyses to 21 human prostate tumor biopsies and two genetically engineered mouse models, together with tissue microarray analysis on 131 tumor samples. Not only did we observe a higher degree of phenotypic heterogeneity in castrate-resistant PRAD and NEPC than previously anticipated but also found that the expression of molecules targeted therapeutically, namely PSMA, STEAP1, STEAP2, TROP2, CEACAM5, and DLL3, varied within a subset of gene-regulatory networks (GRNs). We also noted that NEPC and small cell lung cancer subtypes shared a set of GRNs, indicative of conserved biologic pathways that may be exploited therapeutically across tumor types. While this extreme level of transcriptional heterogeneity, particularly in cell surface marker expression, may mitigate the durability of clinical responses to current and future antigen-directed therapies, its delineation may yield signatures for patient selection in clinical trials, potentially across distinct cancer types.
    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
    肺癌是癌症死亡的主要原因,包括两种主要类型:非小细胞肺癌(NSCLC)和小细胞肺癌(SCLC)。占病例的85%和15%,分别。通常用分子靶向单个癌基因的酪氨酸激酶抑制剂(TKIs)治疗具有可操作驱动突变的非小细胞肺癌。尽管TKIs极大地促进了更好的临床结果,对它们的抵抗不可避免地发生。组织学或谱系转化是一种罕见但有据可查的与获得性抗性相关的脱靶机制。并且在使用多种不同TKIs和其他药物治疗后的环境中被鉴定。它包括神经内分泌转化,鳞状细胞转化,和上皮-间质转化。这里,我们回顾了转化肿瘤的临床病理特征,以及对NSCLC谱系转化的关键遗传改变和生物学机制的最新认识。特别是TKI触发的转换。
    Lung cancer is the leading cause of cancer death and includes two major types: non-small-cell lung cancer (NSCLC) and small-cell lung cancer (SCLC), accounting for 85% and 15% of cases, respectively. Non-small-cell lung cancer harboring actionable driver mutations is generally treated with tyrosine kinase inhibitors (TKIs) molecularly targeting individual oncogenes. Although TKIs have greatly contributed to better clinical outcomes, acquired resistance to them inevitably occurs. Histologic or lineage transformation is a rare but well-documented off-target mechanism associated with acquired resistance, and has been identified in settings following treatment with multiple different TKIs and other drugs. It includes neuroendocrine transformation, squamous cell transformation, and epithelial-to-mesenchymal transition. Here, we review the clinicopathologic features of transformed tumors and current understanding of the key genetic alterations and biologic mechanism of lineage transformation in NSCLC, particularly TKI-triggered transformation.
    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
    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
    免疫系统在其形成的记忆细胞的数量和类型中编码有关致病威胁的严重性的信息。这种编码来自淋巴细胞决定在攻击期间维持或失去自我更新和记忆潜力。通过使用时间分辨转录组学在单细胞和克隆谱系水平跟踪CD8+T细胞,定量实时成像,和急性感染模型,我们发现T细胞会在抗原识别后早期维持或丧失记忆潜能。然而,病原体清除后,如果最初丢失,T细胞可能会恢复记忆潜能。机械上,这种灵活性是通过一个随机的顺式-表观遗传开关来实现的,该开关可调谐和可逆地使记忆调节器沉默,TCF1,对刺激的反应。数学建模显示了这种灵活性如何使记忆T细胞数量随着病原体毒力和免疫应答幅度而稳健地扩展。我们建议细胞决策的灵活性和随机性确保针对各种威胁的最佳免疫反应。
    The immune system encodes information about the severity of a pathogenic threat in the quantity and type of memory cells it forms. This encoding emerges from lymphocyte decisions to maintain or lose self-renewal and memory potential during a challenge. By tracking CD8+ T cells at the single-cell and clonal lineage level using time-resolved transcriptomics, quantitative live imaging, and an acute infection model, we find that T cells will maintain or lose memory potential early after antigen recognition. However, following pathogen clearance, T cells may regain memory potential if initially lost. Mechanistically, this flexibility is implemented by a stochastic cis-epigenetic switch that tunably and reversibly silences the memory regulator, TCF1, in response to stimulation. Mathematical modeling shows how this flexibility allows memory T cell numbers to scale robustly with pathogen virulence and immune response magnitudes. We propose that flexibility and stochasticity in cellular decisions ensure optimal immune responses against diverse threats.
    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
    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
    肺鳞状细胞癌(LUSC)与高死亡率和有限的靶向治疗相关。USP13是LUSC中扩增最多的基因之一,然而,它在肺癌中的作用在很大程度上是未知的。这里,我们通过在KrasG12D/+;Trp53flox/flox背景(KPU)上过度表达USP13,建立了一种新型的LUSC小鼠模型。KPU驱动的肺鳞状肿瘤忠实地概括了关键的病理组织学,分子特征,和人类LUSC的细胞途径。我们发现USP13改变了气道俱乐部细胞中的谱系决定因子,例如NKX2-1和SOX2,并增强了俱乐部细胞向鳞状细胞发展的命运。我们显示了USP13和c-MYC之间的强分子关联,导致鼠和人肺癌细胞鳞状细胞程序的上调。总的来说,我们的数据表明,USP13是俱乐部细胞谱系可塑性的分子驱动因素,并提供了可能对LUSC治疗有潜在影响的机制见解.
    Lung squamous cell carcinoma (LUSC) is associated with high mortality and limited targeted therapies. USP13 is one of the most amplified genes in LUSC, yet its role in lung cancer is largely unknown. Here, we established a novel mouse model of LUSC by overexpressing USP13 on KrasG12D/+; Trp53flox/flox background (KPU). KPU-driven lung squamous tumors faithfully recapitulate key pathohistological, molecular features, and cellular pathways of human LUSC. We found that USP13 altered lineage-determining factors such as NKX2-1 and SOX2 in club cells of the airway and reinforced the fate of club cells to squamous carcinoma development. We showed a strong molecular association between USP13 and c-MYC, leading to the upregulation of squamous programs in murine and human lung cancer cells. Collectively, our data demonstrate that USP13 is a molecular driver of lineage plasticity in club cells and provide mechanistic insight that may have potential implications for the treatment of LUSC.
    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
    炎症对于组织稳态的破坏是必不可少的,并且可以破坏谱系定型上皮细胞的身份。这里,我们使用谱系追踪小鼠模型,单细胞转录组学和染色质分析,和CUT&TAG来鉴定胰腺腺泡细胞区室炎症损伤的表观遗传记忆。尽管胰腺炎消退,我们的数据显示腺泡细胞无法恢复到它们的分子基线,随着染色质可及性升高和H3K4me1保留在化生基因,这样记忆代表了一个不完整的细胞命运决定。在体内,我们发现这种表观遗传记忆控制着谱系可塑性,对第二次损伤的反应,化生减少,但伴随致癌Kras突变的肿瘤发生增加。降低了致癌转化的阈值,反过来,可以通过阻断MAPK信号来恢复。一起,我们定义染色质动力学,分子编码,和回忆炎症损伤的长期表观遗传记忆,影响未来的反应,但仍然是可逆的。
    Inflammation is essential to the disruption of tissue homeostasis and can destabilize the identity of lineage-committed epithelial cells. Here, we employ lineage-traced mouse models, single-cell transcriptomic and chromatin analyses, and CUT&TAG to identify an epigenetic memory of inflammatory injury in the pancreatic acinar cell compartment. Despite resolution of pancreatitis, our data show that acinar cells fail to return to their molecular baseline, with retention of elevated chromatin accessibility and H3K4me1 at metaplasia genes, such that memory represents an incomplete cell fate decision. In vivo, we find this epigenetic memory controls lineage plasticity, with diminished metaplasia in response to a second insult but increased tumorigenesis with an oncogenic Kras mutation. The lowered threshold for oncogenic transformation, in turn, can be restored by blockade of MAPK signaling. Together, we define the chromatin dynamics, molecular encoding, and recall of a prolonged epigenetic memory of inflammatory injury that impacts future responses but remains reversible.
    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
    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)

公众号