Cell plasticity

细胞可塑性
  • 文章类型: Journal Article
    在几种癌症中,包括肝细胞癌,已经证明,与分化的癌细胞相比,癌症干细胞(CSC)具有增强的侵袭力和治疗抗性.数学计算工具对于整合实验结果和理解CSC出现的表型可塑性机制可能是有价值的。在文献综述的基础上,我们构建了一个布尔模型,该模型恢复了八个稳定状态(吸引子),对应于衰老期肝细胞和间充质细胞的基因表达谱,静止,增殖性,和类似茎的状态。分析了与调控网络相关的表观遗传景观。我们观察到p53,p16,RB的丢失,或β-连环蛋白和YAP1的组成型激活增加了增殖茎样表型的稳健性。此外,我们发现p53失活促进了增殖肝细胞向干细胞样间充质表型的转化.因此,表型可塑性可能会改变,与CSC相关的茎样表型可能在突变获得后更容易获得。
    In several carcinomas, including hepatocellular carcinoma, it has been demonstrated that cancer stem cells (CSCs) have enhanced invasiveness and therapy resistance compared to differentiated cancer cells. Mathematical-computational tools could be valuable for integrating experimental results and understanding the phenotypic plasticity mechanisms for CSCs emergence. Based on the literature review, we constructed a Boolean model that recovers eight stable states (attractors) corresponding to the gene expression profile of hepatocytes and mesenchymal cells in senescent, quiescent, proliferative, and stem-like states. The epigenetic landscape associated with the regulatory network was analyzed. We observed that the loss of p53, p16, RB, or the constitutive activation of β-catenin and YAP1 increases the robustness of the proliferative stem-like phenotypes. Additionally, we found that p53 inactivation facilitates the transition of proliferative hepatocytes into stem-like mesenchymal phenotype. Thus, phenotypic plasticity may be altered, and stem-like phenotypes related to CSCs may be easier to attain following the mutation acquisition.
    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
    炎性肿瘤微环境(TME)是肿瘤促进过程的关键驱动因素。肿瘤相关巨噬细胞是TME中主要的免疫细胞类型之一,其密度增加与前列腺癌的不良预后有关。这里,我们研究了促炎(M1)和免疫抑制(M2)巨噬细胞对前列腺癌谱系可塑性的影响.我们的发现表明,M1巨噬细胞分泌因子上调与干细胞相关的基因,同时下调与前列腺癌细胞雄激素反应相关的基因。肿瘤干细胞(CSC)可塑性标志物NANOG的表达,来自M1巨噬细胞的分泌因子刺激KLF4、S0X2、OCT4和CD44。此外,在用M1巨噬细胞分泌因子处理的LNCaP细胞中观察到AR及其靶基因PSA被抑制。使用IKK16抑制剂抑制NFκB信号导致NANOG下调,SOX2、CD44和CSC可塑性。我们的研究强调,M1巨噬细胞的分泌因子通过NFκB信号通路上调CSC可塑性标志物的表达来驱动前列腺癌细胞的可塑性。
    The inflammatory tumor microenvironment (TME) is a key driver for tumor-promoting processes. Tumor-associated macrophages are one of the main immune cell types in the TME and their increased density is related to poor prognosis in prostate cancer. Here, we investigated the influence of pro-inflammatory (M1) and immunosuppressive (M2) macrophages on prostate cancer lineage plasticity. Our findings reveal that M1 macrophage secreted factors upregulate genes related to stemness while downregulating genes associated with androgen response in prostate cancer cells. The expression of cancer stem cell (CSC) plasticity markers NANOG, KLF4, SOX2, OCT4, and CD44 was stimulated by the secreted factors from M1 macrophages. Moreover, AR and its target gene PSA were observed to be suppressed in LNCaP cells treated with secreted factors from M1 macrophages. Inhibition of NFκB signaling using the IKK16 inhibitor resulted in downregulation of NANOG, SOX2, and CD44 and CSC plasticity. Our study highlights that the secreted factors from M1 macrophages drive prostate cancer cell plasticity by upregulating the expression of CSC plasticity markers through NFκB signaling pathway.
    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
    肿瘤是一种复杂且侵袭性的疾病,对健康构成重大挑战。了解其进展背后的细胞机制对于开发有效的治疗方法至关重要。在这项研究中,我们开发了一个新的数学框架来研究细胞可塑性和异质性在肿瘤进展中的作用。通过利用时间单细胞数据,我们提出了一个反应-对流-扩散模型,该模型可以有效地捕获肿瘤微环境中肿瘤细胞和巨噬细胞的时空动力学。通过理论分析,我们获得了脉搏波速度的估计,并分析了齐次稳态解的稳定性。值得注意的是,我们使用AddModuleScore函数来量化细胞可塑性。我们方法的亮点之一是引入脉搏波速度作为一种定量测量,以精确测量细胞表型转变的速率,以及高可塑性细胞状态/低可塑性细胞状态比率作为肿瘤恶性指标的新实现。此外,分叉分析揭示了肿瘤细胞群的复杂动力学。我们广泛的分析表明,表型转变率的增加与恶性程度的增加有关。归因于肿瘤探索更宽表型空间的能力。该研究还调查了肿瘤细胞的增殖率和死亡率,表型对流速度,表型转变阶段的中点影响肿瘤细胞表型转变的速度和向腺癌的进展。这些见解和定量措施可以帮助指导靶向治疗策略的发展,以有效调节细胞可塑性和控制肿瘤进展。
    Tumor is a complex and aggressive type of disease that poses significant health challenges. Understanding the cellular mechanisms underlying its progression is crucial for developing effective treatments. In this study, we develop a novel mathematical framework to investigate the role of cellular plasticity and heterogeneity in tumor progression. By leveraging temporal single-cell data, we propose a reaction-convection-diffusion model that effectively captures the spatiotemporal dynamics of tumor cells and macrophages within the tumor microenvironment. Through theoretical analysis, we obtain the estimate of the pulse wave speed and analyze the stability of the homogeneous steady state solutions. Notably, we employe the AddModuleScore function to quantify cellular plasticity. One of the highlights of our approach is the introduction of pulse wave speed as a quantitative measure to precisely gauge the rate of cell phenotype transitions, as well as the novel implementation of the high-plasticity cell state/low-plasticity cell state ratio as an indicator of tumor malignancy. Furthermore, the bifurcation analysis reveals the complex dynamics of tumor cell populations. Our extensive analysis demonstrates that an increased rate of phenotype transition is associated with heightened malignancy, attributable to the tumor\'s ability to explore a wider phenotypic space. The study also investigates how the proliferation rate and the death rate of tumor cells, phenotypic convection velocity, and the midpoint of the phenotype transition stage affect the speed of tumor cell phenotype transitions and the progression to adenocarcinoma. These insights and quantitative measures can help guide the development of targeted therapeutic strategies to regulate cellular plasticity and control tumor progression effectively.
    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
    这篇综合综述旨在强调研究AHCYL1(也称为IRBIT)的功能作用的重要性,在癌细胞中。最近有人提出AHCYL1调节细胞存活/死亡,干性能力,以及宿主对肿瘤微环境的适应性反应。尽管有这些知识,AHCYL1在癌症中的作用仍然存在争议,可能是由于其能够以组织特异性方式与多种因素相互作用。了解调节肿瘤和控制AHCYL1表达的肿瘤微环境之间的功能相互作用的机制可以提供对肿瘤发展调节的更深入的理解。解决AHCYL1如何在肿瘤环境中调节细胞可塑性过程可能与开发癌症生物学中的翻译方法有关。
    This integrative review aims to highlight the importance of investigating the functional role of AHCYL1, also known as IRBIT, in cancer cells. It has recently been suggested that AHCYL1 regulates cell survival/death, stemness capacity, and the host adaptive response to the tumor microenvironment. Despite this knowledge, the role of AHCYL1 in cancer is still controversial, probably due to its ability to interact with multiple factors in a tissue-specific manner. Understanding the mechanisms regulating the functional interplay between the tumor and the tumor microenvironment that controls the expression of AHCYL1 could provide a deeper comprehension of the regulation of tumor development. Addressing how AHCYL1 modulates cellular plasticity processes in a tumoral context is potentially relevant to developing translational approaches in cancer biology.
    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
    对内分泌治疗的获得性抗性仍然是主要的临床挑战。在这项研究中,我们发现desmoglein-2(DSG2)在ER+乳腺癌(BC)的获得性内分泌抵抗和细胞可塑性中起主要作用。通过使用单细胞RNA-seq分析完善的氟维司群抗性ER+BC模型,我们发现,ER抑制导致癌细胞群中DSG2的特异性增加,这反过来又增强了桥粒在体外和体内的形成和细胞表型可塑性,促进对治疗的抗性。DSG2耗竭减少氟维司群耐药异种移植模型中的肿瘤发生和转移,并提高氟维司群效率。机械上,DSG2与JUP和波形蛋白形成桥粒复合体并触发Wnt/PCP信号传导。我们发现DSG2水平升高,随着ER水平的降低和Wnt/PCP通路的激活,预测生存不佳,这表明DSG2high签名可用于治疗干预。我们的分析强调了抗雌激素治疗后DSG2介导的桥粒连接的关键作用。
    Acquired resistance to endocrine treatments remains a major clinical challenge. In this study, we found that desmoglein-2 (DSG2) plays a major role in acquired endocrine resistance and cellular plasticity in ER+ breast cancer (BC). By analysing the well-established fulvestrant-resistant ER+ BC model using single-cell RNA-seq, we revealed that ER inhibition leads to a specific increase in DSG2 in cancer cell populations, which in turn enhances desmosome formation in vitro and in vivo and cell phenotypic plasticity that promotes resistance to treatment. DSG2 depletion reduced tumorigenesis and metastasis in fulvestrant-resistant xenograft models and promoted fulvestrant efficiency. Mechanistically, DSG2 forms a desmosome complex with JUP and Vimentin and triggers Wnt/PCP signalling. We showed that elevated DSG2 levels, along with reduced ER levels and an activated Wnt/PCP pathway, predicted poor survival, suggesting that a DSG2high signature could be exploited for therapeutic interventions. Our analysis highlighted the critical role of DSG2-mediated desmosomal junctions following antiestrogen 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
    非遗传机制最近已成为抗癌药物抗性的重要驱动因素。其中,药物耐受耐受者(DTP)细胞表型越来越受到关注,并在癌症治疗耐药中发挥主要的非遗传作用。DTP表型的特征是癌细胞亚群的静止或慢细胞周期可逆状态以及对刺激的惰性特化。通过多种潜在机制的相互作用,在一定程度上耐受抗癌药物的暴露,并在停药后恢复生长和增殖,最终导致治疗耐药和癌症复发。因此,靶向DTP细胞有望为癌症患者提供新的治疗机会,尽管我们目前对这些DTP细胞治疗耐药性的了解仍然有限。在这次审查中,我们提供了DTP细胞的形成特征和潜在的药物耐受机制的全面概述,调查DTP的潜在药物(包括临床前药物,旧药物的新用途,和天然产物)基于不同的药物模型,探讨抗DTP治疗的必要性和可行性,相关申请表,以及未来需要解决的问题,以将这一新兴领域推向临床应用。尽管如此,了解DTP细胞的新功能可能使我们能够开发新的更有效的抗癌疗法,并改善癌症患者的临床预后。
    Non-genetic mechanisms have recently emerged as important drivers of anticancer drug resistance. Among these, the drug tolerant persister (DTP) cell phenotype is attracting more and more attention and giving a predominant non-genetic role in cancer therapy resistance. The DTP phenotype is characterized by a quiescent or slow-cell-cycle reversible state of the cancer cell subpopulation and inert specialization to stimuli, which tolerates anticancer drug exposure to some extent through the interaction of multiple underlying mechanisms and recovering growth and proliferation after drug withdrawal, ultimately leading to treatment resistance and cancer recurrence. Therefore, targeting DTP cells is anticipated to provide new treatment opportunities for cancer patients, although our current knowledge of these DTP cells in treatment resistance remains limited. In this review, we provide a comprehensive overview of the formation characteristics and underlying drug tolerant mechanisms of DTP cells, investigate the potential drugs for DTP (including preclinical drugs, novel use for old drugs, and natural products) based on different medicine models, and discuss the necessity and feasibility of anti-DTP therapy, related application forms, and future issues that will need to be addressed to advance this emerging field towards clinical applications. Nonetheless, understanding the novel functions of DTP cells may enable us to develop new more effective anticancer therapy and improve clinical outcomes for cancer patients.
    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
    内皮细胞(EC)是心血管系统的重要结构单元,具有两个主要的独特特性:异质性和可塑性。内皮异质性由组织特异性内皮表型的差异及其沿血管床长度的高度修饰倾向定义。异质性的这一方面与可塑性密切相关,ECs通过动员遗传来适应环境线索的能力,分子,和结构改变。特定的内皮细胞结构学可促进快速的结构细胞重组,此外,容易适应外在和内在的环境刺激,被称为表观遗传景观。EC,作为人体普遍分布和无处不在的细胞,在心血管系统中的作用远远超出了它们的结构功能。它们在屏障功能方面起着至关重要的作用,细胞到细胞的通信,以及无数的生理和病理过程。这些包括发展,本体发生,疾病启动,和进步,以及增长,再生,和修复。尽管在了解内皮细胞生物学方面取得了实质性进展,EC在健康状况和病理中的作用仍然是一个令人着迷的探索领域。本文旨在总结内皮生物学的知识和概念。它着重于健康和病理条件下内皮细胞的发育和功能特征,特别强调内皮表型和功能异质性。
    Endothelial cells (ECs) are vital structural units of the cardiovascular system possessing two principal distinctive properties: heterogeneity and plasticity. Endothelial heterogeneity is defined by differences in tissue-specific endothelial phenotypes and their high predisposition to modification along the length of the vascular bed. This aspect of heterogeneity is closely associated with plasticity, the ability of ECs to adapt to environmental cues through the mobilization of genetic, molecular, and structural alterations. The specific endothelial cytoarchitectonics facilitate a quick structural cell reorganization and, furthermore, easy adaptation to the extrinsic and intrinsic environmental stimuli, known as the epigenetic landscape. ECs, as universally distributed and ubiquitous cells of the human body, play a role that extends far beyond their structural function in the cardiovascular system. They play a crucial role in terms of barrier function, cell-to-cell communication, and a myriad of physiological and pathologic processes. These include development, ontogenesis, disease initiation, and progression, as well as growth, regeneration, and repair. Despite substantial progress in the understanding of endothelial cell biology, the role of ECs in healthy conditions and pathologies remains a fascinating area of exploration. This review aims to summarize knowledge and concepts in endothelial biology. It focuses on the development and functional characteristics of endothelial cells in health and pathological conditions, with a particular emphasis on endothelial phenotypic and functional heterogeneity.
    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
    头颈癌(HNC)引起一种异质性肿瘤疾病,由上呼吸道和胃肠道的粘膜上皮引起。它的特点是发病率和死亡率高,是全球第八大最常见的癌症。认为存在于肿瘤环境中的间充质/干基质细胞(MSC)在肿瘤起始的调节中起关键作用。发育和患者预后;它们也影响对顺铂为基础的化疗的耐药性,高级HNC的黄金标准。MSC是多能的,异构和移动小区。虽然没有MSC特异性标记存在,它们可以根据其他几个来识别,例如CD73,CD90和CD105,而缺乏CD45,CD34,CD14或CD11b的存在,CD79α,或CD19和HLA-DR抗原;它们与基质细胞具有表型相似性以及它们分化成其他细胞类型的能力。在肿瘤利基中,MSC群体的特征是细胞静止,自我更新能力,低活性氧的产生和上皮-间质转化特性的获得。它们可能在获得耐药性的过程中起关键作用,从而导致治疗失败。本叙述性综述审查了MSCs与HNC之间的联系,以及HNC对当前化学放射疗法产生耐药性的不同机制。它还研究了HNSCC中与干性相关的化学耐药性的药理靶向的可能性。它描述了有希望的优化放化疗的新策略,具有个性化患者治疗方法的潜力,并强调了HNC未来的治疗前景。
    Head and neck cancer (HNC) entails a heterogenous neoplastic disease that arises from the mucosal epithelium of the upper respiratory system and the gastrointestinal tract. It is characterized by high morbidity and mortality, being the eighth most common cancer worldwide. It is believed that the mesenchymal/stem stromal cells (MSCs) present in the tumour milieu play a key role in the modulation of tumour initiation, development and patient outcomes; they also influence the resistance to cisplatin-based chemotherapy, the gold standard for advanced HNC. MSCs are multipotent, heterogeneous and mobile cells. Although no MSC-specific markers exist, they can be recognized based on several others, such as CD73, CD90 and CD105, while lacking the presence of CD45, CD34, CD14 or CD11b, CD79α, or CD19 and HLA-DR antigens; they share phenotypic similarity with stromal cells and their capacity to differentiate into other cell types. In the tumour niche, MSC populations are characterized by cell quiescence, self-renewal capacity, low reactive oxygen species production and the acquisition of epithelial-to-mesenchymal transition properties. They may play a key role in the process of acquiring drug resistance and thus in treatment failure. The present narrative review examines the links between MSCs and HNC, as well as the different mechanisms involved in the development of resistance to current chemo-radiotherapies in HNC. It also examines the possibilities of pharmacological targeting of stemness-related chemoresistance in HNSCC. It describes promising new strategies to optimize chemoradiotherapy, with the potential to personalize patient treatment approaches, and highlights future therapeutic perspectives in HNC.
    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
    表型可塑性是驱动前列腺癌(PCa)患者治疗抵抗的公认机制。虽然已经确定了驱动表型可塑性的潜在分子原因,治疗成功尚未实现。确定假定的主调节转录因子(MR-TF)驱动PCa的表型可塑性,这项工作利用多组学方法,使用基因工程的前列腺癌小鼠模型,结合患者数据,鉴定MYBL2是PCa中具有表型可塑性的显著富集的转录因子.使用代表表型可塑性的独立鼠PCa细胞系对Mybl2的遗传抑制表明Mybl2损失显著降低了体内生长以及细胞适应性和参与多能性和干性的抑制基因表达特征。因为MYBL2目前不可用药,MYBL2基因签名用于鉴定细胞周期蛋白依赖性激酶-2(CDK2)作为潜在的治疗靶标。CDK2抑制表型改变了Mybl2的遗传丢失,并显着降低了与DNA损伤富集相关的体内肿瘤生长。一起,这项工作证明了MYBL2是PCa中重要的MR-TF驱动表型可塑性。Further,高MYBL2活性鉴定了对CDK2抑制有反应的PCa。
    Phenotypic plasticity is a recognized mechanism driving therapeutic resistance in patients with prostate cancer. Although underlying molecular causations driving phenotypic plasticity have been identified, therapeutic success is yet to be achieved. To identify putative master regulator transcription factors (MR-TF) driving phenotypic plasticity in prostate cancer, this work utilized a multiomic approach using genetically engineered mouse models of prostate cancer combined with patient data to identify MYB proto-oncogene like 2 (MYBL2) as a significantly enriched transcription factor in prostate cancer exhibiting phenotypic plasticity. Genetic inhibition of Mybl2 using independent murine prostate cancer cell lines representing phenotypic plasticity demonstrated Mybl2 loss significantly decreased in vivo growth as well as cell fitness and repressed gene expression signatures involved in pluripotency and stemness. Because MYBL2 is currently not druggable, a MYBL2 gene signature was employed to identify cyclin-dependent kinase-2 (CDK2) as a potential therapeutic target. CDK2 inhibition phenocopied genetic loss of Mybl2 and significantly decreased in vivo tumor growth associated with enrichment of DNA damage. Together, this work demonstrates MYBL2 as an important MR-TF driving phenotypic plasticity in prostate cancer. Furthermore, high MYBL2 activity identifies prostate cancer that would be responsive to CDK2 inhibition.
    UNASSIGNED: Prostate cancers that escape therapy targeting the androgen receptor signaling pathways via phenotypic plasticity are currently untreatable. Our study identifies MYBL2 as a MR-TF in phenotypic plastic prostate cancer and implicates CDK2 inhibition as a novel therapeutic target for this most lethal subtype of prostate 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
    目前,癌症仍然是全球人类死亡的主要原因。肿瘤恶化包括多个事件,包括转移,治疗抗性和免疫逃避,所有这些都与表型可塑性密切相关,尤其是上皮间质可塑性(EMP)。具有EMP的肿瘤细胞表现为上皮-间质转化(EMT)三种状态,部分EMT,和间质-上皮转化,通过转录调控和一系列信号通路协调肿瘤细胞的表型转换和异质性,包括转化生长因子-β,Wnt/β-catenin,还有Notch.然而,由于EMP的复杂性,EMP的多样化过程仍未完全理解。在这次审查中,我们系统地总结生物学背景,EMP的调节机制以及EMP在治疗反应中的作用。我们还总结了一系列小分子抑制剂,免疫相关的治疗方法,以及针对EMP开发的联合疗法对EMP驱动的肿瘤恶化的突出作用。此外,我们探索基于EMP的肿瘤机制研究和治疗研究的潜在技术,这可能会爆发出强劲的前景。总的来说,我们阐明了EMP在肿瘤进展中的多方面,并提出了基于靶向EMP的癌症治疗方向.
    Currently, cancer is still a leading cause of human death globally. Tumor deterioration comprises multiple events including metastasis, therapeutic resistance and immune evasion, all of which are tightly related to the phenotypic plasticity especially epithelial-mesenchymal plasticity (EMP). Tumor cells with EMP are manifest in three states as epithelial-mesenchymal transition (EMT), partial EMT, and mesenchymal-epithelial transition, which orchestrate the phenotypic switch and heterogeneity of tumor cells via transcriptional regulation and a series of signaling pathways, including transforming growth factor-β, Wnt/β-catenin, and Notch. However, due to the complicated nature of EMP, the diverse process of EMP is still not fully understood. In this review, we systematically conclude the biological background, regulating mechanisms of EMP as well as the role of EMP in therapy response. We also summarize a range of small molecule inhibitors, immune-related therapeutic approaches, and combination therapies that have been developed to target EMP for the outstanding role of EMP-driven tumor deterioration. Additionally, we explore the potential technique for EMP-based tumor mechanistic investigation and therapeutic research, which may burst vigorous prospects. Overall, we elucidate the multifaceted aspects of EMP in tumor progression and suggest a promising direction of cancer treatment based on targeting EMP.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号