Cancer stem cells

癌症干细胞
  • 文章类型: Journal Article
    乳腺癌是一种复杂的疾病,具有显著的细胞表型异质性,甚至在一个乳腺肿瘤内.新出现的证据强调了肿瘤内竞争的重要性,这可以成为癌症耐药性的关键因素,赋予实质性的临床意义。了解竞争动态至关重要,因为它可以显着影响疾病进展和治疗结果。在目前的工作中,使用微分方程系统建立了一个数学模型来描述两种癌症亚型(每种亚型进一步分为癌症干细胞和肿瘤细胞)和先天免疫细胞之间的动态相互作用.该模型的目的是全面了解异质亚群之间的竞争相互作用。建立了平衡点和每个平衡点的稳定性分析。模型模拟表明,两种癌症亚型之间的竞争直接影响两种物种的数量。当两种癌症亚型之间的竞争很激烈时,增加对更具竞争性的物种特异性的免疫应答率有效地减小了肿瘤的大小。然而,如果竞争相对较弱,需要最佳的免疫反应率来最小化肿瘤细胞的总数。低于最佳水平的比率无法减少更强物种的种群,而高于最佳水平的比率会导致较弱物种的复发。总的来说,该模型提供了对乳腺癌动力学的见解,并指导了有效治疗策略的开发。
    Breast cancer is a complex disease with significant phenotypic heterogeneity of cells, even within a single breast tumor. Emerging evidence underscores the significance of intratumoral competition, which can serve as a key contributor to cancer drug resistance, imparting substantial clinical implications. Understanding the competitive dynamics is paramount as it can significantly influence disease progression and treatment outcomes. In the present work, a mathematical model was developed using a system of differential equations to describe the dynamic interactions between two cancer subtypes (each further classified into cancer stem cells and tumor cells) and innate immune cells. The purpose of the model is to comprehensively understand the competitive interactions between the heterogeneous subpopulations. The equilibrium points and stability analysis for each equilibrium point were established. Model simulations showed that the competition between two cancer subtypes directly affects the number of both species. When competition between two cancer subtypes is strong, increasing the immune response rate specific to the more competitive species effectively reduces the tumor size. However, if the competition is relatively weak, an optimal immune response rate is required to minimize the total number of tumor cells. Rates below the optimal level fail to reduce the population of the stronger species, whereas rates above the optimal level can lead to the recurrence of the weaker species. Overall, this model provides insights into breast cancer dynamics and guides the development of effective treatment strategies.
    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:胶质母细胞瘤(GBM)是成人神经胶质瘤中最普遍和侵袭性的形式。尽管实施了涉及手术的强化治疗方法,辐射,和化疗,胶质母细胞瘤干细胞有助于肿瘤复发和不良预后。通过操纵转录机制诱导成胶质细胞瘤干细胞分化已成为GBM治疗的有希望的策略。这里,我们通过研究在患者来源的GBM细胞(GSC)中观察到的去极化静息膜电位(RMP)的作用,探索了一种创新的方法,当它们处于细胞周期的G0期时,它们可以保持干性。
    方法:我们进行了分子生物学和电生理实验,在体外和体内,为了检查GSCs中电压门控钠通道(Nav)的功能表达,特别关注其细胞周期依赖性功能表达。导航活动是在药理学上操纵的,通过活成像细胞周期分析评估其对GSCs行为的影响,自我更新测定,和化学敏感性测定。通过体外途径分析和体内肿瘤增殖测定研究了Nav在调节GBM干性中的作用的机制见解。
    结果:我们证明了Nav主要在细胞周期的G0阶段由GSCs功能性表达,提示其在调节RMP中的关键作用。Nav的药理学阻断使GBM细胞对替莫唑胺(TMZ)更敏感,这类肿瘤的标准药物,通过诱导细胞周期从G0期重新进入G1/S过渡。此外,Nav的抑制实质上影响了GSCs的自我更新和多潜能特征,同时提高他们的分化程度。最后,我们的数据表明,Nav通过使RMP去极化和抑制ERK信号通路来正向调节GBM的干性.值得注意的是,体内增殖评估证实了药物阻断Nav后对TMZ的易感性增加。
    结论:这一见解将Nav定位为GBM患者的有希望的预后生物标志物和治疗靶点,特别是与替莫唑胺联合治疗。
    BACKGROUND: Glioblastoma (GBM) stands as the most prevalent and aggressive form of adult gliomas. Despite the implementation of intensive therapeutic approaches involving surgery, radiation, and chemotherapy, Glioblastoma Stem Cells contribute to tumor recurrence and poor prognosis. The induction of Glioblastoma Stem Cells differentiation by manipulating the transcriptional machinery has emerged as a promising strategy for GBM treatment. Here, we explored an innovative approach by investigating the role of the depolarized resting membrane potential (RMP) observed in patient-derived GBM sphereforming cell (GSCs), which allows them to maintain a stemness profile when they reside in the G0 phase of the cell cycle.
    METHODS: We conducted molecular biology and electrophysiological experiments, both in vitro and in vivo, to examine the functional expression of the voltage-gated sodium channel (Nav) in GSCs, particularly focusing on its cell cycle-dependent functional expression. Nav activity was pharmacologically manipulated, and its effects on GSCs behavior were assessed by live imaging cell cycle analysis, self-renewal assays, and chemosensitivity assays. Mechanistic insights into the role of Nav in regulating GBM stemness were investigated through pathway analysis in vitro and through tumor proliferation assay in vivo.
    RESULTS: We demonstrated that Nav is functionally expressed by GSCs mainly during the G0 phase of the cell cycle, suggesting its pivotal role in modulating the RMP. The pharmacological blockade of Nav made GBM cells more susceptible to temozolomide (TMZ), a standard drug for this type of tumor, by inducing cell cycle re-entry from G0 phase to G1/S transition. Additionally, inhibition of Nav substantially influenced the self-renewal and multipotency features of GSCs, concomitantly enhancing their degree of differentiation. Finally, our data suggested that Nav positively regulates GBM stemness by depolarizing the RMP and suppressing the ERK signaling pathway. Of note, in vivo proliferation assessment confirmed the increased susceptibility to TMZ following pharmacological blockade of Nav.
    CONCLUSIONS: This insight positions Nav as a promising prognostic biomarker and therapeutic target for GBM patients, particularly in conjunction with temozolomide 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: Letter
    三阴性乳腺癌(TNBC)是由癌症干细胞(CSC)和免疫抑制微环境驱动的特别致命的乳腺癌(BC)亚型。我们的研究表明,伏隔核相关蛋白1(NAC1),BTB/POZ基因家族的成员,通过维持肿瘤干性和影响髓源性抑制细胞(MDSC)在TNBC中发挥关键作用。高NAC1表达与较差的TNBC预后相关。NAC1敲低降低CSC标志物和肿瘤细胞增殖,迁移,和入侵。此外,NAC1影响致癌途径,如CD44-JAK1-STAT3轴和免疫抑制信号(TGFβ,IL-6).有趣的是,NAC1对肿瘤生长的影响随宿主免疫状态而变化,在具有自然杀伤(NK)细胞能力的小鼠中显示出致瘤性降低,但在缺乏NK细胞的小鼠中显示出致瘤性增加。这突出了宿主免疫系统在TNBC进展中的重要作用。此外,MDSC中的高NAC1水平也支持TNBC的干性。一起,本研究提示NAC1是一个有前景的治疗靶点,能够同时根除CSCs和减轻免疫逃避.
    Triple negative breast cancer (TNBC) is a particularly lethal breast cancer (BC) subtype driven by cancer stem cells (CSCs) and an immunosuppressive microenvironment. Our study reveals that nucleus accumbens associated protein 1 (NAC1), a member of the BTB/POZ gene family, plays a crucial role in TNBC by maintaining tumor stemness and influencing myeloid-derived suppressor cells (MDSCs). High NAC1 expression correlates with worse TNBC prognosis. NAC1 knockdown reduced CSC markers and tumor cell proliferation, migration, and invasion. Additionally, NAC1 affects oncogenic pathways such as the CD44-JAK1-STAT3 axis and immunosuppressive signals (TGFβ, IL-6). Intriguingly, the impact of NAC1 on tumor growth varies with the host immune status, showing diminished tumorigenicity in natural killer (NK) cell-competent mice but increased tumorigenicity in NK cell-deficient ones. This highlights the important role of the host immune system in TNBC progression. In addition, high NAC1 level in MDSCs also supports TNBC stemness. Together, this study implies NAC1 as a promising therapeutic target able to simultaneously eradicate CSCs and mitigate immune evasion.
    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
    与其他蒽环类药物相比,吡柔比星具有较高的治疗效果和降低的毒性,因此在临床研究中引起了广泛的关注。然而,~30%接受PIRA治疗的患者仍经历复发和转移。临床进展揭示了存在于肿瘤中的癌症干细胞(CSC)构成了这种限制的主要因素,并且随后是治疗失败的原因。因此,与大块肿瘤一起根除CSC是获得最大治疗效果的关键任务。然而,目前正在检查的大多数CSC抑制剂缺乏特异性,与其他主要治疗方法表现出不同步的生物利用度,并在其治疗应用中表现出明显的毒性,这主要归因于它们的肿瘤靶向能力不足。因此,我们开发了一种可生物降解的聚乳酸基共混嵌段共聚物NP,用于同时递送CSC抑制剂盐霉素(SAL)和化疗药物吡柔比星(PIRA),旨在提高治疗疗效和预防癌症复发.制备的NP显示<100nm的尺寸和优异的负载,这两种药物持续释放。此外,PIRA:SAL共负载的NP表现出协同增强的针对癌细胞以及CSC的细胞毒性。最重要的是,NPs介导的药物共同递送显示肿瘤完全根除,在整个监测期间没有任何复发。此外,NPs治疗未显示重要器官的任何组织病理学改变,证实其无毒性质。总之,本研究得出结论,开发的PIRA:SALNP对肿瘤消退以及预防癌症复发具有优异的疗效,因此可用作癌症治疗的潜在联合疗法。
    Pirarubicin attracted considerable attention in clinical studies because of its high therapeutic efficacy and reduced toxicity in comparison with other anthracyclines. Nevertheless, ~ 30% patients undergoing PIRA treatment still experience relapse and metastasis. Clinical advancements unveiled that cancer stem cells (CSCs) residing in the tumor constitutes a major factor for such limitations and subsequently are the reason for treatment failure. Consequently, eradicating CSCs alongside bulk tumor is a crucial undertaking to attain utmost therapeutic efficacy of the treatment. Nevertheless, majority of the CSCs inhibitors currently under examination lack specificity, show unsynchronized bioavailability with other primary treatments and exhibit notable toxicity in their therapeutic applications, which is primarily attributable to their inadequate tumor-targeting capabilities. Therefore, we have developed a biodegradable polylactic acid based blend block copolymeric NPs for concomitant delivery of CSCs inhibitor Salinomycin (SAL) & chemotherapeutic drug Pirarubicin (PIRA) with an aim to improve the efficacy of treatment and prevent cancer relapse. Prepared NPs showed < 100 nm size and excellent loading with sustained release for both the drugs. Also, PIRA:SAL co-loaded NPs exhibits synergistically enhanced cytotoxicity against cancer cell as well as CSCs. Most importantly, NPs mediated co-delivery of the drugs showed complete tumor eradication, without any reoccurrence throughout the surveillance period. Additionally, NPs treatment didn\'t show any histopathological alteration in vital organs confirming their non-toxic nature. Altogether, present study concludes that the developed PIRA:SAL NPs have excellent efficacy for tumor regression as well as prevention of cancer relapse, hence can be used as a potential combination therapy for cancer 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
    尽管最近在癌症诊断和治疗方面取得了进展,与肺癌相关的死亡率仍然是世界上最高的。后期诊断,常伴有转移,是高死亡率的主要原因,强调迫切需要可靠且易于获得的诊断工具,以检测肺癌特有的生物标志物。循环因素,如循环肿瘤DNA和细胞外囊泡,液体活检已被认为是肺癌的诊断或预后标志物。目前正在进行许多临床研究,以研究循环肿瘤DNA的潜力,循环肿瘤RNA,外泌体,和外泌体微小RNA在肺癌的背景下。这些临床研究旨在解决肺癌诊断不佳和治疗选择有限的问题。以开发临床标志物和个性化治疗为最终目标。在这次审查中,我们讨论了每个循环因子的作用,其研究现状,和正在进行的非小细胞肺癌循环因子的临床研究。此外,我们讨论了在肺癌干细胞中发现的循环因子,并研究了已批准的用于检测肺癌患者循环生物标志物的诊断试验.
    Despite recent advances in cancer diagnostics and treatment, the mortality associated with lung cancer is still the highest in the world. Late-stage diagnosis, often accompanied by metastasis, is a major contributor to the high mortality rates, emphasizing the urgent need for reliable and readily accessible diagnostic tools that can detect biomarkers unique to lung cancer. Circulating factors, such as circulating tumor DNA and extracellular vesicles, from liquid biopsy have been recognized as diagnostic or prognostic markers in lung cancer. Numerous clinical studies are currently underway to investigate the potential of circulating tumor DNA, circulating tumor RNA, exosomes, and exosomal microRNA within the context of lung cancer. Those clinical studies aim to address the poor diagnostics and limited treatment options for lung cancer, with the ultimate goal of developing clinical markers and personalized therapies. In this review, we discuss the roles of each circulating factor, its current research status, and ongoing clinical studies of circulating factors in non-small cell lung cancer. Additionally, we discuss the circulating factors specifically found in lung cancer stem cells and examine approved diagnostic assays designed to detect circulating biomarkers in lung 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
    已经提出乳腺癌干细胞(BCSC)负责乳腺癌(BC)的发展。本研究的目的是评估BCSCs和靶器官微环境免疫表型标志物在常见BC转移中的作用。和关于上述标准的治疗目标。
    此叙述性审查涉及搜索国际数据库;PubMed,谷歌学者使用预先确定的关键词,包括乳腺癌,乳腺癌干细胞,乳腺癌转移,免疫表型,免疫组织化学和转移。搜索结果是根据标题进行评估的,abstract,和文章全文,和相关的调查结果被纳入审查。
    BCSC表达大量的醛脱氢酶1(ALDH1),神经节苷脂2(GD2),CD44和CD133,但CD24标记阴性。CXCR4和OPN在细胞中高表达,可能有助于BC向骨骼的转移。Nestin,已报道BCSC中的CK5、显著蛋白-1(CD133)标志物与脑转移相关。CD44在BCSCs中的高表达和CXCL12在肝脏微环境中的表达可能有助于BC向肝脏的转移。异常表达的血管细胞粘附分子-1(VCAM-1)与肺实质上的胶原蛋白和弹性蛋白纤维结合,肺微环境中BCSCs的CXCR4和CXCL12可能促进细胞归巢和转移到肺。
    在各种类型的BC转移中,由细胞和靶器官微环境表达的不同标志物负责,BCSCs免疫分型可作为预测疾病预后和治疗的靶标志物。
    UNASSIGNED: Breast cancer stem cells (BCSCs) have been suggested to be responsible for the development of Breast cancer (BC). The aim of this study was to evaluate BCSCs and the target organs microenvironment immunophenotyping markers in common BC metastases, and therapeutic targets regarding to the mentioned criteria.
    UNASSIGNED: This narrative review involved searching international databases; PubMed, Google Scholar using predetermined keywords including breast cancer, breast cancer stem cells, breast cancer metastases, immunophenotyping, immunohistochemistry and metastases. The search results were assessed based on the title, abstract, and full text of the articles, and relevant findings were included in the review.
    UNASSIGNED: BCSCs express high amounts of aldehyde dehydrogenase 1 (ALDH1), Ganglioside 2 (GD2), CD44 and CD133 but are negative for CD24 marker. CXCR4 and OPN have high expression in the cells and may contribute in BC metastasis to the bone. Nestin, CK5, prominin-1 (CD133) markers in BCSCs have been reported to correlate with brain metastasis. High expression of CD44 in BCSCs and CXCL12 expression in the liver microenvironment may contribute to BC metastasis to the liver. Aberrantly expressed vascular cell adhesion molecule-1 (VCAM-1) that binds to collagen and elastin fibers on pulmonary parenchyma, and CXCR4 of BCSCs and CXCL12 in lung microenvironment may promote the cells homing and metastasis to lung.
    UNASSIGNED: As in various types of BC metastases different markers that expressed by the cells and target organ microenvironment are responsible, BCSCs immunophenotyping can be used as target markers to predict the disease prognosis and treatment.
    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:增加癌症干细胞(CSC)含量和SOX2过表达是激素依赖性乳腺癌对治疗产生耐药性的共同特征,这仍然是一个重要的临床挑战。SOX2具有作为治疗抵抗的生物标志物和作为治疗靶标的潜力,但是靶向转录因子也具有挑战性。这里,我们研究了不同多金属氧酸盐(POM)衍生物对他莫昔芬耐药乳腺癌细胞中SOX2转录因子的潜在抑制作用.
    方法:合成了各种POM衍生物,并通过红外光谱表征,粉末X射线衍射图和核磁共振波谱。雌激素受体(ER)阳性乳腺癌细胞,和他们的同行,对他莫昔芬的激素疗法产生了抗药性,用POM处理,并通过凝胶阻滞和染色质免疫沉淀评估其后果,以确定SOX2与DNA的结合。对增殖的影响,迁移,使用显微镜监测和定量侵袭和致瘤性,克隆形成,transwell,伤口愈合试验,流式细胞术和体内鸡绒毛尿囊膜(CAM)模型。应用使用CRISPR-Cas9基因组编辑的慢病毒稳定基因沉默和基因敲除的产生来验证所选择的POM的抑制作用。癌症干细胞亚群通过乳腺球形成试验进行定量,ALDEFLUOR活性和CD44/CD24染色。流式细胞术和蛋白质印迹用于测量活性氧(ROS)和细胞凋亡。
    结果:POM阻断了内源性SOX2的体外结合活性。[P2W18O62]6-(PW)Wells-Dawson型阴离子在抑制他莫昔芬抗性的各种细胞系模型中最有效。10µMPW还减少了癌细胞的迁移和侵袭,以及SNAI2表达水平。通过降低CSC含量治疗具有PW受损肿瘤形成的他莫昔芬抗性细胞,以SOX2依赖的方式,导致体内干细胞消耗。机械上,PW诱导活性氧(ROS)的形成并抑制Bcl-2,导致他莫昔芬耐药细胞死亡。PW处理的他莫昔芬抗性细胞显示恢复对他莫昔芬的敏感性。
    结论:一起,这些观察结果强调了PW作为SOX2抑制剂的潜在用途,以及靶向SOX2治疗他莫昔芬耐药乳腺癌的治疗相关性.
    BACKGROUND: Increased cancer stem cell (CSC) content and SOX2 overexpression are common features in the development of resistance to therapy in hormone-dependent breast cancer, which remains an important clinical challenge. SOX2 has potential as biomarker of resistance to treatment and as therapeutic target, but targeting transcription factors is also challenging. Here, we examine the potential inhibitory effect of different polyoxometalate (POM) derivatives on SOX2 transcription factor in tamoxifen-resistant breast cancer cells.
    METHODS: Various POM derivatives were synthesised and characterised by infrared spectra, powder X-ray diffraction pattern and nuclear magnetic resonance spectroscopy. Estrogen receptor (ER) positive breast cancer cells, and their counterparts, which have developed resistance to the hormone therapy tamoxifen, were treated with POMs and their consequences assessed by gel retardation and chromatin immunoprecipitation to determine SOX2 binding to DNA. Effects on proliferation, migration, invasion and tumorigenicity were monitored and quantified using microscopy, clone formation, transwell, wound healing assays, flow cytometry and in vivo chick chorioallantoic membrane (CAM) models. Generation of lentiviral stable gene silencing and gene knock-out using CRISPR-Cas9 genome editing were applied to validate the inhibitory effects of the selected POM. Cancer stem cell subpopulations were quantified by mammosphere formation assays, ALDEFLUOR activity and CD44/CD24 stainings. Flow cytometry and western blotting were used to measure reactive oxygen species (ROS) and apoptosis.
    RESULTS: POMs blocked in vitro binding activity of endogenous SOX2. [P2W18O62]6- (PW) Wells-Dawson-type anion was the most effective at inhibiting proliferation in various cell line models of tamoxifen resistance. 10 µM PW also reduced cancer cell migration and invasion, as well as SNAI2 expression levels. Treatment of tamoxifen-resistant cells with PW impaired tumour formation by reducing CSC content, in a SOX2-dependent manner, which led to stem cell depletion in vivo. Mechanistically, PW induced formation of reactive oxygen species (ROS) and inhibited Bcl-2, leading to the death of tamoxifen-resistant cells. PW-treated tamoxifen-resistant cells showed restored sensitivity to tamoxifen.
    CONCLUSIONS: Together, these observations highlight the potential use of PW as a SOX2 inhibitor and the therapeutic relevance of targeting SOX2 to treat tamoxifen-resistant breast 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
    人ALDH包含19个亚家族,其中ALDH1A1、ALDH1A3、ALDH3A1、ALDH5A1、ALDH7A1和ALDH18A1涉及CSC。研究表明,ALDH也可能与耐药性有关,标准化疗方案对疾病复发阶段的患者治疗无效。现有的化疗药物消除了大部分肿瘤,但通常对表达ALDH+群体的CSC无效。从今以后,靶向ALDH是令人信服的治疗患者的复发后。将信号传导机制相互联系的联合疗法似乎有望提高整体无病生存率。因此,通过ALDH抑制剂和免疫疗法靶向ALDH可能为转化研究创造一个新的平台.这篇综述旨在填补ALDH1家族成员之间在细胞信号传导机制方面的空白。强调它们作为分子靶标致敏复发性肿瘤的潜力,并提出关于当前进展和倒退的未来发展。这篇综述总结了癌症干细胞的作用及其通过维持ALDH被特别突出的肿瘤微环境的上调。它讨论了ALDH家族蛋白的调节以及ALDH和CSC之间与癌症代谢有关的串扰。此外,它建立了ALDH参与的信号机制与其特异性靶向抑制剂之间的相关性,以及它们的功能模块化,生物利用度,以及在各种癌症中的机制作用。
    Human ALDH comprise 19 subfamilies in which ALDH1A1, ALDH1A3, ALDH3A1, ALDH5A1, ALDH7A1, and ALDH18A1 are implicated in CSC. Studies have shown that ALDH can also be involved in drug resistance and standard chemotherapy regimens are ineffective in treating patients at the stage of disease recurrence. Existing chemotherapeutic drugs eliminate the bulk of tumors but are usually not effective against CSC which express ALDH+ population. Henceforth, targeting ALDH is convincing to treat the patient\'s post-relapse. Combination therapies that interlink signaling mechanisms seem promising to increase the overall disease-free survival rate. Therefore, targeting ALDH through ALDH inhibitors along with immunotherapies may create a novel platform for translational research. This review aims to fill in the gap between ALDH1 family members in relation to its cell signaling mechanisms, highlighting their potential as molecular targets to sensitize recurrent tumors and bring forward the future development concerning the current progress and draw backs. This review summarizes the role of cancer stem cells and their upregulation by maintaining the tumor microenvironment in which ALDH is specifically highlighted. It discusses the regulation of ALDH family proteins and the crosstalk between ALDH and CSC in relation to cancer metabolism. Furthermore, it establishes the correlation between ALDH involved signaling mechanisms and their specific targeted inhibitors, as well as their functional modularity, bioavailability, and mechanistic role in various cancers.
    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
    大多数被诊断为胰腺癌的患者最初处于晚期,和放疗阻力影响治疗的有效性。本研究旨在探讨内分泌干扰物邻苯二甲酸二(2-乙基己基)酯(DEHP)对胰腺癌细胞多种生物学行为及放疗敏感性的影响。以及它的潜在机制。我们的发现表明,暴露于DEHP促进各种癌细胞的增殖,包括肺部的,乳房,胰腺,还有肝脏,以时间和浓度依赖的方式。此外,DEHP暴露可以影响胰腺癌细胞的体内和体外生物学行为。这些作用包括减少细胞凋亡,导致G0/G1相停滞,不断增加的迁移能力,增强致瘤性,提高癌症干细胞(CSC)的比例,并上调CSCs标志物如CD133和BMI1的表达水平。DEHP暴露还可以增加抗辐射能力,可以通过下调BMI1表达来逆转。总之,我们的研究表明,DEHP暴露可导致胰腺癌进展和放疗抵抗,其机制可能与BMI1表达上调有关,这导致CSC特性的增加。
    Most patients diagnosed with pancreatic cancer are initially at an advanced stage, and radiotherapy resistance impact the effectiveness of treatment. This study aims to investigate the effects of endocrine disruptor Di-(2-ethylhexyl) phthalate (DEHP) on various biological behaviors and the radiotherapy sensitivity of pancreatic cancer cells, as well as its potential mechanisms. Our findings indicate that exposure to DEHP promotes the proliferation of various cancer cells, including those from the lung, breast, pancreas, and liver, in a time- and concentration-dependent manner. Furthermore, DEHP exposure could influence several biological behaviors of pancreatic cancer cells in vivo and vitro. These effects include reducing cell apoptosis, causing G0/G1 phase arrest, increasing migration capacity, enhancing tumorigenicity, elevating the proportion of cancer stem cells (CSCs), and upregulating expression levels of CSCs markers such as CD133 and BMI1. DEHP exposure can also increase radiation resistance, which can be reversed by downregulating BMI1 expression. In summary our research suggests that DEHP exposure can lead to pancreatic cancer progression and radiotherapy resistance, and the mechanism may be related to the upregulation of BMI1 expression, which leads to the increase of CSCs properties.
    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
    癌症干细胞(CSC)肿瘤异质性和进展的主要调节因子,对癌症转移产生深远的影响,通过各种分泌囊泡。从CSCs中脱颖而出,外泌体是肿瘤微环境中细胞间通讯的关键介质,调节入侵,血管生成,和免疫反应。此外,CSC衍生的外泌体在雕刻动态景观中起着核心作用,有助于恶性表型。在几个外泌体货物中,错误折叠的蛋白质最近因其维持蛋白质稳态和促进肿瘤进展的双重功能而受到关注。中断这些通讯途径可能会阻止CSC的维持和扩展,克服治疗阻力,并抑制肿瘤微环境创造的支持性环境,从而提高癌症治疗的有效性并降低肿瘤复发和转移的风险。此外,外泌体也显示出潜在的治疗应用,例如在药物递送中或作为癌症诊断和预后的生物标志物。因此,理解源自CSC的外泌体的生物学是一个多方面的研究领域,对基础科学和临床应用都有影响。这篇综述探讨了CSCs释放的外泌体错误折叠蛋白之间的复杂相互作用,肿瘤异质性的有力贡献者,以及它们对细胞过程的影响,阐明它们在癌症进展中的作用。
    Cancer stem cells (CSCs), the master regulators of tumor heterogeneity and progression, exert profound influence on cancer metastasis, via various secretory vesicles. Emerging from CSCs, the exosomes serve as pivotal mediators of intercellular communication within the tumor microenvironment, modulating invasion, angiogenesis, and immune responses. Moreover, CSC-derived exosomes play a central role in sculpting a dynamic landscape, contributing to the malignant phenotype. Amidst several exosomal cargoes, misfolded proteins have recently gained attention for their dual functions in maintaining protein homeostasis and promoting tumor progression. Disrupting these communication pathways could potentially prevent the maintenance and expansion of CSCs, overcome treatment resistance, and inhibit the supportive environment created by the tumor microenvironment, thereby improving the effectiveness of cancer therapies and reducing the risk of tumor recurrence and metastasis. Additionally, exosomes have also shown potential therapeutic applications, such as in drug delivery or as biomarkers for cancer diagnosis and prognosis. Therefore, comprehending the biology of exosomes derived from CSCs is a multifaceted area of research with implications in both basic sciences and clinical applications. This review explores the intricate interplay between exosomal misfolded proteins released by CSCs, the potent contributor in tumor heterogeneity, and their impact on cellular processes, shedding light on their role in cancer progression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号