Cancer stem cells

癌症干细胞
  • 文章类型: Journal Article
    背景:实体瘤中的主要免疫细胞是M2样肿瘤相关巨噬细胞(M2样TAMs),显着影响促进肿瘤的上皮-间质转化(EMT),增强干性,促进肿瘤侵袭和转移。然而,M2样TAM对胆囊癌(GBC)肿瘤进展的贡献部分已知.
    方法:免疫组织化学用于评估M2样TAMs和癌症干细胞(CSC)标志物在24对GBC患者的GBC和邻近非癌组织中的表达。随后,GBC细胞和M2样TAM共培养以检测CSC标志物的表达,EMT标记,和迁徙行为。对M2样TAM的培养上清液进行蛋白质组学研究。stemness,使用蛋白质组学和转录组学阐明了M2样TAM在GBC中的转移。将GBC细胞与未分化的巨噬细胞(M0)共培养并分析。在体内观察到吉西他滨联合趋化因子(C-C基序)受体2(CCR2)拮抗剂对GBC的治疗效果。
    结果:与邻近的非癌组织相比,CD68和CD163在M2样TAMs中的表达水平以及CD44和CD133在胆囊癌干细胞(GBCSCs)中的表达水平升高,并呈正相关。M2样TAM分泌大量趋化细胞因子配体2(CCL2),与GBC细胞上的受体CCR2结合后,激活MEK/细胞外调节蛋白激酶(ERK)途径并增强SNAIL表达。ERK途径的激活导致ELK1的核易位,随后导致SNAIL表达增加。GBCSC通过CCL2分泌介导GBC微环境内M0募集和极化为M2样TAM。在鼠类模型中,CCR2拮抗剂和吉西他滨的组合有效地抑制了GBC中皮下肿瘤的生长。
    结论:M2样TAM与GBC细胞之间的相互作用是由趋化因子CCL2介导的,它激活了GBC细胞中的MEK/ERK/ELK1/SNAIL通路,促进EMT,stemness,和转移。CCR2抑制剂和吉西他滨的组合有效抑制皮下肿瘤的生长。因此,我们的研究确定了治疗GBC的有希望的治疗靶点和策略.
    BACKGROUND: The predominant immune cells in solid tumors are M2-like tumor-associated macrophages (M2-like TAMs), which significantly impact the promotion of epithelial-mesenchymal transition (EMT) in tumors, enhancing stemness and facilitating tumor invasion and metastasis. However, the contribution of M2-like TAMs to tumor progression in gallbladder cancer (GBC) is partially known.
    METHODS: Immunohistochemistry was used to evaluate the expression of M2-like TAMs and cancer stem cell (CSC) markers in 24 pairs of GBC and adjacent noncancerous tissues from patients with GBC. Subsequently, GBC cells and M2-like TAMs were co-cultured to examine the expression of CSC markers, EMT markers, and migratory behavior. Proteomics was performed on the culture supernatant of M2-like TAMs. The mechanisms underlying the induction of EMT, stemness, and metastasis in GBC by M2-like TAMs were elucidated using proteomics and transcriptomics. GBC cells were co-cultured with undifferentiated macrophages (M0) and analyzed. The therapeutic effect of gemcitabine combined with a chemokine (C-C motif) receptor 2 (CCR2) antagonist on GBC was observed in vivo.
    RESULTS: The expression levels of CD68 and CD163 in M2-like TAMs and CD44 and CD133 in gallbladder cancer stem cells (GBCSCs) were increased and positively correlated in GBC tissues compared with those in neighboring noncancerous tissues. M2-like TAMs secreted a significant amount of chemotactic cytokine ligand 2 (CCL2), which activated the MEK/extracellular regulated protein kinase (ERK) pathway and enhanced SNAIL expression after binding to the receptor CCR2 on GBC cells. Activation of the ERK pathway caused nuclear translocation of ELK1, which subsequently led to increased SNAIL expression. GBCSCs mediated the recruitment and polarization of M0 into M2-like TAMs within the GBC microenvironment via CCL2 secretion. In the murine models, the combination of a CCR2 antagonist and gemcitabine efficiently inhibited the growth of subcutaneous tumors in GBC.
    CONCLUSIONS: The interaction between M2-like TAMs and GBC cells is mediated by the chemokine CCL2, which activates the MEK/ERK/ELK1/SNAIL pathway in GBC cells, promoting EMT, stemness, and metastasis. A combination of a CCR2 inhibitor and gemcitabine effectively suppressed the growth of subcutaneous tumors. Consequently, our study identified promising therapeutic targets and strategies for treating GBC.
    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
    基因融合是癌症中常见的体细胞改变,和具有致瘤特征的融合已被确定为癌症和治疗靶标的新驱动因素。很少有研究确定融合基因的致癌能力是否与细胞中干细胞的诱导有关。癌症干细胞(CSC)是有助于癌症进展的细胞亚群,转移,和复发,是癌症侵袭性特征的关键组成部分。这里,我们研究了CD63-BCAR4融合基因诱导的CSC样特性,先前报道为致癌融合,及其对CD63-BCAR4的显着特征增强的转移的潜在贡献。CD63-BCAR4过表达促进永生化支气管上皮细胞中的球体形成。在来自CD63-BCAR4过表达细胞的异种移植小鼠的肿瘤衍生细胞中观察到的显著增强的球体形成活性被BCAR4的沉默抑制。RNA微阵列分析显示ALDH1A1在BCAR4融合过表达细胞中上调。与空载体相比,在CD63-BCAR4过表达细胞的球体中观察到ALDH1A1的活性和表达增加。在BCAR4融合过表达细胞中CD133和CD44水平也升高。NANOG增加,在源自注射了CD63-BCAR4过表达细胞的小鼠的转移性肿瘤细胞中观察到S0X2和0CT-3/4蛋白水平。此外,DEAB,ALDH1A1抑制剂,降低了CD63-BCAR4诱导的迁移活性以及球体形成活性。我们的发现表明,CD63-BCAR4融合通过上调ALDH1A1诱导CSC样特性,这有助于其转移特征。
    Gene fusions are common somatic alterations in cancers, and fusions with tumorigenic features have been identified as novel drivers of cancer and therapeutic targets. Few studies have determined whether the oncogenic ability of fusion genes is related to the induction of stemness in cells. Cancer stem cells (CSCs) are a subset of cells that contribute to cancer progression, metastasis, and recurrence, and are critical components of the aggressive features of cancer. Here, we investigated the CSC-like properties induced by CD63-BCAR4 fusion gene, previously reported as an oncogenic fusion, and its potential contribution for the enhanced metastasis as a notable characteristic of CD63-BCAR4. CD63-BCAR4 overexpression facilitates sphere formation in immortalized bronchial epithelial cells. The significantly enhanced sphere-forming activity observed in tumor-derived cells from xenografted mice of CD63-BCAR4 overexpressing cells was suppressed by silencing of BCAR4. RNA microarray analysis revealed that ALDH1A1 was upregulated in the BCAR4 fusion-overexpressing cells. Increased activity and expression of ALDH1A1 were observed in the spheres of CD63-BCAR4 overexpressing cells compared with those of the empty vector. CD133 and CD44 levels were also elevated in BCAR4 fusion-overexpressing cells. Increased NANOG, SOX2, and OCT-3/4 protein levels were observed in metastatic tumor cells derived from mice injected with CD63-BCAR4 overexpressing cells. Moreover, DEAB, an ALDH1A1 inhibitor, reduced the migration activity induced by CD63-BCAR4 as well as the sphere-forming activity. Our findings suggest that CD63-BCAR4 fusion induces CSC-like properties by upregulating ALDH1A1, which contributes to its metastatic features.
    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: 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
    在C57BL/6小鼠中,在联合病理(肺气肿和肺癌)模型中研究了多巴胺D2受体拮抗剂螺培酮的抗肿瘤和抗转移活性。通过施用LPS和香烟烟雾提取物诱导肺气肿。通过将Lewis肺癌细胞注射到肺中诱导肺癌。研究表明,在合并肺部病理的情况下,螺哌酮可防止肺部的炎症浸润和气肿性扩张,并减少原发性肿瘤淋巴结的大小。转移的数量,和受转移影响的肺部区域。Spiperone减少了具有联合病理的小鼠的肺和血液中的癌症干细胞(CSC)的数量。与未治疗的患有肺气肿和肺癌的小鼠的CSC相比,从用螺哌酮治疗的组合病理的小鼠的肺和血液中分离的CSC在体外形成肿瘤圈的可能性显着降低。因此,多巴胺D2受体的阻断是纠正合并肺部病理的有前途的方法,可用于开发治疗肺气肿患者肺癌的方法。
    The antitumor and antimetastatic activity of dopamine D2 receptor antagonists spiperone was studied in C57BL/6 mice in a model of combined pathology (emphysema and lung cancer). Emphysema was induced by administration of LPS and cigarette smoke extract. Lung cancer was induced by injection of Lewis lung carcinoma cells into the lung. It has been shown that under conditions of combined lung pathology, spiperone prevents inflammatory infiltration and emphysematous expansion of the lungs and reduces the size of the primary tumor node, the number of metastases, and the area of the lungs affected by metastases. Spiperone reduces the number of cancer stem cells (CSCs) in the lungs and blood of mice with combined pathology. CSCs isolated from the lungs and blood of mice with combined pathology treated with spiperone had a significantly lower potential to form a tumorosphere in vitro than CSCs from untreated mice with emphysema and lung carcinoma. Thus, blockade of dopamine D2 receptors is a promising approach for correcting combined lung pathology and can be used in the development of a method for treating lung cancer in patients with emphysema.
    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)被认为是癌症表型和细胞异质性的原因。在这里,我们证明了人结肠癌细胞系DLD1包含两种类型的CSC样细胞,它们在重构的基底膜凝胶Matrigel中经历不同的形态发生。在我们用癌细胞球体的方法中,亲本细胞系(DLD1-P)发育出葡萄样的出芽结构,而另一个(DLD1-Wm)及其单细胞克隆动态地发展了蠕虫样克隆。基因表达分析表明,前者模仿肠道隐窝绒毛形态发生,而后者模仿胚胎后肠发育。DLD1-Wm细胞的类器官通过表达偶极蛋白水解活性在两个相反的方向上快速延伸。侵袭性形态发生需要MMP-2和CD133基因的表达和ROCK活性。即使在没有Matrigel的二维培养物中,这些细胞也表现出胃肠病样形态发生。此外,两种DLD1细胞系在细胞生长方面表现出明显的差异,肿瘤生长和对紫杉醇的敏感性。这项研究还为人类癌细胞系提供了一种简单的类器官培养方法。HT-29和其他癌细胞系经历了与正常成纤维细胞直接接触的特征性形态发生。此类类器官培养物可用于研究CSC的性质和用于筛选抗癌药物。我们的结果导致假设CSC样细胞具有侵袭活性和胎儿表型,即,.癌胎儿CSC,在某些类型的结肠癌中产生。
    Cancer stem cells (CSC) are thought to be responsible for cancer phenotypes and cellular heterogeneity. Here we demonstrate that the human colon cancer cell line DLD1 contains two types of CSC-like cells that undergo distinct morphogenesis in the reconstituted basement membrane gel Matrigel. In our method with cancer cell spheroids, the parent cell line (DLD1-P) developed grape-like budding structures, whereas the other (DLD1-Wm) and its single-cell clones dynamically developed worm-like ones. Gene expression analysis suggested that the former mimicked intestinal crypt-villus morphogenesis, while the latter mimicked embryonic hindgut development. The organoids of DLD1-Wm cells rapidly extended in two opposite directions by expressing dipolar proteolytic activity. The invasive morphogenesis required the expression of MMP-2 and CD133 genes and ROCK activity. These cells also exhibited gastrula-like morphogenesis even in two-dimensional cultures without Matrigel. Moreover, the two DLD1 cell lines showed clear differences in cellular growth, tumor growth and susceptibility to paclitaxel. This study also provides a simple organoid culture method for human cancer cell lines. HT-29 and other cancer cell lines underwent characteristic morphogenesis in direct contact with normal fibroblasts. Such organoid cultures would be useful for investigating the nature of CSCs and for screening anti-cancer drugs. Our results lead to the hypothesis that CSC-like cells with both invasive activity and a fetal phenotype, i.e,. oncofetal CSCs, are generated in some types of colon 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
    背景:使癌细胞中耐药状态和药物敏感状态之间发生动态变化的机制仍未得到充分研究。这项研究调查了靶向自噬蛋白降解在调节卵巢癌干细胞(CSC)命运决定和化疗抗性中的作用。
    方法:使用免疫印迹法比较了多种卵巢癌细胞系中CSC富集侧群(SP)和非SP细胞(NSP)之间的自噬水平,免疫荧光,和透射电子显微镜。通过流式细胞术评估自噬调节对CSC标记和分化的影响,免疫印迹和qRT-PCR。计算机建模和免疫共沉淀鉴定了ID1相互作用蛋白。药理学和遗传学方法以及膜联蛋白-PI测定,ChIP分析,西方印迹,qRT-PCR和ICP-MS用于评估对顺铂敏感性的影响,凋亡,SLC31A1表达式,启动子结合,和细胞内铂积累在ID1耗尽的背景下。分析患者来源的肿瘤球体的自噬和SLC31A1水平。
    结果:与非CSC相比,卵巢CSC表现出增加的基础自噬。通过血清饥饿和化学模式的进一步自噬刺激触发了干性调节剂ID1的蛋白水解,从而驱动化学抗性CSC分化为化学敏感性非CSC。在计算机建模中,TCF12被预测为有效的ID1交互因子,通过免疫共沉淀进行验证。ID1消耗释放TCF12以反式激活顺铂流入转运蛋白SLC31A1,从而增加细胞内顺铂水平和细胞毒性。患者来源的肿瘤球体表现出自噬之间的功能关联,ID1、SLC31A1和铂的敏感度。
    结论:这项研究揭示了一种新的自噬-ID1-TCF12-SLC31A1轴,其中ID1的靶向自噬降解能够快速重塑CSCs以逆转化疗抗性。调节该途径可以对抗卵巢癌的耐药性。
    BACKGROUND: The mechanisms enabling dynamic shifts between drug-resistant and drug-sensitive states in cancer cells are still underexplored. This study investigated the role of targeted autophagic protein degradation in regulating ovarian cancer stem cell (CSC) fate decisions and chemo-resistance.
    METHODS: Autophagy levels were compared between CSC-enriched side population (SP) and non-SP cells (NSP) in multiple ovarian cancer cell lines using immunoblotting, immunofluorescence, and transmission electron microscopy. The impact of autophagy modulation on CSC markers and differentiation was assessed by flow cytometry, immunoblotting and qRT-PCR. In silico modeling and co-immunoprecipitation identified ID1 interacting proteins. Pharmacological and genetic approaches along with Annexin-PI assay, ChIP assay, western blotting, qRT-PCR and ICP-MS were used to evaluate effects on cisplatin sensitivity, apoptosis, SLC31A1 expression, promoter binding, and intracellular platinum accumulation in ID1 depleted backdrop. Patient-derived tumor spheroids were analyzed for autophagy and SLC31A1 levels.
    RESULTS: Ovarian CSCs exhibited increased basal autophagy compared to non-CSCs. Further autophagy stimulation by serum-starvation and chemical modes triggered proteolysis of the stemness regulator ID1, driving the differentiation of chemo-resistant CSCs into chemo-sensitive non-CSCs. In silico modeling predicted TCF12 as a potent ID1 interactor, which was validated by co-immunoprecipitation. ID1 depletion freed TCF12 to transactivate the cisplatin influx transporter SLC31A1, increasing intracellular cisplatin levels and cytotoxicity. Patient-derived tumor spheroids exhibited a functional association between autophagy, ID1, SLC31A1, and platinum sensitivity.
    CONCLUSIONS: This study reveals a novel autophagy-ID1-TCF12-SLC31A1 axis where targeted autophagic degradation of ID1 enables rapid remodeling of CSCs to reverse chemo-resistance. Modulating this pathway could counter drug resistance in ovarian 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
    血小板反应蛋白-2(THBS2),分泌的细胞外基质蛋白,在包括血管生成在内的各种生物过程中起着至关重要的作用,组织重塑,和炎症。我们的研究重点是其在人胃癌(GC)中的功能。通过生物信息学和肿瘤组织分析,我们比较了THBS2在GC组织和邻近组织中的表达,和预测的调节上游和下游分子。miR-29b-3p对THBS2的直接调节作用通过双荧光素酶报告基因测定进行评估。显示miR-29b-3p靶向THBS2mRNA的3'-UTR,降低其在GC细胞中的表达。通过蛋白质印迹检查THBS2对肿瘤发生和干性的影响。在体内,通过裸鼠异种移植和转移试验研究了THBS2的作用,证明THBS2的下调会损害GC肿瘤发生和肝转移。我们的研究确定THBS2是GC患者中高表达的预后因子。功能上,THBS2通过调节Notch3、NEY1和HES1蛋白通过Notch信号通路促进GC进展,并通过Notch3维持癌症干细胞样特征,包括CD44,Nanog,OCT4和SOX2。总之,我们的研究表明,THBS2促进GC进展和干性,由miR-29b-3p负调控。这表明THBS2/Notch信号轴内用于对抗胃癌的潜在治疗靶标。
    Thrombospondin-2 (THBS2), a secreted extracellular matrix protein, plays a crucial role in various biological processes including angiogenesis, tissue remodeling, and inflammation. Our study focuses on its function in human gastric cancer (GC). Through bioinformatics and tumor tissue analysis, we compared THBS2 expression in GC tissues and adjacent tissues, and predicted regulatory upstream and downstream molecules. The direct regulatory effect of miR-29b-3p on THBS2 was evaluated through dual-luciferase reporter assays, showing that miR-29b-3p targets the 3\'-UTR of THBS2 mRNA, reducing its expression in GC cells. The influence of THBS2 on tumorigenesis and stemness was examined on protein expression levels via Western blot. In vivo, THBS2\'s role was investigated through xenograft and metastasis assays in nude mice, demonstrating that downregulation of THBS2 impairs GC tumorigenesis and liver metastasis. Our study identified THBS2 as a highly expressed prognostic factor in GC patients. Functionally, THBS2 promotes GC progression through the Notch signaling pathway by regulating Notch3, NEY1, and HES1 proteins, and sustains cancer stem cell-like characteristics by Notch3, including the expression of CD44, Nanog, OCT4, and SOX2. In sum, our study reveals that THBS2 promotes GC progression and stemness, modulated negatively by miR-29b-3p. This suggests potential therapeutic targets within the THBS2/Notch signaling axis for combating gastric 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
    三阴性乳腺癌(TNBC)是乳腺癌的一种侵袭性亚型,缺乏可行的靶标,除了常规化疗之外,治疗选择有限。由于对化疗的固有或获得性抗性,经常遇到治疗失败。先前的研究暗示PI3K/Akt/mTOR信号通路在癌症干细胞(CSC)富集和因此的化学抗性中。本研究旨在研究胡椒碱(PIP)的潜在作用,一种从黑胡椒中分离出的酰胺生物碱,在MDA-MB-231细胞系上和在Ehrlich腹水癌实体瘤动物模型中体内增强TNBC细胞对多柔比星(DOX)的敏感性。结果显示在MDA-MB-231细胞上DOX和PIP之间的协同相互作用。此外,该组合引起PI3K/Akt/mTOR信号传导的抑制增强,与该途径的负调节因子的上调平行,PTEN,随着CSC替代标志物水平的削减,醛脱氢酶-1(ALDH-1)。同时,体内研究表明,联合治疗方案可增强坏死,同时下调PTEN和抑制PI3K水平以及p-Akt,mTOR,和ALDH-1免疫反应性。值得注意的是,该组合未能改变裂解的聚ADP核糖聚合酶水平,提示存在促坏死而非促凋亡机制.总的来说,这些发现表明PIP在体外和体内降低对DOX的抗性方面的潜在作用,可能是通过干扰PI3K/Akt/mTOR通路和CSC。
    Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that lacks an actionable target with limited treatment options beyond conventional chemotherapy. Therapeutic failure is often encountered due to inherent or acquired resistance to chemotherapy. Previous studies implicated PI3K/Akt/mTOR signaling pathway in cancer stem cells (CSCs) enrichment and hence chemoresistance. The present study aimed at investigating the potential effect of piperine (PIP), an amide alkaloid isolated from Piper nigrum, on enhancing the sensitivity of TNBC cells to doxorubicin (DOX) in vitro on MDA-MB-231 cell line and in vivo in an animal model of Ehrlich ascites carcinoma solid tumor. Results showed a synergistic interaction between DOX and PIP on MDA-MB-231 cells. In addition, the combination elicited enhanced suppression of PI3K/Akt/mTOR signaling that paralleled an upregulation in this pathway\'s negative regulator, PTEN, along with a curtailment in the levels of the CSCs surrogate marker, aldehyde dehydrogenase-1 (ALDH-1). Meanwhile, in vivo investigations demonstrated the potential of the combination regimen to enhance necrosis while downregulating PTEN and curbing PI3K levels as well as p-Akt, mTOR, and ALDH-1 immunoreactivities. Notably, the combination failed to change cleaved poly-ADP ribose polymerase levels suggesting a pro-necrotic rather than pro-apoptotic mechanism. Overall, these findings suggest a potential role of PIP in decreasing the resistance to DOX in vitro and in vivo, likely by interfering with the PI3K/Akt/mTOR pathway and CSCs.
    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
    虽然癌症治疗的进展仍在继续,癌症转移和复发带来的持久挑战仍然是癌症患者死亡率升高的强大因素.在与肿瘤复发和转移有关的多方面因素中,癌症干细胞(CSC)由于其对传统疗法的固有抵抗力和增强的侵袭能力而成为值得注意的实体。以他们显著的自我更新能力为特征,分化,和肿瘤发生的开始,根除CSC成为首要目标。最近的研究越来越强调翻译后蛋白质修饰(PTM)在控制CSC的自我更新和复制能力中的关键作用。这篇评论强调了几种流行的PTM的关键意义以及PTM串扰在调节CSC行为中的复杂相互作用。此外,它认为PTM的操纵可能为靶向和消除CSC人群提供了一种新的途径,对癌症治疗提出了令人信服的观点,具有未来应用的巨大潜力。
    While strides in cancer treatment continue to advance, the enduring challenges posed by cancer metastasis and recurrence persist as formidable contributors to the elevated mortality rates observed in cancer patients. Among the multifaceted factors implicated in tumor recurrence and metastasis, cancer stem cells (CSCs) emerge as noteworthy entities due to their inherent resistance to conventional therapies and heightened invasive capacities. Characterized by their notable abilities for self-renewal, differentiation, and initiation of tumorigenesis, the eradication of CSCs emerges as a paramount objective. Recent investigations increasingly emphasize the pivotal role of post-translational protein modifications (PTMs) in governing the self-renewal and replication capabilities of CSCs. This review accentuates the critical significance of several prevalent PTMs and the intricate interplay of PTM crosstalk in regulating CSC behavior. Furthermore, it posits that the manipulation of PTMs may offer a novel avenue for targeting and eliminating CSC populations, presenting a compelling perspective on cancer therapeutics with substantial potential for future applications.
    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
    全球癌症发病率不断上升,据估计,到2030年,每年将有2600万新病例和1700万人死亡。癌症干细胞(CSC)和细胞外囊泡(EV)是癌症抵抗和发展的关键。它们在肿瘤动力学和对治疗的抵抗中起着至关重要的作用。CSCs,最初在急性髓系白血病中发现,以参与肿瘤启动而闻名,programming,和复发,主要是因为它们独特的特点,例如对药物的抗性和自我更新的能力。电动汽车,其中包括外泌体,微泡,和凋亡体,在促进肿瘤微环境(TME)内细胞之间的通讯中起着至关重要的作用。它们对细胞行为有重大影响,并有助于遗传和表观遗传变化。本文分析了CSCs与电动汽车之间的互利联系,强调它们在促进肿瘤扩散和发展耐药机制中的作用。这篇综述旨在研究这些实体之间的相互作用,以发现攻击癌细胞复杂机制的新方法。它强调了CSC和EV作为新型癌症治疗进展中的关键靶标的重要性。这有助于刺激更多的研究,促进癌症治疗理念的进步,并为减轻癌症负担的努力提供新的乐观态度。
    The global incidence of cancer is increasing, with estimates suggesting that there will be 26 million new cases and 17 million deaths per year by 2030. Cancer stem cells (CSCs) and extracellular vesicles (EVs) are key to the resistance and advancement of cancer. They play a crucial role in tumor dynamics and resistance to therapy. CSCs, initially discovered in acute myeloid leukemia, are well-known for their involvement in tumor initiation, progression, and relapse, mostly because of their distinct characteristics, such as resistance to drugs and the ability to self-renew. EVs, which include exosomes, microvesicles, and apoptotic bodies, play a vital role in facilitating communication between cells within the tumor microenvironment (TME). They have a significant impact on cellular behaviors and contribute to genetic and epigenetic changes. This paper analyzes the mutually beneficial association between CSCs and EVs, emphasizing their role in promoting tumor spread and developing resistance mechanisms. This review aims to investigate the interaction between these entities in order to discover new approaches for attacking the complex machinery of cancer cells. It highlights the significance of CSCs and EVs as crucial targets in the advancement of novel cancer treatments, which helps stimulate additional research, promote progress in ideas for cancer treatment, and provide renewed optimism in the effort to reduce the burden of cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号