cancer stemness

癌症干性
  • 文章类型: Journal Article
    癌症进展涉及分化表型的逐渐丧失和祖细胞和干细胞样特征的获得。是免疫疗法抵抗的潜在元凶。尽管最先进的预测计算方法促进了癌症干性的预测,目前还没有能够满足各种使用需求的高效资源。这里,我们在网上展示癌症干细胞,在批量和单细胞水平上有效评估癌症干细胞潜能的综合资源。该资源集成了8个强大的预测算法以及与癌症干性相关的27个签名基因集,用于预测干性评分。从五个不同的方面进行了下游分析,包括确定癌症干性的特征基因,探索与癌症标志的关联,细胞状态,免疫反应,以及与免疫细胞的交流;调查对患者生存的贡献;并在不同方法中对癌症干性进行稳健性分析。此外,用户可以访问40多种癌症类型的预先计算的癌症干性图谱。可以下载分析结果的表格和各种可视化。一起,癌症干细胞在线是癌症干细胞评分和扩展下游功能解释的强大资源,包括免疫反应以及癌症标志。癌症干细胞在线可以在http://bio-bigdata上免费访问。hrbmu.edu.cn/CancerStemnessOnline.
    Cancer progression involves the gradual loss of a differentiated phenotype and the acquisition of progenitor and stem-cell-like features, which are potential culprits of immunotherapy resistance. Although the state-of-art predictive computational methods have facilitated the prediction of cancer stemness, currently there is no efficient resource that can meet various usage requirements. Here, we present the Cancer Stemness Online, an integrated resource for efficiently scoring cancer stemness potential at the bulk and single-cell levels. The resource integrates 8 robust predictive algorithms as well as 27 signature gene sets associated with cancer stemness for predicting stemness scores. Downstream analyses were performed from five different aspects, including identifying the signature genes of cancer stemness, exploring the associations with cancer hallmarks, cellular states, the immune response, and communication with immune cells; investigating the contributions to patient survival; and performing a robustness analysis of cancer stemness among different methods. Moreover, the pre-calculated cancer stemness atlas for more than 40 cancer types can be accessed by users. Both the tables and diverse visualizations of the analytical results are available for download. Together, Cancer Stemness Online is a powerful resource for scoring cancer stemness and expanding the downstream functional interpretation, including immune response as well as cancer hallmarks. Cancer Stemness Online is freely accessible at http://bio-bigdata.hrbmu.edu.cn/CancerStemnessOnline.
    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:肝癌干细胞(CSCs)有助于肿瘤的发生,programming,和肝细胞癌(HCC)的复发。Wnt/β-catenin通路在肝癌干性、programming,转移,和抗药性,但是到目前为止,还没有临床批准的药物有效地靶向这一途径。我们旨在阐明COLEC10在HCC干性中的作用。
    方法:采用癌症基因组图谱(TCGA)和临床蛋白质组学肿瘤分析联盟(CPTAC)数据库来搜索COLEC10表达与HCC干性之间的关联。殖民地的形成,球体形成,侧面人口,和有限稀释肿瘤起始测定用于鉴定COLEC10过表达在HCC细胞系干性中的调节作用。进行Wnt/β-catenin报告蛋白测定和免疫沉淀以探索潜在的机制。
    结果:COLEC10水平与HCC干性呈负相关。升高的COLEC10导致EpCAM和AFP(甲胎蛋白)的表达降低,肝脏CSC的两种常见标志物。COLEC10的过表达抑制HCC细胞形成集落和球体,并减少了体外的侧生数量,以及体内的致瘤能力。机械上,我们证明,COLEC10的过表达通过上调Wnt抑制因子WIF1和降低细胞质β-catenin水平来抑制Wnt/β-catenin信号的活性。COLEC10过表达促进了β-catenin与破坏复合物CK1α成分的相互作用。此外,KLHL22(KelchLikeFamilyMember22),报道的E3连接酶衔接子预测与CK1α相互作用,可以促进COLEC10的单同质化和降解。
    结论:COLEC10通过下调Wnt/β-catenin通路抑制HCC的干性,这是肝脏CSC治疗的一个有希望的目标。
    BACKGROUND: Liver cancer stem cells (CSCs) contribute to tumor initiation, progression, and recurrence in hepatocellular carcinoma (HCC). The Wnt/β-catenin pathway plays a crucial role in liver cancer stemness, progression, metastasis, and drug resistance, but no clinically approved drugs have targeted this pathway efficiently so far. We aimed to elucidate the role of COLEC10 in HCC stemness.
    METHODS: The Cancer Genome Atlas (TCGA) and the Clinical Proteomic Tumor Analysis Consortium (CPTAC) databases were employed to search for the association between COLEC10 expression and HCC stemness. Colony formation, sphere formation, side population, and limiting dilution tumor initiation assays were used to identify the regulatory role of COLEC10 overexpression in the stemness of HCC cell lines. Wnt/β-catenin reporter assay and immunoprecipitation were performed to explore the underlying mechanism.
    RESULTS: COLEC10 level was negatively correlated with HCC stemness. Elevated COLEC10 led to decreased expressions of EpCAM and AFP (alpha-fetoprotein), two common markers of liver CSCs. Overexpression of COLEC10 inhibited HCC cells from forming colonies and spheres, and reduced the side population numbers in vitro, as well as the tumorigenic capacity in vivo. Mechanically, we demonstrated that overexpression of COLEC10 suppressed the activity of Wnt/β-catenin signaling by upregulating Wnt inhibitory factor WIF1 and reducing the level of cytoplasmic β-catenin. COLEC10 overexpression promoted the interaction of β-catenin with the component of destruction complex CK1α. In addition, KLHL22 (Kelch Like Family Member 22), a reported E3 ligase adaptor predicted to interact with CK1α, could facilitate COLEC10 monoubiquitination and degradation.
    CONCLUSIONS: COLEC10 inhibits HCC stemness by downregulating the Wnt/β-catenin pathway, which is a promising target for liver CSC therapy.
    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
    组蛋白脱乙酰酶(HDACs)的失调与癌症的发生和发展密切相关。这里,我们全面分析了所有HDAC家族成员与22种不同肿瘤类型实体瘤的几种临床病理和分子特征之间的关联,主要关注癌症的干性和免疫力。为此,我们使用了公开可用的TCGA数据和几种生物信息学工具(即,GEPIA2,TISIDB,GSCA,Enrichr,GSEA)。我们的分析表明,I类和II类HDAC蛋白与不同的癌症表型相关。转录组分析表明I类HDAC成员,包括HDAC2,与癌症干性呈正相关,而IIA类HDAC蛋白,以HDAC7为代表,显示与实体瘤中癌症干细胞样表型呈负相关。与含有大量HDAC7蛋白的肿瘤相反,HDAC2过表达癌症的转录组特征显著富集了先前确定为干性相关基因的生物学术语.此外,高表达HDAC2的肿瘤被免疫相关过程耗尽,HDAC2表达与肿瘤免疫抑制微环境相关。相反,HDAC7上调与增强的免疫反应显着相关,其次是富集的CD4+和CD8+T细胞浸润。这是第一份全面的报告,展示了特定HDAC家族成员之间强大而通用的关联。癌症去分化,和实体瘤中的抗肿瘤免疫状态。
    Dysregulation of histone deacetylases (HDACs) is closely associated with cancer development and progression. Here, we comprehensively analyzed the association between all HDAC family members and several clinicopathological and molecular traits of solid tumors across 22 distinct tumor types, focusing primarily on cancer stemness and immunity. To this end, we used publicly available TCGA data and several bioinformatic tools (i.e., GEPIA2, TISIDB, GSCA, Enrichr, GSEA). Our analyses revealed that class I and class II HDAC proteins are associated with distinct cancer phenotypes. The transcriptomic profiling indicated that class I HDAC members, including HDAC2, are positively associated with cancer stemness, while class IIA HDAC proteins, represented by HDAC7, show a negative correlation to cancer stem cell-like phenotypes in solid tumors. In contrast to tumors with high amounts of HDAC7 proteins, the transcriptome signatures of HDAC2-overexpressing cancers are significantly enriched with biological terms previously determined as stemness-associated genes. Moreover, high HDAC2-expressing tumors are depleted with immune-related processes, and HDAC2 expression correlates with tumor immunosuppressive microenvironments. On the contrary, HDAC7 upregulation is significantly associated with enhanced immune responses, followed by enriched infiltration of CD4+ and CD8+ T cells. This is the first comprehensive report demonstrating robust and versatile associations between specific HDAC family members, cancer dedifferentiation, and anti-tumor immune statuses in solid tumors.
    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
    由于低反应率,结直肠癌(CRC)对KRAS靶向抑制的反应受到限制。其机制仍然未知。在这里,我们探索了癌症相关成纤维细胞(CAFs)分泌组作为KRAS沉默抗性的介导因子.将CRC细胞系HCT15、HCT116和SW480在推荐培养基中或在来自用rhTGF-β1活化的正常结肠成纤维细胞细胞系(CCD-18Co)的条件培养基中培养,以诱导CAF样表型。通过流式细胞术分析膜干细胞标志物的表达。通过球体形成测定评估干细胞潜能。RNAseq在用来自CAF的对照培养基或条件培养基处理的KRAS沉默的HCT116结肠球中进行。我们的结果表明,KRAS沉默在三种细胞系中上调CD24和下调CD49f和CD104,导致球体形成效率降低。然而,CAF分泌因子恢复干细胞标志物表达并增加干性。RNA测序显示CAF分泌因子上调KRAS沉默细胞中与促瘤通路相关的基因,包括KRAS,TGFβ,NOTCH,WNT,MYC,细胞周期进展和退出静止,上皮-间质转化,和免疫调节。总的来说,我们的结果表明,对KRAS靶向抑制的抗性可能不仅来自细胞内在原因,而且来自外部因素,如成纤维细胞分泌因子。
    Colorectal cancer (CRC) responses to KRAS-targeted inhibition have been limited due to low response rates, the mechanisms of which remain unknown. Herein, we explored the cancer-associated fibroblasts (CAFs) secretome as a mediator of resistance to KRAS silencing. CRC cell lines HCT15, HCT116, and SW480 were cultured either in recommended media or in conditioned media from a normal colon fibroblast cell line (CCD-18Co) activated with rhTGF-β1 to induce a CAF-like phenotype. The expression of membrane stem cell markers was analyzed by flow cytometry. Stem cell potential was evaluated by a sphere formation assay. RNAseq was performed in KRAS-silenced HCT116 colonospheres treated with either control media or conditioned media from CAFs. Our results demonstrated that KRAS-silencing up-regulated CD24 and down-regulated CD49f and CD104 in the three cell lines, leading to a reduction in sphere-forming efficiency. However, CAF-secreted factors restored stem cell marker expression and increased stemness. RNA sequencing showed that CAF-secreted factors up-regulated genes associated with pro-tumorigenic pathways in KRAS-silenced cells, including KRAS, TGFβ, NOTCH, WNT, MYC, cell cycle progression and exit from quiescence, epithelial-mesenchymal transition, and immune regulation. Overall, our results suggest that resistance to KRAS-targeted inhibition might derive not only from cell-intrinsic causes but also from external elements, such as fibroblast-secreted factors.
    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:奥氮平(OLZ)可逆转慢性应激诱导的焦虑。慢性应激通过异常神经内分泌激活促进癌症发展。然而,脑-体相互作用的干预如何逆转慢性应激诱导的肿瘤发生仍然难以捉摸。
    方法:使用KrasLSL-G12D/WT肺癌模型和LLC1同源肿瘤模型研究OLZ对癌症干性和焦虑样行为的影响。通过qPCR评估癌症的干性,西方印迹,免疫组织学染色和干细胞标记的流式细胞术分析,通过小鼠中的连续稀释肿瘤发生和原代肿瘤细胞中的极限稀释分析来评估癌症干细胞样功能。通过高架迷宫和野外试验检测小鼠的焦虑样行为。通过悬尾试验检测小鼠的抑郁样行为。通过医院焦虑和抑郁量表(HADS)评估人类的焦虑和抑郁状态。通过体内同系肿瘤模型和肺癌细胞系中的体外CCK-8测定来评估肺癌的化学敏感性。
    结果:在这项研究中,我们发现在KrasLSL-G12D/WT肺癌模型和LLC1同系肿瘤模型中,OLZ逆转了慢性应激增强的肺肿瘤发生.OLZ通过抑制mPFC中的神经活性并减少慢性压力下的去甲肾上腺素(NE)释放来缓解焦虑和抑郁样行为。NE激活ADRB2-cAMP-PKA-CREB通路促进CLOCK转录,导致癌症干细胞样特征。因此,CLOCK缺乏或OLZ逆转肺癌中NE/慢性应激诱导的吉西他滨(GEM)耐药性。值得注意的是,肿瘤CLOCK表达与应激状态呈正相关,血清NE水平与肺癌患者预后不良的关系。
    结论:我们确定了OLZ改善慢性应激增强的肿瘤发生和化疗耐药的新机制。OLZ抑制mPFC-NE-CLOCK轴以逆转慢性应激诱导的焦虑样行为和肺癌干性。NE释放减少阻止ADRB2-cAMP-PKA-CREB通路的激活以抑制CLOCK转录,从而逆转慢性应激下的肺癌干细胞样性状和化疗耐药。
    BACKGROUND: Olanzapine (OLZ) reverses chronic stress-induced anxiety. Chronic stress promotes cancer development via abnormal neuro-endocrine activation. However, how intervention of brain-body interaction reverses chronic stress-induced tumorigenesis remains elusive.
    METHODS: KrasLSL-G12D/WT lung cancer model and LLC1 syngeneic tumor model were used to study the effect of OLZ on cancer stemness and anxiety-like behaviors. Cancer stemness was evaluated by qPCR, western-blotting, immunohistology staining and flow-cytometry analysis of stemness markers, and cancer stem-like function was assessed by serial dilution tumorigenesis in mice and extreme limiting dilution analysis in primary tumor cells. Anxiety-like behaviors in mice were detected by elevated plus maze and open field test. Depression-like behaviors in mice were detected by tail suspension test. Anxiety and depression states in human were assessed by Hospital Anxiety and Depression Scale (HADS). Chemo-sensitivity of lung cancer was assessed by in vivo syngeneic tumor model and in vitro CCK-8 assay in lung cancer cell lines.
    RESULTS: In this study, we found that OLZ reversed chronic stress-enhanced lung tumorigenesis in both KrasLSL-G12D/WT lung cancer model and LLC1 syngeneic tumor model. OLZ relieved anxiety and depression-like behaviors by suppressing neuro-activity in the mPFC and reducing norepinephrine (NE) releasing under chronic stress. NE activated ADRB2-cAMP-PKA-CREB pathway to promote CLOCK transcription, leading to cancer stem-like traits. As such, CLOCK-deficiency or OLZ reverses NE/chronic stress-induced gemcitabine (GEM) resistance in lung cancer. Of note, tumoral CLOCK expression is positively associated with stress status, serum NE level and poor prognosis in lung cancer patients.
    CONCLUSIONS: We identify a new mechanism by which OLZ ameliorates chronic stress-enhanced tumorigenesis and chemoresistance. OLZ suppresses mPFC-NE-CLOCK axis to reverse chronic stress-induced anxiety-like behaviors and lung cancer stemness. Decreased NE-releasing prevents activation of ADRB2-cAMP-PKA-CREB pathway to inhibit CLOCK transcription, thus reversing lung cancer stem-like traits and chemoresistance under chronic stress.
    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
    目的:化疗耐药是化疗后常见的事件,包括口腔鳞状细胞癌(OSCC)。越来越多的证据表明,癌症的干性显著有助于治疗抗性。关于如何通过靶向干细胞有效地克服OSCC化学抗性,仍然存在一个未解决的问题。本研究旨在探讨二甲双胍的抗肿瘤作用并阐明其潜在的分子机制。
    方法:建立化疗耐药细胞模型,使用CCK-8和软琼脂形成试验评估了它们的活力和成球能力,分别。采用RNA-seq和Western印迹分析来深入研究分子途径。此外,为了证实二甲双胍和顺铂在动物水平的抑制作用,建立皮下肿瘤移植模型.
    结果:二甲双胍作为单一疗法通过Krüppel样因子4(KLF4)抑制了干性性状。二甲双胍和顺铂可以协同抑制细胞增殖和诱导细胞凋亡。动物实验证实了顺铂和二甲双胍对小鼠肿瘤的抑制作用。
    结论:我们的研究提出了一种潜在的治疗方法,即联合化疗和二甲双胍来克服OSCC的化疗耐药性。
    OBJECTIVE: Chemoresistance is a common event after chemotherapy, including oral squamous cell carcinoma (OSCC). Accumulated evidence suggests that the cancer stemness significantly contributes to therapy resistance. An unresolved question remains regarding how to effectively overcome OSCC chemoresistance by targeting stemness. This study aims to investigate the antitumor effect of metformin and clarify the potential molecular mechanisms.
    METHODS: Cellular models resistant to chemotherapy were established, and their viability and sphere-forming ability were assessed using CCK-8 and soft agar formation assays, respectively. RNA-seq and Western blotting analyses were employed to delve into the molecular pathways. Furthermore, to corroborate the inhibitory effects of metformin and cisplatin at an animal level, a subcutaneous tumor transplantation model was instituted.
    RESULTS: Metformin as a monotherapy exhibited inhibition of stemness traits via Krüppel-like factor 4 (KLF4). Metformin and cisplatin can synergically inhibit cell proliferation and induce cell apoptosis. Animal experiments confirmed the inhibitory effect of cisplatin and metformin on tumor in mice.
    CONCLUSIONS: Our study proposes a potential therapeutic approach of combining chemotherapy with metformin to overcome chemoresistance in OSCC.
    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
    越来越多的证据表明,癌症治疗的治疗失败可归因于癌症干细胞(CSC)。在众多癌症干细胞调控者中,非编码RNA(ncRNAs)最近获得了广泛的关注。在这项研究中,我们研究了胃腺癌预测长基因间非编码RNA(GAPLINC)在口腔CSCs(OCSCs)中的作用.
    RNA测序和定量实时聚合酶链反应(qRT-PCR)用于确定GAPLINC的表达。利用流式细胞术和球体形成测定来分离OCSC。醛脱氢酶1(ALDH1)活性的测量,CD44表达细胞,和各种表型分析,比如自我更新,迁移,入侵,和群体形成能力,在敲除GAPLINC后,在两种类型的口腔癌细胞(SAS和GNM)的CSC中进行。还进行了荧光素酶报告基因以验证GAPLINC和microRNA(miR)-331-3p之间的直接相互作用。
    我们的结果表明,GAPLINC在来自患者来源和口腔癌细胞系的OCSC中过表达。我们证明了OCSCs中GAPLINC的沉默下调了各种CSC标志,例如ALDH1活动,CD44表达细胞的百分比,自我更新能力,和菌落形成能力。此外,我们的结果显示,GAPLINC对癌症干性的影响是由miR-331-3p的直接抑制介导的.
    这些数据具有潜在的临床意义,因为我们揭示了GAPLINC的异常上调,并证明抑制GAPLINC可能通过隔离miR-331-3p来降低癌症干性。
    UNASSIGNED: Accumulating evidence has suggested that treatment failure of cancer therapy can be attributed to cancer stem cells (CSCs). Among numerous regulators of cancer stemness, non-coding RNAs (ncRNAs) have gained significant attention recently. In this study, we examined the role of gastric adenocarcinoma predictive long intergenic noncoding RNA (GAPLINC) in oral CSCs (OCSCs).
    UNASSIGNED: RNA Sequencing and quantitative real-time polymerase chain reaction (qRT-PCR) were used to determine the expression of GAPLINC. Flow cytometry and sphere-forming assay were exploited to isolate OCSCs. Measurement of aldehyde dehydrogenase 1 (ALDH1) activity, CD44 expressing cells, and various phenotypic assays, such as self-renewal, migration, invasion, and colony-forming abilities, were conducted in CSCs of two types of oral cancer cells (SAS and GNM) following the knockdown of GAPLINC. A luciferase reporter was also carried out to validate the direct interaction between GAPLINC and microRNA (miR)-331-3p.
    UNASSIGNED: Our results showed that GAPLINC was overexpressed in OCSCs from patient-derived and oral cancer cell lines. We demonstrated that silencing of GAPLINC in OCSCs downregulated various CSC hallmarks, such as ALDH1 activity, percentage of CD44-expressing cells, self-renewal capacity, and colony-forming ability. Moreover, our results revealed that the effect of GAPLINC on cancer stemness was mediated by direct repression of miR-331-3p.
    UNASSIGNED: These data have potential clinical implications in that we unraveled the aberrant upregulation of GAPLINC and demonstrated that suppression of GAPLINC may reduce cancer stemness via sequestering miR-331-3p.
    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:结直肠癌(CRC)是癌症相关死亡的最常见原因之一,并显着损害生活质量。黄芪郁金(AC)在中医治疗CRC中被广泛使用,但确切的机制尚不清楚。
    目的:本研究旨在阐明AC抑制CRC进展的机制。
    方法:使用UPLC-MS/MS分析鉴定AC的活性组分。使用CT26-Lucifer细胞系在BALB/c小鼠中建立原位移植结直肠肿瘤模型以评估AC的作用。使用IVIS成像技术监测肿瘤体积。用苏木精和伊红(H&E)染色进行肿瘤形态的组织学检查。进行小鼠肿瘤样品的转录组测序以鉴定关键途径和分子靶标。使用CCK-8和伤口愈合试验评估AC对细胞活力和迁移的影响。分别。为了研究AC对CRC细胞的影响,使用氯化钴(CoCl2)建立了体外低氧模型,缺氧诱导剂.通过构建稳定的慢病毒载体实现HIF-2α过表达。从RNA-seq中确定的关键靶标,比如c-Myc,Ki-67,β-连环蛋白,使用蛋白质印迹法评估切割的胱天蛋白酶3,CD133和CD44,qRT-PCR,和免疫荧光分析。采用上皮-间充质转化(EMT)和球体克隆测定来评估癌症干细胞的表型变化。
    结果:确定了12种AC成分。AC在体内有效抑制CRC进展。转录组分析强调低氧信号是一个显著富集的途径,暗示其在AC抑制CRC进展中的作用。在低氧模型中,AC在体外抑制CRC细胞的增殖和迁移。此外,AC通过下调干性标志物来降低癌症干性,抑制EMT,减少肿瘤球的形成。低氧反应的下调和AC引起的干性改变涉及HIF-2α和WNT/β-catenin信号传导的减弱。
    结论:这项研究提供了第一个证据,表明AC降低CRC的干性,AC抑制CRC向干细胞样细胞的转变与HIF-2α/β-catenin通路的下调密切相关,尤其是在缺氧条件下。
    BACKGROUND: Colorectal cancer (CRC) is one of the most common causes of cancer-related mortality and significantly impairs quality of life. Astragali Radix-Curcumae Rhizoma (AC) is widely employed in the treatment of CRC in Chinese medicine, but the precise mechanisms remain unclear.
    OBJECTIVE: This study aimed to elucidate the mechanisms by which AC inhibits CRC progression.
    METHODS: The active components of AC were identified using UPLC-MS/MS analysis. An orthotopic transplantation colorectal tumor model was established in BALB/c mice using the CT26-Lucifer cell line to evaluate the effects of AC. Tumor volumes were monitored using IVIS imaging technology. Histological examination of tumor morphology was performed with hematoxylin and eosin (H&E) staining. Transcriptomic sequencing of mouse tumor samples was conducted to identify critical pathways and molecular targets. The impact of AC on cell viability and migration was assessed using CCK-8 and wound healing assays, respectively. To investigate the effects of AC on CRC cells, an in vitro hypoxic model was established using cobalt chloride (CoCl2), a hypoxia inducer. HIF-2α overexpression was achieved by constructing stable lentiviral vectors. Key targets identified from RNA-seq, such as c-Myc, Ki-67, β-catenin, cleaved caspase 3, CD133, and CD44, were evaluated using western blotting, qRT-PCR, and immunofluorescence assays. Epithelial-Mesenchymal Transition (EMT) and spheroid cloning assays were employed to evaluate phenotypic changes in cancer stem cells.
    RESULTS: Twelve components of AC were identified. AC effectively inhibited CRC progression in vivo. Transcriptomic analysis highlighted hypoxic signaling as a significantly enriched pathway, implicating its role in suppressing CRC progression by AC. In the hypoxic model, AC inhibited the proliferation and migration of CRC cells in vitro. Furthermore, AC reduced cancer stemness by downregulating stemness markers, inhibiting EMT, and decreasing tumor sphere formation. The downregulation of hypoxic responses and the shift in stemness by AC involved attenuation of HIF-2α and WNT/β-catenin signaling.
    CONCLUSIONS: This study provides the first evidence that AC reduces the stemness of CRC and the inhibition of the transition of CRC to stem-like cells by AC is closely related to the downregulation of the HIF-2α/β-catenin pathway, especially under hypoxic conditions.
    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
    细胞外囊泡之间复杂的相互作用,癌症的干性特性,和免疫系统显着影响肝细胞癌(HCC)的进展,治疗反应,和患者预后。细胞外囊泡(EV),它们是膜结合结构,在输送蛋白质方面发挥着关键作用,脂质,细胞之间的核酸,从而充当细胞间通讯的基本介体。由于目前很多研究都集中在小的细胞外囊泡(sEV)上,直径范围从30nm到200nm,这篇综述强调了sEV在HCC干细胞与免疫细胞之间相互作用的背景下的作用。sEV为HCC创新诊断和预后生物标志物的临床应用提供了有希望的机会。通过专门针对电动汽车,可以开发针对癌症干性的新疗法。正在进行的研究sEV在肝癌的癌症干性和免疫调节中的作用将扩大我们的理解,并最终为开创性的治疗干预铺平道路。
    The intricate interplay among extracellular vesicles, cancer stemness properties, and the immune system significantly impacts hepatocellular carcinoma (HCC) progression, treatment response, and patient prognosis. Extracellular vesicles (EVs), which are membrane-bound structures, play a pivotal role in conveying proteins, lipids, and nucleic acids between cells, thereby serving as essential mediators of intercellular communication. Since a lot of current research focuses on small extracellular vesicles (sEVs), with diameters ranging from 30 nm to 200 nm, this review emphasizes the role of sEVs in the context of interactions between HCC stemness-bearing cells and the immune cells. sEVs offer promising opportunities for the clinical application of innovative diagnostic and prognostic biomarkers in HCC. By specifically targeting sEVs, novel therapeutics aimed at cancer stemness can be developed. Ongoing investigations into the roles of sEVs in cancer stemness and immune regulation in HCC will broaden our understanding and ultimately pave the way for groundbreaking therapeutic interventions.
    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
    含肿瘤抑制基因F-box和WD重复结构域(FBXW)7通过促进多能干细胞标志物的蛋白质降解来降低癌症干性。我们最近通过几种癌细胞的三维(3D)球体形成证明了FBXW7的转录抑制。在本研究中,我们发现FBXW7的转录活性受到Ca2+激活的K+通道的抑制,KCa1.1,在人前列腺癌LNCaP细胞通过Akt-Nrf2信号通路的3D球体模型中。在LNCaP球体模型中转染miR223模拟物后,通过siRNA介导的CCAAT增强子结合蛋白C/EBPδ(CEBPD)的抑制,FBXW7的转录活性降低,提示在LNCaP球体模型中通过Akt-Nrf2-CEBPD-miR223转录轴对FBXW7进行转录调节。此外,KCa1.1抑制诱导的FBXW7的激活降低了(1)KCa1.1活性和质膜中的蛋白质水平,以及(2)癌症干细胞(CSC)标志物的蛋白质水平,c-Myc,在LNCaP球体模型中,它是被FBXW7降解的分子,表明KCa1.1抑制诱导的FBXW7激活抑制了KCa1.1阳性癌细胞中的CSC转化。
    The tumor suppressor gene F-box and WD repeat domain-containing (FBXW) 7 reduces cancer stemness properties by promoting the protein degradation of pluripotent stem cell markers. We recently demonstrated the transcriptional repression of FBXW7 by the three-dimensional (3D) spheroid formation of several cancer cells. In the present study, we found that the transcriptional activity of FBXW7 was promoted by the inhibition of the Ca2+-activated K+ channel, KCa1.1, in a 3D spheroid model of human prostate cancer LNCaP cells through the Akt-Nrf2 signaling pathway. The transcriptional activity of FBXW7 was reduced by the siRNA-mediated inhibition of the CCAAT-enhancer-binding protein C/EBP δ (CEBPD) after the transfection of miR223 mimics in the LNCaP spheroid model, suggesting the transcriptional regulation of FBXW7 through the Akt-Nrf2-CEBPD-miR223 transcriptional axis in the LNCaP spheroid model. Furthermore, the KCa1.1 inhibition-induced activation of FBXW7 reduced (1) KCa1.1 activity and protein levels in the plasma membrane and (2) the protein level of the cancer stem cell (CSC) markers, c-Myc, which is a molecule degraded by FBXW7, in the LNCaP spheroid model, indicating that KCa1.1 inhibition-induced FBXW7 activation suppressed CSC conversion in KCa1.1-positive cancer cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号