Colon cancer stem cells

结肠癌干细胞
  • 文章类型: Journal Article
    目的:探讨miR-513b-5p通过IL-6/STAT3抑制结肠癌干细胞(CCSCs)在HCT116细胞中转移的机制。
    方法:球体形成培养基和磁性细胞分选用于富集和筛选CCSCs。我们用了集落形成试验,细胞增殖和活力测定,和裸鼠移植瘤试验以鉴定CCSCs。进行ELISA以鉴定细胞培养基中的IL-6,和增长,生存能力,伤口愈合,并比较不同细胞群的transwell迁移以区分它们。双荧光素酶报告基因测定,RT-PCR,进行和/或WesternBlot分析以确定它们之间的相关性。
    结果:CD133+CD44+HCT116细胞具有更高的克隆效率,更大的增殖能力和生存能力,和更强的致瘤性。双荧光素酶报告基因分析显示miR-513b-5p对STAT3表达有负面影响。RT-PCR和/或WesternBlot分析提示miR-513b-5p对STAT3和波形蛋白有负面影响,同时积极影响E-cadherin表达。STAT3过表达载体+miR-513b-5p抑制剂细胞组有效率最高,最大的增殖能力和生存能力,和实验中最高的IL-6水平。
    结论:Mir-513b-5p通过IL-6/STAT3抑制CCSCs的上皮-间质转化(EMT)。这种潜在的机制可能为结肠癌提供新的治疗靶点。
    OBJECTIVE: To reveal the mechanisms by which miR-513b-5p inhibits metastasis of colon cancer stem cells (CCSCs) through IL-6/STAT3 in HCT116 cells.
    METHODS: Sphere formation media and magnetic cell sorting were used to enrich and screen CCSCs. We used a colony formation assay, cell proliferation and viability assays, and a nude mouse transplantation tumor assay to identify CCSCs. ELISA was performed to identify IL-6 in the cell culture medium, and the growth, viability, wound healing, and transwell migration of distinct cell groups were compared to differentiate them. Dual-luciferase reporter assay, RT-PCR, and/or Western Blot analysis were conducted to determine the correlation between them.
    RESULTS: CD133+CD44+ HCT116 cells were shown to have higher cloning efficiency, greater proliferation ability and viability, and stronger tumorigenicity. A dual-luciferase reporter assay revealed that miR-513b-5p negatively affected STAT3 expression. RT-PCR and/or Western Blot analysis suggested that miR-513b-5p negatively affected STAT3 and Vimentin, while positively affecting E-cadherin expression. The STAT3 overexpression vector + miR-513b-5p inhibitor cell group had the highest efficiency, greatest proliferation ability and viability, and the highest IL-6 level in the experiments.
    CONCLUSIONS: Mir-513b-5p inhibited the epithelial-mesenchymal transition (EMT) of CCSCs through IL-6/STAT3. This potential mechanism may provide a new therapeutic target for colon 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
    UNASSIGNED: To explore whether metformin (MET) can affect the biological behaviour and CD133 mRNA expression of CD133+ colon cancer stem cells (CCSCs) through miR-342-3p.
    UNASSIGNED: The direct immunomagnetic bead method was used to select CD133+ CCSCs from the SW480 and HCT116 cell lines, and miRNA-tailing qRT-PCR was used to detect the expression changes of tumor suppressor-related miRNAs (miR-34a, miR-126, miR-143, miR-145, miR-342-3p, miR-342-5p) after MET intervention. Then, miR-342-3p with markedly significant differential expression was selected as the target miRNA. The lentiviruses LV16-hsa-miR-342-3p inhibitor and LV16-NC were used for the transfection inhibition test. CCK-8, flow cytometry, and qRT-PCR were used to detect the cell viability, apoptosis rate, and CD133 mRNA expression of CD133+ CCSCs.
    UNASSIGNED: Under the high-glucose environment, the expression of tumor suppressor-related miRNAs in CCSCs changed differently (p <0.05), MET also had different effects on the expression of tumor suppressor-related miRNA under different glucose concentrations (p<0.05). Among them, MET upregulates the expression of miR-342-3p in CCSCs for the first time. The results of the lentiviruses transfection inhibition test showed that after miR-342-3p was inhibited, the cell viability and apoptosis rate of CD133+ CCSCs did not change significantly compared with before inhibition (p>0.05), but the expression of CD133 mRNA markedly increased (p<0.05). Meanwhile, after MET intervention, the apoptosis rate and the expression of CD133 mRNA of CD133+ CCSCs was significantly increased, and the proliferation of CD133+ CCSCs was obviously inhibited (p<0.05).
    UNASSIGNED: MET upregulating the expression of miR-342-3p may not have a significant effect on the proliferation and apoptosis of CD133+ CCSCs, but it can reduce the expression of CD133 mRNA in CD133+ CCSCs.
    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
    Colon cancer stem cells (CCSCs) play an important role in facilitating colon cancer occurrence, metastasis and drug resistance. The results of our previous studies confirmed that the well-studied antioxidant gene peroxiredoxin-2 (PRDX2) promotes colon cancer progression. However, the underlying function and mechanisms associated with PRDX2 remodeling in the context of CCSCs have remained poorly studied. In our present study, we demonstrated that PRDX2 is highly expressed in CD133/CD44-positive colon cancer tissues and spheroid CD133+CD44+ CCSCs. PRDX2 overexpression was shown to be closely correlated with CD133+CD44+ CCSCs in colon cancer. Furthermore, PRDX2 depletion markedly suppressed CD133+CD44+ CCSC stemness maintenance, tumor initiation, migration and invasion and liver metastasis. Furthermore, the expression of various EMT markers and Wnt/β-catenin signaling proteins was altered after PRDX2 inhibition. In addition, PRDX2 knockdown led to increased ROS production in CD133+CD44+ CCSCs, sensitizing CCSCs to oxidative stress and chemotherapy. These results suggest that PRDX2 could be a possible therapeutic target in CCSCs.
    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
    BACKGROUND: Celecoxib (CXB), a selective COX-2 inhibitor NSAID, has exhibited prominent anti-proliferative potential against numerous cancers. However, its low bioavailability and long term exposure related cardiovascular side effects, limit its clinical application. In order to overcome these limitations, natural bioactive compounds with lower toxicity profile are used in combination with therapeutic drugs. Therfore, in this study Piperine (PIP), a natural chemo-preventive agent possessing drug bioavailability enhancing properties, was considered to be used in combination with low doses of CXB.
    OBJECTIVE: We hypothesized that the combination of PIP with CXB will have a synergistic anti-proliferative effect on colon cancer cells.
    METHODS: The potency of PIP and CXB alone and in combination was evaluated in HT-29 human colon adenocarcinoma cells and mechanism of growth inhibition was investigated by analyzing the players in apoptotic and Wnt/β-catenin signaling pathways.
    METHODS: The effect of PIP on the oral bioavailability of CXB in mice was investigated using HPLC analysis. The study investigated the synergistic anti-proliferative effect of CXB and PIP on HT-29 cells and IEC-6 non-tumorigenic rat intestinal epithelial cells by SRB cell viability assay. Further, the cellular and molecular mechanism(s) involved in the anti-proliferative combinatorial effect was extensively explored in HT-29 cells by flow cytometry and western blotting. The in vivo efficacy of this combination was studied in CT26.WT tumor syngeneic Balb/c mice model.
    RESULTS: PIP as a bioenhancer increased the oral bioavailability of CXB (129%). The IC50 of CXB and PIP were evaluated to select doses for combination treatment of HT-29 cells. The drug combinations having combination index (CI) less than 1 were screened using CompuSyn software. These combinations were significantly cytotoxic to HT-29 cells but IEC-6 were least effected. Further, the mechanism behind CXB and PIP mediated cell death was explored. The co-treatment led to reactive oxygen species generation, mitochondrial dysfunction, caspase activation and enhanced apoptosis in HT-29 cells. Additionally, the combination treatment synergistically modulated Wnt/β-catenin pathway, downregulated the stemness markers and boosted therapeutic response in CT26 syngeneic Balb/c mice.
    CONCLUSIONS: The outcomes of the study suggests that combining CXB and PIP offers a novel approach for the treatment of colon cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • DOI:
    文章类型: Journal Article
    Patients with advanced-stage colon cancer often exhibit resistance against treatment and distant metastasis, both key contributors to poor prognosis. Emerging evidence indicates that cancer stem cells (CSCs), characterized by the enhanced ability to self-renew, resist therapeutics, and promote metastasis, represents a clinical challenge to target. Alternative therapeutic approaches are urgently required. Here, we explored the feasibility of disrupting the intracellular communications between CSCs and the tumor microenvironment by way of exosomes. First, we demonstrated that exosomes secreted by colon tumorspheres (Exosp) promoted 5-FU resistance, migration, and tumorsphere formation. Exosp also increased the generation of cancer-associated fibroblasts and M2 polarized macrophages in vitro. Oncogenic molecules, including IL-6, p-STAT3, TGF-β1, and β-catenin, were identified as the cargoes of Exosp. Furthermore, the public database revealed the high abundance of miR-1246 in serum exosomes from colon cancer patients, and we verified in the Exosp from HCT116 and HT29 cells. Therapeutically, we demonstrated the ovatodiolide treatment reduced exosomal cargoes from tumorspheres (Exosp_OV). Exosp_OV were significantly less capable of promoting 5-FU resistance, migration, and tumorsphere formation when co-cultured with HCT116 and HT29 cells. Notably, Exosp_OV was less CAF- and M2 TAM-transformative. Computational docking analysis revealed that OV could bind and significantly reduced β-catenin activity. Finally, mouse xenograft data indicated that ovatodiolide suppressed tumor growth via down-regulating IL-6, STAT3, β-catenin expression, and serum exosomal miR-1246. In conclusion, our findings provided preclinical supports for ovatodiolide as a colon CSC inhibitor by reducing β-catenin/STAT3/miR-1246 signaling conveyed by CSC derived exosomes.
    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
    结直肠癌(CRC)是由非典型结肠细胞的不受控制的繁殖和耐力引起的致命疾病。一个人的生活方式和饮食习惯对CRC有积极和/或消极的影响。饮食衍生的植物化学物质调节微生物组以及靶向结肠癌干细胞(CSCs),这些细胞被发现对CRC具有显著的保护作用。它们被组织在纸上的适当位置。所有关于膳食植物化学物质的信息,肠道微生物组,CSCs,它们对CRC的影响是从各种数据库和电子搜索引擎中获取的。使用各种膳食植物化学物质或调节微生物组可以降低CRC的有效性,从而减少或逆转肿瘤以及CSC的进展,这可能是减轻CRC负担的一种有希望和有效的方法。通过明显的抗肿瘤作用和对CSC的目标,具有调节肠道微生物组的植物化学物质继续是CRC的吉祥研究,这为癌症抑制和治疗提供了新的机会。
    Colorectal cancer (CRC) is a fatal disease caused by the uncontrolled propagation and endurance of atypical colon cells. A person\'s lifestyle and eating pattern have significant impacts on the CRC in a positive and/or negative way. Diet-derived phytochemicals modulate the microbiome as well as targeting colon cancer stem cells (CSCs) that are found to offer significant protective effects against CRC, which were organized in an appropriate spot on the paper. All information on dietary phytochemicals, gut microbiome, CSCs, and their influence on CRC were accessed from the various databases and electronic search engines. The effectiveness of CRC can be reduced using various dietary phytochemicals or modulating microbiome that reduces or inverses the progression of a tumor as well as CSCs, which could be a promising and efficient way to reduce the burden of CRC. Phytochemicals with modulation of gut microbiome continue to be auspicious investigations in CRC through noticeable anti-tumorigenic effects and goals to CSCs, which provides new openings for cancer inhibition and treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:结肠癌干细胞(CSCs),被认为是肿瘤发生和癌症复发的原因,不断暴露于肿瘤微环境中存在的邻近细胞发出的调控线索。这些细胞是肠神经胶质细胞(EGC),它们是健康肠道中上皮功能的有效调节剂。然而,EGC是否影响CSC驱动的肿瘤发生尚不清楚.
    方法:使用鼠异种移植模型和3D共培养系统,确定了人EGC原代培养物或非转化EGC细胞系对从人原发性结肠腺癌或结肠癌细胞系中分离的CSC的影响。患者匹配的人原发性结肠腺癌和非邻近健康粘膜的上清液用于模拟肿瘤与健康粘膜的分泌体,并比较其对EGC的影响。
    结果:我们的数据显示,EGCs刺激CSC扩增,并通过旁分泌信号产生肿瘤。重要的是,只有被肿瘤上皮细胞来源的可溶性因子预激活的EGC增加了CSC的致瘤性.EGC中PGE2生物合成的药理学抑制或肿瘤上皮细胞中IL-1敲低阻止了EGC获得促致瘤表型。CSC中PGE2受体EP4和EGFR的抑制抑制了肿瘤激活的EGC的作用。
    结论:总而言之,我们的结果表明,EGCs,一旦被肿瘤激活,通过PGE2/EP4/EGFR依赖性途径获得促瘤表型并刺激CSC驱动的肿瘤发生。
    背景:这项工作得到了法国国家癌症研究所的资助,LaLigueContreleCancer,“卢瓦尔河军团”和UNCLineberger综合癌症中心。
    BACKGROUND: Colon cancer stem cells (CSCs), considered responsible for tumor initiation and cancer relapse, are constantly exposed to regulatory cues emanating from neighboring cells present in the tumor microenvironment. Among these cells are enteric glial cells (EGCs) that are potent regulators of the epithelium functions in a healthy intestine. However, whether EGCs impact CSC-driven tumorigenesis remains unknown.
    METHODS: Impact of human EGC primary cultures or a non-transformed EGC line on CSCs isolated from human primary colon adenocarcinomas or colon cancer cell lines with different p53, MMR system and stemness status was determined using murine xenograft models and 3D co-culture systems. Supernatants of patient-matched human primary colon adenocarcinomas and non-adjacent healthy mucosa were used to mimic tumor versus healthy mucosa secretomes and compare their effects on EGCs.
    RESULTS: Our data show that EGCs stimulate CSC expansion and ability to give rise to tumors via paracrine signaling. Importantly, only EGCs that were pre-activated by tumor epithelial cell-derived soluble factors increased CSC tumorigenicity. Pharmacological inhibition of PGE2 biosynthesis in EGCs or IL-1 knockdown in tumor epithelial cells prevented EGC acquisition of a pro-tumorigenic phenotype. Inhibition of PGE2 receptor EP4 and EGFR in CSCs inhibited the effects of tumor-activated EGCs.
    CONCLUSIONS: Altogether, our results show that EGCs, once activated by the tumor, acquire a pro-tumorigenic phenotype and stimulate CSC-driven tumorigenesis via a PGE2/EP4/EGFR-dependent pathway.
    BACKGROUND: This work was supported by grants from the French National Cancer Institute, La Ligue contre le Cancer, the \'Région des Pays de la Loire\' and the UNC Lineberger Comprehensive Cancer Center.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    尽管在结肠癌治疗方面取得了许多进展和优化,肿瘤复发和转移使得开发新的治疗方法成为必要.结肠癌干细胞(CCSCs)被认为是癌症进展的主要触发因素。复发,和转移。由于积累的遗传和表观遗传改变以及与肿瘤微环境(TME)的复杂互连,CCSC可以进化并转化为完整的恶性细胞。越来越多的证据表明,在癌症治疗中,TME中的CCSC和非CCSC都必须被认为突破了当前治疗的局限性。在这方面,一些非CCSC的干细胞能力可能出现在TME条件内。因此,对监管机制有深刻的了解,异质性,特定标记,需要CCSCs的信号通路及其与TME成分的相互联系,以改善结直肠癌的治疗和患者的生活质量。在这次审查中,我们探讨了目前靶向CCSCs和TME其他成分的细胞表面标志物和信号通路的不同靶向治疗方案.本文还提供了结肠癌个性化治疗的当前挑战和未来前景。一起来看,基于对CCSCs生物学的深刻理解,利用三维培养技术,有可能成功根除结肠癌,并针对CCSCs和TME进行联合靶向治疗.
    Despite many advances and optimization in colon cancer treatment, tumor recurrence and metastases make the development of new therapies necessary. Colon cancer stem cells (CCSCs) are considered as the main triggering factor of cancer progression, recurrence, and metastasis. CCSCs as a result of accumulated genetic and epigenetic alterations and also complex interconnection with the tumor microenvironment (TME) can evolve and convert to full malignant cells. Mounting evidence suggests that in cancer therapy both CCSCs and non-CCSCs in TME have to be regarded to break through the limitation of current therapies. In this regard, stem cell capabilities of some non-CCSCs may arise inside the TME condition. Therefore, a deep knowledge of regulatory mechanisms, heterogeneity, specific markers, and signaling pathways of CCSCs and their interconnection with TME components is needed to improve the treatment of colorectal cancer and the patient\'s life quality. In this review, we address current different targeted therapeutic options that target cell surface markers and signaling pathways of CCSCs and other components of TME. Current challenges and future perspectives of colon cancer personalized therapy are also provided here. Taken together, based on the deep understanding of biology of CCSCs and using three-dimensional culture technologies, it can be possible to reach successful colon cancer eradication and improvise combination targeted therapies against CCSCs and TME.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    目的:研究具有基因特征的结直肠癌干细胞(CR-CSC)系全基因组甲基化。材料和方法:从原发性结直肠癌(CRC)组织中分离出8个CR-CSC系,培养并表征为非整倍性,CRC相关基因的突变状态和微卫星不稳定性(MSI)。通过MethylationEPIC微阵列评估全基因组DNA甲基化。结果:我们描述了在免疫受损小鼠体内传代后保持的独特甲基化模式。我们鉴定了与MSI相关的表观遗传CR-CSC特征。我们注意到,差异甲基化位置的优势不存在于CpG岛,而是蔓延到陆架和公海地区。结论:鉴于MSI高状态的CRC具有较低的转移潜力,MSI相关甲基化特征的鉴定可以为转移性CRC提供新的见解和可能的靶标.
    Aim: To investigate the genome-wide methylation of genetically characterized colorectal cancer stem cell (CR-CSC) lines. Materials & methods: Eight CR-CSC lines were isolated from primary colorectal cancer (CRC) tissues, cultured and characterized for aneuploidy, mutational status of CRC-related genes and microsatellite instability (MSI). Genome-wide DNA methylation was assessed by MethylationEPIC microarray. Results: We describe a distinctive methylation pattern that is maintained following in vivo passages in immune-compromised mice. We identified an epigenetic CR-CSC signature associated with MSI. We noticed that the preponderance of the differentially methylated positions do not reside at CpG islands, but spread to shelf and open sea regions. Conclusion: Given that CRCs with MSI-high status have a lower metastatic potential, the identification of a MSI-related methylation signature could provide new insights and possible targets into metastatic CRC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Despite incessant research, colon cancer still is one of the most common causes of fatalities in both men and women worldwide. Also, nearly 50% of patients with colorectal cancer show tumor recurrence. Recent investigations have highlighted the involvement of colon cancer stem cells (CCSCs) in cancer relapse and chemoresistance. CCSCs deliver a significant protumorigenic niche through persistent overexpression of self-renewal capabilities. Moreover, CSCs cross network with stromal cells, immune infiltrates, and cyotokine-chemokine, which potentiate their aggressive proliferative potential. Targeting CCSCs through small molecule inhibitors, miRNAs, and monoclonal antibodies (mAbs) in in vivo studies has generated compelling evidence for the effectiveness of these various treatments. This review effectively compiles the role of CCSC surface markers and dysregulated and/or upregulated pathways in the pathogenesis of colorectal cancer that can be used to target CCSCs for effective colorectal cancer treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号