cancer‐associated fibroblasts

  • 文章类型: Journal Article
    肝细胞癌(HCC)是肝癌的一种致命形式,和肿瘤微环境,特别是癌症相关成纤维细胞(CAFs),在其发展中起着至关重要的作用。本研究旨在阐明CAF来源的外泌体调节HCC发展的机制。该研究采用定量实时聚合酶链反应进行mRNA表达分析,蛋白质印迹分析进行蛋白质表达检测。进行染色质免疫沉淀测定和双荧光素酶报告基因测定以研究锌指蛋白250(ZNF250)与程序性细胞死亡1配体1(PD-L1)之间的关系。透射电子显微镜和蛋白质印迹分析用于表征分离的外来体。使用绿色荧光标记染料PKH67分析了CAF衍生的外泌体和正常成纤维细胞(NFs)衍生的外泌体向HCC细胞的可转移性。通过5-乙炔基-2'-脱氧尿苷测定法评估细胞增殖,同时进行Transwell测定以评估细胞迁移和侵袭。流式细胞术检测细胞凋亡,而酶联免疫吸附试验用于评估肿瘤坏死因子-α和穿孔素的水平。最后,构建了异种移植小鼠模型,以检查ZNF250缺陷型CAF来源的外泌体对HCC细胞肿瘤特性的影响.该研究显示ZNF250在肝癌组织和细胞中的表达增加,ZNF250转录激活HCC细胞中的PD-L1。ZNF250表达与HbsAg相关,肝癌患者的临床分期和肿瘤大小。CAF来源的外泌体ZNF250可以调节HCC细胞中PD-L1的表达。此外,来自ZNF250缺陷型CAFs的外泌体抑制了增殖,迁移,入侵,通过下调PD-L1表达来实现肝癌细胞的免疫逃逸。此外,CAF来源的外泌体ZNF250在体内促进肿瘤形成。这些发现为CAF衍生的外泌体在抑制HCC发展中的作用提供了见解。强调ZNF250和PD-L1调节在肿瘤进展中的意义。
    Hepatocellular carcinoma (HCC) is a lethal form of liver cancer, and the tumor microenvironment, particularly cancer-associated fibroblasts (CAFs), plays a critical role in its progression. This study aimed to elucidate the mechanism by which CAF-derived exosomes regulate the development of HCC. The study employed quantitative real-time polymerase chain reaction for mRNA expression analysis and western blot analysis for protein expression detection. Chromatin immunoprecipitation assay and dual-luciferase reporter assay were performed to investigate the relationship between zinc finger protein 250 (ZNF250) and programmed cell death 1 ligand 1 (PD-L1). Transmission electron microscopy and western blot analysis were used to characterize the isolated exosomes. The transferability of CAF-derived exosomes and normal fibroblasts (NFs)-derived exosomes into HCC cells was analyzed using a green fluorescent labeling dye PKH67. Cell proliferation was assessed via a 5-Ethynyl-2\'-deoxyuridine assay, while Transwell assays were conducted to evaluate cell migration and invasion. Flow cytometry was performed to measure cell apoptosis, while enzyme-linked immunosorbent assays were used to assess the levels of tumor necrosis factor-α and perforin. Finally, a xenograft mouse model was constructed to examine the effects of exosomes derived from ZNF250-deficient CAFs on the tumor properties of HCC cells. The study revealed increased expression of ZNF250 in HCC tissues and cells, with ZNF250 transcriptionally activating PD-L1 in HCC cells. ZNF250 expression was associated with HbsAg, clinical stage and tumor size of HCC patients. CAF-derived exosomal ZNF250 can regulate PD-L1 expression in HCC cells. Furthermore, exosomes derived from ZNF250-deficient CAFs inhibited the proliferation, migration, invasion, and immune escape of HCC cells by downregulating PD-L1 expression. Moreover, CAF-derived exosomal ZNF250 promoted tumor formation in vivo. These findings provide insights into the role of CAF-derived exosomes in the suppression of HCC development, highlighting the significance of ZNF250 and PD-L1 regulation in tumor progression.
    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
    目前,脊索瘤内质网应激-CAF(ERS-CAF)亚群的致癌机制尚不清楚.这里,单细胞RNA测序,空间转录组学,GeoMx数字空间分析器,独立于数据的采集蛋白质组学,批量RNA-seq,和多重定量免疫荧光用于揭示ERS-CAF如何影响脊索瘤进展的精确分子机制。结果表明,低氧微环境将CAF重新编程为ERS-CAF亚型。机械上,这是通过缺氧介导的IER2转录上调而发生的。CAFs中IER2的过表达促进脊索瘤的进展,这可能会受到IER2敲低或使用ERS抑制剂的阻碍。IER2还诱导ERS-CAF标记基因的表达,并导致产生促肿瘤性旁分泌GMFG信号,通过与肿瘤细胞上的ITGB1直接结合发挥其生物学功能。ITGB1抑制减弱肿瘤恶性进展,外源性GMFG干预可以部分逆转。进一步分析显示,ITGB1高肿瘤细胞计数与SPP1+巨噬细胞密度呈正相关,以及这两种细胞类型的空间接近度。临床上,观察到IER2/ITGB1高表达与肿瘤侵袭性表型和低患者生存率显著相关.总的来说,研究结果表明,ERS-CAF通过IER2/GMFG/ITGB1轴调节SPP1+巨噬细胞加重脊索瘤进展,将来可能会有针对性的治疗。
    Currently, the oncogenic mechanism of endoplasmic reticulum stress-CAF (ERS-CAF) subpopulation in chordoma remains unknown. Here, single-cell RNA sequencing, spatial transcriptomics, GeoMx Digital Spatial Profiler, data-independent acquisition proteomics, bulk RNA-seq, and multiplexed quantitative immunofluorescence are used to unveil the precise molecular mechanism of how ERS-CAF affected chordoma progression. Results show that hypoxic microenvironment reprograms CAFs into ERS-CAF subtype. Mechanistically, this occurrs via hypoxia-mediated transcriptional upregulation of IER2. Overexpression of IER2 in CAFs promotes chordoma progression, which can be impeded by IER2 knockdown or use of ERS inhibitors. IER2 also induces expression of ERS-CAF marker genes and results in production of a pro-tumorigenic paracrine GMFG signaling, which exert its biological function via directly binding to ITGB1 on tumor cells. ITGB1 inhibition attenuates tumor malignant progression, which can be partially reversed by exogenous GMFG intervention. Further analyses reveal a positive correlation between ITGB1high tumor cell counts and SPP1+ macrophage density, as well as the spatial proximity of these two cell types. Clinically, a significant correlation of high IER2/ITGB1 expression with tumor aggressive phenotype and poor patient survival is observed. Collectively, the findings suggest that ERS-CAF regulates SPP1+ macrophage to aggravate chordoma progression via the IER2/GMFG/ITGB1 axis, which may be targeted therapeutically in future.
    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
    转移是肿瘤进展的关键阶段,和癌症相关成纤维细胞(CAFs)通过参与细胞外基质(ECM)刚度来支持转移。CD248是非小细胞肺癌(NSCLC)衍生的CAFs的可能生物标志物,但其在介导ECM硬度促进NSCLC转移中的作用尚不清楚。我们研究了CD248+CAFs在激活Hippo轴和促进结缔组织生长因子(CTGF)表达中的意义。这影响基质胶原蛋白I环境并提高ECM硬度,从而促进NSCLC转移。在这项研究中,我们发现CAFs中较高的CD248水平诱导了I型胶原的形成,这反过来又增加了细胞外基质的硬度,从而使NSCLC细胞浸润和迁移。CD248+CAFs激活河马轴诱导CTGF表达,这促进了基质基质中胶原蛋白I环境的形成。在使用成纤维细胞特异性CD248基因敲除小鼠的肿瘤肺转移模型中,与WT小鼠相比,CD248基因敲除小鼠显示出发生肿瘤肺转移的能力显着降低。我们的研究结果表明,CD248+CAFs激活了河马通路,从而诱导CTGF表达,这反过来又促进了基质基质的胶原蛋白I环境,促进NSCLC转移。
    Metastasis is a crucial stage in tumour progression, and cancer-associated fibroblasts (CAFs) support metastasis through their participation in extracellular matrix (ECM) stiffness. CD248 is a possible biomarker for non-small cell lung cancer (NSCLC)-derived CAFs, but its role in mediating ECM stiffness to promote NSCLC metastasis is unknown. We investigated the significance of CD248+ CAFs in activating the Hippo axis and promoting connective tissue growth factor (CTGF) expression, which affects the stromal collagen I environment and improves ECM stiffness, thereby facilitating NSCLC metastasis. In this study, we found that higher levels of CD248 in CAFs induced the formation of collagen I, which in turn increased extracellular matrix stiffness, thereby enabling NSCLC cell infiltration and migration. Hippo axis activation by CD248+ CAFs induces CTGF expression, which facilitates the formation of the collagen I milieu in the stromal matrix. In a tumour lung metastasis model utilizing fibroblast-specific CD248 gene knockout mice, CD248 gene knockout mice showed a significantly reduced ability to develop tumour lung metastasis compared to that of WT mice. Our findings demonstrate that CD248+ CAFs activate the Hippo pathway, thereby inducing CTGF expression, which in turn facilitates the collagen I milieu of the stromal matrix, which promotes NSCLC metastasis.
    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:结肠癌(CC)患者的癌相关成纤维细胞(CAFs)丰富,预后较差。这里,探讨了CAFs对CC生长和转移的分子调控机制。
    方法:使用RT-qPCR监测基因的表达,免疫印迹,和免疫组织化学。使用CCK-8和克隆形成测定发现细胞活力和增殖。使用伤口愈合和Transwell探测细胞迁移和侵袭。Co-IP用于确定AKT和无名指蛋白之间的相互作用,LIM域交互(RLIM)。建立体内鼠皮下肿瘤模型和转移模型以进一步确定轴。
    结果:结果表明,CAFs通过旁分泌软骨寡聚基质蛋白(COMP)促进CC细胞的生长并激活PI3K/AKT通路。此外,RLIM促进CC细胞的生长,AKT通过磷酸化调节其蛋白稳定性。Further,RLIM促进早幼粒细胞白血病蛋白(PML)的泛素化和降解。体外和体内试验发现,PML过表达可抑制CC的生长和转移,由CAF增强。
    结论:从CAFs排出的COMP通过调节RLIM/PML轴促进CC的生长和转移,为CC的治愈提供新的潜在目标。
    OBJECTIVE: Cancer-associated fibroblasts (CAFs) are abundant in colon cancer (CC) patients with a poor prognosis. Here, the molecular regulatory mechanism of CAFs on CC growth and metastasis was explored.
    METHODS: The genes\' expression was monitored using RT-qPCR, immunoblotting, and immunohistochemistry. Cell viability and proliferation were found using CCK-8 and clone formation assays. The cell migration and invasion were probed using wound healing and Transwell. Co-IP was utilized for ascertaining the interaction between AKT and the ring finger protein, LIM domain interacting (RLIM). The in vivo murine subcutaneous tumor model and the metastasis model were built to further ascertain the axis.
    RESULTS: The result showed that CAFs motivate the growth and activate the PI3K/AKT pathway of CC cells via paracrine cartilage oligomeric matrix protein (COMP). Moreover, RLIM promoted the growth of CC cells, and its protein stability was regulated by AKT through its phosphorylation. Further, RLIM facilitated the ubiquitination and degradation of promyelocytic leukemia protein (PML). The in vitro and in vivo tests found that PML overexpression could inhibit CC\'s growth and metastasis, which were enhanced by CAFs.
    CONCLUSIONS: The COMP excreted from CAFs enhances the CC\'s growth and metastasis through regulating the RLIM/PML axis, supplying a new potential target for the cure of CC.
    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
    目的:尽管我们以前的数据表明claudin18同工型2(CLDN18.2)靶向的嵌合抗原受体(CAR)T细胞在CLDN18.2阳性胃癌中表现出显著的临床疗效,它们在胰腺导管腺癌(PDAC)中的疗效有限.肿瘤微环境(TME)是CAR-T功效的主要障碍之一,重塑TME可能是克服这一障碍的可能方法。PDAC的TME以丰富的癌症相关成纤维细胞(CAFs)为特征,这阻碍了CLDN18.2靶向CAR-T细胞的浸润和功能。成纤维细胞活化卵白α(FAP)的表达是活性CAFs的重要特征,提供消除CAF的潜在目标。
    方法:在本研究中,我们产生了10个FAP/CLDN18.2双靶向CAR-T细胞,并在体外和体内评估了它们的抗肿瘤能力。
    结果:与常规CAR-T细胞相比,一些双靶向CAR-T细胞在小鼠胰腺癌中显示出改善的治疗效果.Further,具有更好抗肿瘤作用的双靶向CAR-T细胞可以抑制髓源性抑制细胞(MDSCs)的募集,以改善免疫抑制TME,这有助于CD8+T细胞的存活。此外,双靶向CAR-T细胞以转化TGF-β依赖性方式减少了T细胞的耗竭。
    结论:双靶向CAR-T细胞获得了T效应子功能的增强,抑制T细胞耗尽,改善肿瘤微环境。我们的发现为PDAC中的双靶向FAP/CLDN18.2CAR-T细胞治疗提供了理论基础。
    OBJECTIVE: Although our previous data indicated that claudin 18 isoform 2 (CLDN18.2)-targeted chimeric antigen receptor (CAR) T cells displayed remarkable clinical efficacy in CLDN18.2-positive gastric cancer, their efficacy is limited in pancreatic ductal adenocarcinoma (PDAC). The tumour microenvironment (TME) is one of the main obstacles to the efficacy of CAR-T and remodelling the TME may be a possible way to overcome this obstacle. The TME of PDAC is characterized by abundant cancer-related fibroblasts (CAFs), which hinder the infiltration and function of CLDN18.2-targeted CAR-T cells. The expression of fibroblast activation protein alpha (FAP) is an important feature of active CAFs, providing potential targets for eliminating CAFs.
    METHODS: In this study, we generated 10 FAP/CLDN 18.2 dual-targeted CAR-T cells and evaluated their anti-tumour ability in vitro and in vivo.
    RESULTS: Compared with conventional CAR-T cells, some dual-targeted CAR-T cells showed improved therapeutic effects in mouse pancreatic cancers. Further, dual-targeted CAR-T cells with better anti-tumour effect could suppress the recruitment of myeloid-derived suppressor cells (MDSCs) to improve the immunosuppressive TME, which contributes to the survival of CD8+ T cells. Moreover, dual-targeted CAR-T cells reduced the exhaustion of T cells in transforming TGF-β dependent manner.
    CONCLUSIONS: The dual-targeted CAR-T cells obtained enhancement of T effector function, inhibition of T cell exhaustion, and improvement of tumour microenvironment. Our findings provide a theoretical rationale for dual-targeted FAP/CLDN 18.2 CAR-T cells therapy in PDAC.
    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
    获得性耐药是奥希替尼治疗肺腺癌(LUAD)疗效的主要障碍,但是潜在的机制仍然没有完全理解。癌症相关成纤维细胞(CAF)是LUAD肿瘤微环境(TME)中最丰富的基质细胞类型,并已成为化学耐药性的关键参与者。然而,CAFs在奥希替尼耐药中的作用尚不清楚.这里,我们显示,来自奥希替尼耐药LUAD组织(CAFOR)的CAFs产生的集落刺激因子2(CSF2)比来自奥希替尼敏感组织的CAFs产生的集落刺激因子2(CSF2)多得多.CAFOR衍生的CSF2激活了LUAD细胞中的Janus激酶2(JAK2)/信号转导和转录激活因子3(STAT3)信号通路,并上调了lnc-CSRNP3。然后,Lnc-CSRNP3通过募集色域解旋酶DNA结合蛋白9(CHD9)促进附近基因CSRNP3的表达,并抑制丝氨酸/苏氨酸蛋白磷酸酶1催化亚基α(PP1α)的磷酸酶活性,从而通过增强核糖体生物发生诱导奥希替尼耐药。总的来说,我们的研究揭示了CAFs在奥希替尼耐药发展中的关键作用,并确定CSF2途径是监测LUAD患者奥希替尼疗效和克服奥希替尼耐药的有吸引力的靶标.
    Acquired resistance is a major obstacle to the therapeutic efficacy of osimertinib in lung adenocarcinoma (LUAD), but the underlying mechanisms are still not fully understood. Cancer-associated fibroblasts (CAFs) are the most abundant stromal cell type in LUAD tumor-microenvironment (TME) and have emerged as a key player in chemoresistance. However, the function of CAFs in osimertinib resistance is still unclear. Here, we showed that CAFs derived from osimertinib-resistant LUAD tissues (CAFOR) produced much more colony-stimulating factor 2 (CSF2) than those isolated from osimertinib-sensitive tissues. CAFOR-derived CSF2 activated the Janus kinase 2 (JAK2)/Signal transducer and activator of transcription 3 (STAT3) signaling pathway and upregulated lnc-CSRNP3 in LUAD cells. Lnc-CSRNP3 then promoted the expression of nearby gene CSRNP3 by recruiting chromodomain helicase DNA binding protein 9 (CHD9) and inhibited the phosphatase activity of the serine/threonine protein phosphatase 1 catalytic subunit α (PP1α), thereby induced osimertinib resistance by enhancing ribosome biogenesis. Collectively, our study reveals a critical role for CAFs in the development of osimertinib resistance and identifies the CSF2 pathway as an attractive target for monitoring osimertinib efficacy and overcoming osimertinib resistance in LUAD.
    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
    癌细胞的一个关键特征是它们能够诱导其微环境的变化,使其允许肿瘤生长。侵袭和转移。的确,这些变化是肿瘤进展所必需的.因此,肿瘤微环境正在成为抗癌新靶点的关键来源,有了旨在逆转肿瘤促进变化的新疗法,恢复对肿瘤不利的微环境并抑制疾病进展。RHO-ROCK信令,以及随之而来的细胞肌动球蛋白细胞骨架内的张力,调节旁分泌信号级联,建立促进肿瘤的微环境。这里,我们证明了与我们在乳腺癌中的观察结果一致,增强的ROCK活性和随后产生的CRELD2与皮肤鳞状细胞癌中癌症相关成纤维细胞的募集和促进肿瘤的极化有关。我们的观察结果为以下观点提供了支持:RHO-ROCK信号传导在建立促进肿瘤的微环境中的作用可能在患者之间以及潜在的不同癌症类型之间得到保留。
    A key characteristic of cancer cells is their ability to induce changes in their microenvironment that render it permissive to tumor growth, invasion and metastasis. Indeed, these changes are required for tumor progression. Consequently, the tumor microenvironment is emerging as a key source of new targets against cancer, with novel therapies aimed at reversing tumor-promoting changes, reinstating a tumor-hostile microenvironment and suppressing disease progression. RHO-ROCK signaling, and consequent tension within the cellular actomyosin cytoskeleton, regulates a paracrine signaling cascade that establishes a tumor-promoting microenvironment. Here, we show that consistent with our observations in breast cancer, enhanced ROCK activity and consequent production of CRELD2 is associated with the recruitment and tumor-promoting polarization of cancer-associated fibroblasts in cutaneous squamous cell carcinoma. Our observations provide support for the notion that the role of RHO-ROCK signaling in establishing a tumor-promoting microenvironment may be conserved across patients and potentially also different cancer types.
    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
    前列腺癌(PCa)中癌症相关成纤维细胞(CAFs)的特异性和临床相关性,以及雄激素剥夺治疗(ADT)对CAF的影响,还有待充分阐明。利用细胞谱系多样性和加权基因共表达网络分析(WGCNA),我们确定了PCa独有的CAF签名。通过单细胞RNA测序(scRNA-seq)验证了该CAF特征的特异性,细胞系RNA测序,和免疫组织化学。该特征将CAFs与肿瘤进展相关联,格里森分数升高,和去势抵抗前列腺癌(CRPC)的出现。在收集的样本上使用scRNA-seq,我们证明了CAF特异性签名不会被ADT改变,保持其峰值信号输出。识别PCa特异性CAF特征并观察ADT后CAF的信号变化为进一步的PCa研究奠定了重要的基础。
    The specificity and clinical relevance of cancer-associated fibroblasts (CAFs) in prostate cancer (PCa), as well as the effect of androgen deprivation therapy (ADT) on CAFs, remain to be fully elucidated. Using cell lineage diversity and weighted gene co-expression network analysis (WGCNA), we pinpointed a unique CAF signature exclusive to PCa. The specificity of this CAF signature was validated through single-cell RNA sequencing (scRNA-seq), cell line RNA sequencing, and immunohistochemistry. This signature associates CAFs with tumor progression, elevated Gleason scores, and the emergence of castration resistant prostate cancer (CRPC). Using scRNA-seq on collected samples, we demonstrated that the CAF-specific signature is not altered by ADT, maintaining its peak signal output. Identifying a PCa-specific CAF signature and observing signaling changes in CAFs after ADT lay essential groundwork for further PCa studies.
    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
    癌症相关成纤维细胞(CAF)是肿瘤微环境中的主要细胞成分,并已被证明在肝细胞癌(HCC)中表现出促肿瘤作用。本研究旨在探讨CAFs在HCC中的肿瘤促进作用的潜在机制。进行小RNA测序以筛选源自CAF和正常成纤维细胞(NFs)的外来体中差异表达的微小RNA。然后使用逆转录酶定量实时PCR在CAFs中测量miR-92a-3p表达,NFs,CAFs来源的外泌体(CAFs-Exo),和NF衍生的外泌体(NFs-Exo)。与NF或NF-Exo相比,CAFs和CAFs-Exo显著促进HCC细胞增殖,迁移,和干劲。此外,与NF或NF-Exo相比,miR-92a-3p水平在CAFs和CAFs-Exo中明显更高,分别。发现外泌体miR-92a-3p可增强HCC细胞增殖,迁移,和干劲。同时,AXIN1被miR-92a-3p靶向。外泌体miR-92a-3p可通过抑制AXIN1信使RNA激活肝癌细胞β-catenin/CD44信号传导。此外,体内研究证实,外泌体miR-92a-3p通过靶向AXIN1/β-catenin轴显着促进肿瘤生长和干性。总的来说,CAFs分泌的外泌体miR-92a-3p能够通过抑制AXIN1激活Wnt/β-catenin信号通路来促进HCC的生长和干性。因此,靶向CAFs来源的miR-92a-3p可能是治疗HCC的潜在策略.
    Cancer-associated fibroblasts (CAFs) are a major cellular component in the tumor microenvironment and have been shown to exhibit protumorigenic effects in hepatocellular carcinoma (HCC). This study aimed to delve into the mechanisms underlying the tumor-promoting effects of CAFs in HCC. Small RNA sequencing was conducted to screen differential expressed microRNAs in exosomes derived from CAFs and normal fibroblasts (NFs). The miR-92a-3p expression was then measured using reverse transcriptase quantitative real-time PCR in CAFs, NFs, CAFs-derived exosomes (CAFs-Exo), and NF-derived exosomes (NFs-Exo). Compared to NFs or NF-Exo, CAFs and CAFs-Exo significantly promoted HCC cell proliferation, migration, and stemness. Additionally, compared to NFs or NF-Exo, miR-92a-3p level was notably higher in CAFs and CAFs-Exo, respectively. Exosomal miR-92a-3p was found to enhance HCC cell proliferation, migration, and stemness. Meanwhile, AXIN1 was targeted by miR-92a-3p. Exosomal miR-92a-3p could activate β-catenin/CD44 signaling in HCC cells by inhibiting AXIN1 messenger RNA. Furthermore, in vivo studies verified that exosomal miR-92a-3p notably promoted tumor growth and stemness through targeting AXIN1/β-catenin axis. Collectively, CAFs secreted exosomal miR-92a-3p was capable of promoting growth and stemness in HCC through activation of Wnt/β-catenin signaling pathway by suppressing AXIN1. Therefore, targeting CAFs-derived miR-92a-3p may be a potential strategy for treating HCC.
    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
    肿瘤相关成纤维细胞(CAFs)是肿瘤微环境中丰富且异质的基质细胞,通过将外泌体转移到癌细胞中,在调节肿瘤进展和治疗抗性方面发挥重要作用。然而,CAFs如何调节食管鳞状细胞癌(ESCC)的进展和放射抗性仍未完全了解.采用免疫组织化学方法对174例接受手术治疗的ESCC患者和78例接受确定性放化疗的ESCC患者的预处理活检标本进行了成纤维细胞活化蛋白(FAP)在CAFs中的表达。我们根据FAP表达对CAFs进行排序,收集条件培养基(CM)以培养ESCC细胞。在源自FAP+CAF和FAP-CAF的外泌体中测量被认为调节ESCC进展和/或放射抗性的几种lncRNA的表达水平。随后,细胞计数试剂盒-8,5-乙炔基-2'-脱氧尿苷,transwell,菌落形成,进行和异种移植物测定以研究FAP+CAF和FAP-CAF之间的功能差异。最后,我们采用一系列体外和体内试验来评价AFAP1-AS1对ESCC细胞放射敏感性的影响.基质CAFs中FAP的表达与神经浸润呈正相关,血管浸润,入侵深度,淋巴结转移,缺乏临床完全缓解和生存率差。用CM/FAP+CAFs培养ESCC细胞显著增加肿瘤增殖,迁移,入侵和抗辐射,与CM/FAP-CAF培养物相比。重要的是,FAP+CAF通过将功能性lncRNAAFAP1-AS1经由外泌体直接转移至ESCC细胞而发挥作用。功能研究表明,AFAP1-AS1通过增强ESCC细胞的DNA损伤修复来促进放射抗性。临床上,ESCC患者血浆AFAP1-AS1水平升高与dCRT反应不良相关.我们的发现表明,FAP+CAFs通过转移外泌体lncRNAAFAP1-AS1促进ESCC细胞的放射抗性;并且可能是ESCC治疗的潜在治疗靶标。
    Cancer-associated fibroblasts (CAFs) are abundant and heterogeneous stromal cells in the tumor microenvironment, which play important roles in regulating tumor progression and therapy resistance by transferring exosomes to cancer cells. However, how CAFs modulate esophageal squamous cell carcinoma (ESCC) progression and radioresistance remains incompletely understood. The expression of fibroblast activation protein (FAP) in CAFs was evaluated by immunohistochemistry in 174 ESCC patients who underwent surgery and 78 pretreatment biopsy specimens of ESCC patients who underwent definitive chemoradiotherapy. We sorted CAFs according to FAP expression, and the conditioned medium (CM) was collected to culture ESCC cells. The expression levels of several lncRNAs that were considered to regulate ESCC progression and/or radioresistance were measured in exosomes derived from FAP+ CAFs and FAP- CAFs. Subsequently, cell counting kit-8, 5-ethynyl-2\'-deoxyuridine, transwell, colony formation, and xenograft assays were performed to investigate the functional differences between FAP+ CAFs and FAP- CAFs. Finally, a series of in vitro and in vivo assays were used to evaluate the effect of AFAP1-AS1 on radiosensitivity of ESCC cells. FAP expression in stromal CAFs was positively correlated with nerve invasion, vascular invasion, depth of invasion, lymph node metastasis, lack of clinical complete response and poor survival. Culture of ESCC cells with CM/FAP+ CAFs significantly increased cancer proliferation, migration, invasion and radioresistance, compared with culture with CM/FAP- CAFs. Importantly, FAP+ CAFs exert their roles by directly transferring the functional lncRNA AFAP1-AS1 to ESCC cells via exosomes. Functional studies showed that AFAP1-AS1 promoted radioresistance by enhancing DNA damage repair in ESCC cells. Clinically, high levels of plasma AFAP1-AS1 correlated with poor responses to dCRT in ESCC patients. Our findings demonstrated that FAP+ CAFs promoted radioresistance in ESCC cells through transferring exosomal lncRNA AFAP1-AS1; and may be a potential therapeutic target for ESCC treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号