cancer-associated fibroblast

癌症相关成纤维细胞
  • 文章类型: Journal Article
    在过去的十年里,表皮生长因子受体(EGFR)靶向治疗改变了晚期实体瘤患者的治疗前景.尽管取得了这些进展,抗EGFR治疗的耐药性仍然是一个重大的临床挑战.虽然细胞自主的抵抗机制是有据可查的,他们没有完全阐明耐药性的复杂性。癌症相关成纤维细胞(CAFs),肿瘤微环境(TME)内的关键介质,已经成为癌症进展和化疗耐药性的关键参与者。最近的证据表明CAFs对抗EGFR治疗耐药,这表明它们可能会破坏治疗效果。这篇综述综合了当前的数据,强调CAFs在耐药发病机制中的关键作用,并总结最近针对CAFs的治疗策略。我们强调挑战,并倡导探索CAF作为一种潜在的双目标方法。
    Over the last decade, epidermal growth factor receptor (EGFR)-targeted therapies have transformed the treatment landscape for patients with advanced solid tumors. Despite these advances, resistance to anti-EGFR therapies is still a significant clinical challenge. While cell-autonomous mechanisms of resistance are well-documented, they do not fully elucidate the complexity of drug resistance. Cancer-associated fibroblasts (CAFs), key mediators within the tumor microenvironment (TME), have emerged as pivotal players in cancer progression and chemoresistance. Recent evidence implicates CAFs in resistance to anti-EGFR therapies, suggesting they may undermine treatment efficacy. This review synthesizes current data, highlighting the critical role of CAFs in resistance pathogenesis and summarizing recent therapeutic strategies targeting CAFs. We underscore the challenges and advocate for the exploration of CAFs as a potential dual-targeted approach.
    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
    肿瘤微环境中的肿瘤-基质相互作用和基质异质性是影响进展的关键因素。转移,和胰腺导管腺癌(PDAC)的化学耐药性。这里,我们使用空间转录组技术,利用PDAC小鼠模型(LSL-KrasG12D/+;LSL-Trp53R172H/+;和Pdx-1-Cre小鼠),对生物活性黑磷纳米材料(生物活性BP)治疗后原发性PDAC和肝转移性PDAC的基因表达情况进行了分析.生物信息学和生化分析表明,生物活性BP通过抑制癌症相关成纤维细胞(CAF)的激活而有助于肿瘤-基质的相互作用。我们的结果表明,生物活性BP有助于CAF异质性通过减少炎性CAF和肌纤维母细胞CAF的量,两个CAF亚群。我们的研究证明了生物活性BP对肿瘤-基质相互作用和CAF异质性的影响,并建议生物活性BP作为潜在的PDAC治疗。
    Tumor-stromal interactions and stromal heterogeneity in the tumor microenvironment are critical factors that influence the progression, metastasis, and chemoresistance of pancreatic ductal adenocarcinoma (PDAC). Here, we used spatial transcriptome technology to profile the gene expression landscape of primary PDAC and liver metastatic PDAC after bioactive black phosphorus nanomaterial (bioactive BP) treatment using a murine model of PDAC (LSL-KrasG12D/+; LSL-Trp53R172H/+; and Pdx-1-Cre mice). Bioinformatic and biochemical analyses showed that bioactive BP contributes to the tumor-stromal interplay by suppressing cancer-associated fibroblast (CAF) activation. Our results showed that bioactive BP contributes to CAF heterogeneity by decreasing the amount of inflammatory CAFs and myofibroblastic CAFs, two CAF subpopulations. Our study demonstrates the influence of bioactive BP on tumor-stromal interactions and CAF heterogeneity and suggests bioactive BP as a potential PDAC 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

    了解肿瘤微环境在确定乳腺癌亚型分子标志物中的关键作用在诊断和治疗中非常重要。因此,在肿瘤微环境(TME)的控制下,管腔状态的相互转换及其特异性标志物改变的可能性,特别是癌症相关成纤维细胞(CAFs)值得进一步研究。
    为了激活正常人成纤维细胞,使用液体覆盖技术或止血技术,并表达α-SMA蛋白,CAFs标记,在成纤维细胞球体中通过印迹测量。管腔A,MCF-7和腔B,将MDA-MB361细胞系用正常和球形/活化的成纤维细胞条件培养基处理48小时。通过倒置光学显微镜评估腔A和B细胞的形态变化,并通过形状因子公式进行分析。此外,化学敏感性,扩散,并通过MTT法分别评估ER相关基因和增殖基因表达水平的变化,Ki67表达免疫荧光测定,实时PCR和膜联蛋白V-FITC技术。
    激活(球形)成纤维细胞,表达αSMA标记比单层培养的成纤维细胞多两倍。我们的研究表明,用条件培养基处理后,腔A和B细胞系的IC50显着增加,特别是在用球形条件培养基处理的组中。使用形状因子公式研究形态变化表明,通过增加暴露时间,在腔A和B亚型中获得间充质特征具有更大的侵略性。用成纤维细胞和球形旁分泌体处理后,观察到Ki67表达的变化。来自Ki67测定的驱动数据通过管腔A中Ki67表达升高和管腔B中Ki67表达降低来支持管腔A和B的相互转换。基因表达分析显示,用条件培养基处理的两种管腔类型中的抗凋亡Bcl2基因表达已经增加,尽管在管腔A(CFM7)和管腔B(MDA-MB361)亚型之间的ER相关和增殖基因的表达没有互换,AnnexinV-FITC流式细胞术检测结果表明,与对照组相比,用成纤维细胞和球形条件培养基处理组的早期和晚期凋亡细胞数量均减少.
    在成纤维细胞的旁分泌作用下,乳腺癌的管腔A(MCF7)和管腔B(MDA-MB)亚型均获得侵袭性,抗凋亡,和化学抗性特征,其主要通过模拟CAF的活化(球状)成纤维细胞条件培养基增加。没有强有力的证据证明管腔A和管腔B在乳腺癌分子亚型之间具有更多的相似性。

    UNASSIGNED: Understanding the key role of the tumor microenvironment in specifying molecular markers of breast cancer subtypes is of a high importance in diagnosis and treatment. Therefore, the possibility of interconversion of luminal states and their specific markers alteration under the control of tumor microenvironment (TME), particularly cancer-associated fibroblasts (CAFs) deserves to be further investigated.
    UNASSIGNED: To activate normal human fibroblasts, liquid overlay technique or nemosis was used and α-SMA protein expression, CAFs marker, in fibroblastic spheroids was measured by blotting. The luminal A, MCF-7, and luminal B, MDA-MB 361, cell lines were treated with normal and spheroidal/activated fibroblast conditioned medium for 48 hours. The morphological changes of both luminal A and B cells were evaluated by invert light microscopy and analyzed through the shape factor formula. Moreover, chemo-sensitivity, proliferation, and changes in ER-related and proliferative genes expression levels were assessed respectively via MTT assay, Ki67 expression Immunofluorescence assay, real time PCR and Annexin V-FITC techniques.
    UNASSIGNED: Activated (spheroidal) fibroblasts, expressed αSMA marker two folds more than monolayer cultured fibroblasts. Our study indicated a significant increase in IC50 of both luminal A and B cell lines after being treated with conditioned medium particularly in treated group with spheroidal conditioned medium. Studying Morphological changes using shape factor formula demonstrated more aggressiveness with gaining mesenchymal features in both luminal A and B subtypes by increasing exposure time. Changes in the expression of Ki67 were observed following treatment with fibroblastic and spheroidal paracrine secretome. Driven Data from Ki67 assay supports the luminal A and B interconversion by elevated Ki67 expression in luminal A and lowered Ki67 expression in luminal B. Gene expression analysis revealed that anti-apoptotic Bcl2 gene expression in both luminal types treated with condition medium has been increased though there has seen no interchange in expression of ER-related and proliferative genes between luminal A (MCF7) and luminal B (MDA-MB361) subtypes, the results of Annexin V-FITC flow cytometry test indicated a decrease in the population of both early and late apoptotic cells in groups treated with both fibroblastic and spheroidal condition medium compared to of control group.
    UNASSIGNED: Under the paracrine influence of fibroblast cells, both luminal A (MCF7) and luminal B (MDA-MB) subtypes of breast cancer gained invasive, anti-apoptotic, and chemoresistance features which are mostly increased by activated(spheroidal) fibroblasts conditioned medium mimicking CAFs. There was no strong proof for interconversion of luminal A and luminal B which share more similarities among breast cancer molecular subtypes.
    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:三阴性乳腺癌(TNBC)的特征在于其高转移潜力,导致患者生存率低下。癌相关成纤维细胞(CAF)在通过诱导线粒体生物发生促进TNBC转移方面至关重要。然而,如何抑制CAF赋予的线粒体生物合成仍需探索。
    方法:我们使用伤口愈合和细胞侵袭试验研究了转移,3D文化,anoikis检测,和NOD/SCID小鼠。线粒体生物发生通过MitoTracker绿色FM染色检测,线粒体DNA水平的定量,和蓝色天然聚丙烯酰胺凝胶电泳。表达式,转录,通过蛋白质印迹法检测过氧化物酶体增殖物激活受体共激活因子1α(PGC-1α)的磷酸化,染色质免疫沉淀,双荧光素酶报告分析,定量聚合酶链反应,免疫沉淀,和液相色谱-串联质谱。使用Kaplan-Meier绘图仪数据库和临床乳腺癌组织样本评估了PGC-1α在TNBC中的预后作用。
    结果:我们证明PGC-1α提示淋巴结转移,肿瘤血栓形成,TNBC患者的生存率很低,它是由CAFs诱导的,在TNBC中充当线粒体生物发生和转移的诱导剂。紫草素阻碍了CAF诱导的PGC-1α表达,核定位,以及与雌激素相关受体α(ERRα)的相互作用,从而抑制靶向PGC-1α/ERRα的线粒体基因。机械上,PGC-1α的下调是由合酶激酶3β诱导的PGC-1α在Thr295的磷酸化介导的,这与神经前体细胞表达的发育下调的4e1识别以及随后通过泛素蛋白水解的降解有关。PGC-1α在Thr295的突变消除了紫草素对CAF刺激的TNBC线粒体生物发生和转移的体外和体内抑制作用。
    结论:我们的研究结果表明,PGC-1α是通过破坏线粒体生物发生来阻断TNBC转移的可行靶标,紫草素具有通过靶向PGC-1α作为线粒体生物发生抑制剂治疗TNBC转移的潜力。
    BACKGROUND: Triple-negative breast cancer (TNBC) is characterized by its high metastatic potential, which results in poor patient survival. Cancer-associated fibroblasts (CAFs) are crucial in facilitating TNBC metastasis via induction of mitochondrial biogenesis. However, how to inhibit CAF-conferred mitochondrial biogenesis is still needed to explore.
    METHODS: We investigated metastasis using wound healing and cell invasion assays, 3D-culture, anoikis detection, and NOD/SCID mice. Mitochondrial biogenesis was detected by MitoTracker green FM staining, quantification of mitochondrial DNA levels, and blue-native polyacrylamide gel electrophoresis. The expression, transcription, and phosphorylation of peroxisome-proliferator activated receptor coactivator 1α (PGC-1α) were detected by western blotting, chromatin immunoprecipitation, dual-luciferase reporter assay, quantitative polymerase chain reaction, immunoprecipitation, and liquid chromatography-tandem mass spectrometry. The prognostic role of PGC-1α in TNBC was evaluated using the Kaplan-Meier plotter database and clinical breast cancer tissue samples.
    RESULTS: We demonstrated that PGC-1α indicated lymph node metastasis, tumor thrombus formation, and poor survival in TNBC patients, and it was induced by CAFs, which functioned as an inducer of mitochondrial biogenesis and metastasis in TNBC. Shikonin impeded the CAF-induced PGC-1α expression, nuclear localization, and interaction with estrogen-related receptor alpha (ERRα), thereby inhibiting PGC-1α/ERRα-targeted mitochondrial genes. Mechanistically, the downregulation of PGC-1α was mediated by synthase kinase 3β-induced phosphorylation of PGC-1α at Thr295, which associated with neural precursor cell expressed developmentally downregulated 4e1 recognition and subsequent degradation by ubiquitin proteolysis. Mutation of PGC-1α at Thr295 negated the suppressive effects of shikonin on CAF-stimulated TNBC mitochondrial biogenesis and metastasis in vitro and in vivo.
    CONCLUSIONS: Our findings indicate that PGC-1α is a viable target for blocking TNBC metastasis by disrupting mitochondrial biogenesis, and that shikonin merits potential for treatment of TNBC metastasis as an inhibitor of mitochondrial biogenesis through targeting PGC-1α.
    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:国际肺癌研究协会(IASLC)分级系统可预测早期肺腺癌的预后。
    方法:本研究的目的是检查IASLC分级系统的预后价值及其与I期EGFR减弱的肺腺癌中肿瘤微环境(TME)的相关性。基于IASLC分级系统,我们比较了EGFR突变肺腺癌的临床病理特征(n=296).此外,我们检测了E-cadherin在肿瘤细胞中的表达水平,并计数了肿瘤浸润淋巴细胞(TIL;CD8,CD20,CD138和Foxp3)的数量,肿瘤相关巨噬细胞(TAMs;CD204),和癌症相关的成纤维细胞(CAFs;podoplanin)使用半自动数字病理图像分析。
    结果:无复发生存期(RFS)曲线显示,3级生存期明显短于1级(P<0.01)和2级(P=0.03)。RFS的多变量分析显示侵入性大小,淋巴渗透,3级(P<0.01)是独立的不良预后因素。3级CD204+TAMs和PDPN+CAFs的数量明显高于1级或2级(均P<0.01)。在中间等级中,按基于主要亚型的分类,新分类分类为3级的病例的CD204+TAM(P<0.01)和PDPN+CAF(P=0.02)数量高于2级.
    结论:IASLC分级系统与EGFR突变肺腺癌的预后相关。发现3级具有对肿瘤进展最重要的TME,这可能解释了他们预后不佳的原因。
    BACKGROUND: The International Association for the Study of Lung Cancer (IASLC) grading system predicts early lung adenocarcinoma outcomes.
    METHODS: The purpose of this study is to examine prognostic value of the IASLC grading system and its association with the tumor microenvironment (TME) in Stage I EGFR-muted lung adenocarcinoma. Based on the IASLC grading system, we compared the clinicopathological characteristics of EGFR-mutated lung adenocarcinoma (n = 296). In addition, we examined the expression level of E-cadherin in tumor cells and counted the number of tumor-infiltrating lymphocytes (TILs; CD8, CD20, CD138, and Foxp3), tumor-associated macrophages (TAMs; CD204), and cancer-associated fibroblasts (CAFs; podoplanin) using semi-automatic digital pathology image analysis.
    RESULTS: Recurrence-free survival (RFS) curve showed that survival of grade 3 was significantly shorter than that of grade 1 (P < 0.01) and grade 2 (P = 0.03). Multivariate analysis of RFS revealed the invasive size, lymphatic permeation, and grade 3 (P < 0.01) as independent poor prognostic factors. The number of CD204 +TAMs and PDPN+CAFs was significantly higher in grade 3 than in grade 1 or 2 (all P < 0.01). Among the intermediate grade by the predominant subtype based classification, cases classified as grade 3 by the new classification had higher number of CD204 +TAMs (P < 0.01) and PDPN+CAFs (P = 0.02) than those classified as grade 2.
    CONCLUSIONS: The IASLC grading system correlated with the outcomes of EGFR-mutated lung adenocarcinoma. Grade 3 was found to have the TME that most contributes to tumor progression, which probably explained their poor prognosis.
    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
    结直肠癌,全球第三大最常见的肿瘤,由于其在转移性病例中的高死亡率和持续的耐药性,因此提出了挑战。我们调查了肿瘤的微环境,强调癌症相关成纤维细胞在结直肠癌进展和化疗耐药中的作用。我们使用包含结肠直肠癌类器官和癌症相关成纤维细胞的间接共培养系统来模拟肿瘤微环境。免疫荧光染色验证了类器官和成纤维细胞的特征,显示上皮细胞标志物(EPCAM)的高表达,结肠癌标志物(CK20),增殖标志物(KI67),和成纤维细胞标记(VIM,SMA)。转录组分析是在用抗癌药物治疗后进行的,如5-氟尿嘧啶和奥沙利铂,鉴定与化学抗性相关的基因。抗癌药物治疗后共培养的结直肠癌类器官中基因表达的变化,与单一类器官相比,特别是在与干扰素-α/β信号传导和主要组织相容性复合物II类蛋白质复合物组装相关的途径中,已确定。这两个基因群可能介导与JAK/STAT信号相关的药物抗性。结直肠癌类器官和成纤维细胞之间的相互作用至关重要地调节与耐药性相关的基因的表达。这些发现表明结直肠癌类器官和成纤维细胞之间的相互作用显著影响与耐药性相关的基因表达。突出潜在的生物标志物和治疗靶点,以克服化学耐药性。增强对癌细胞与其微环境之间相互作用的理解可以导致个性化医学研究的进步。.
    Colorectal cancer, the third most commonly occurring tumor worldwide, poses challenges owing to its high mortality rate and persistent drug resistance in metastatic cases. We investigated the tumor microenvironment, emphasizing the role of cancer-associated fibroblasts in the progression and chemoresistance of colorectal cancer. We used an indirect co-culture system comprising colorectal cancer organoids and cancer-associated fibroblasts to simulate the tumor microenvironment. Immunofluorescence staining validated the characteristics of both organoids and fibroblasts, showing high expression of epithelial cell markers (EPCAM), colon cancer markers (CK20), proliferation markers (KI67), and fibroblast markers (VIM, SMA). Transcriptome profiling was conducted after treatment with anticancer drugs, such as 5-fluorouracil and oxaliplatin, to identify chemoresistance-related genes. Changes in gene expression in the co-cultured colorectal cancer organoids following anticancer drug treatment, compared to monocultured organoids, particularly in pathways related to interferon-alpha/beta signaling and major histocompatibility complex class II protein complex assembly, were identified. These two gene groups potentially mediate drug resistance associated with JAK/STAT signaling. The interaction between colorectal cancer organoids and fibroblasts crucially modulates the expression of genes related to drug resistance. These findings suggest that the interaction between colorectal cancer organoids and fibroblasts significantly influences gene expression related to drug resistance, highlighting potential biomarkers and therapeutic targets for overcoming chemoresistance. Enhanced understanding of the interactions between cancer cells and their microenvironment can lead to advancements in personalized medical research..
    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
    由于癌症相关成纤维细胞的表达上调,成纤维细胞活化蛋白(FAP)已成为实体瘤成像和治疗的有吸引力的生物标志物。虽然许多FAP配体已经被开发用于放射性药物治疗(RPT),大多数人患有肿瘤摄取不足,肿瘤停留时间不足,或者在健康组织中的脱靶积累,表明需要进一步改进。方法:通过结合先前几种配体靶向RPT的理想特征,设计了具有新型配体(FAP8-PEG3-IP-DOTA)的新型FAP靶向RPT。[111In]In或[177Lu]Lu-FAP8-PEG3-IP-DOTA的摄取和保留以KB为单位进行评估,通过放射成像或离体生物分布分析的HT29、MDA-MB-231和4T1鼠肿瘤模型。还通过监测肿瘤生长来研究放射治疗效力和总体毒性。体重,和荷瘤小鼠的组织损伤。结果:FAP8-PEG3-IP-DOTA表现出高亲和力(半最大抑制浓度,1.6nM)和相对于其最接近的同系物对FAP的良好选择性,丙氨酰基寡肽酶(半最大抑制浓度,~14.0nM)和二肽基肽酶-IV(半最大抑制浓度,~860nM)。SPECT/CT扫描在2种不同的实体瘤模型中显示出高保留和在健康组织中的最小摄取。定量生物分布分析显示,所有主要器官的肿瘤与健康组织的比率都超过5倍。活体动物研究表明,在所有4个测试模型中,肿瘤生长抑制65%-93%,具有最小或没有全身毒性的证据。结论:我们得出结论,[177Lu]Lu-FAP8-PEG3-IP-DOTA是FAPα靶向放射性核素治疗实体瘤的有希望且安全的RPT候选物。
    Because of upregulated expression on cancer-associated fibroblasts, fibroblast activation protein (FAP) has emerged as an attractive biomarker for the imaging and therapy of solid tumors. Although many FAP ligands have already been developed for radiopharmaceutical therapies (RPTs), most suffer from inadequate tumor uptake, insufficient tumor residence times, or off-target accumulation in healthy tissues, suggesting a need for further improvements. Methods: A new FAP-targeted RPT with a novel ligand (FAP8-PEG3-IP-DOTA) was designed by combining the desirable features of several previous ligand-targeted RPTs. Uptake and retention of [111In]In or [177Lu]Lu-FAP8-PEG3-IP-DOTA were assessed in KB, HT29, MDA-MB-231, and 4T1 murine tumor models by radioimaging or ex vivo biodistribution analyses. Radiotherapeutic potencies and gross toxicities were also investigated by monitoring tumor growth, body weight, and tissue damage in tumor-bearing mice. Results: FAP8-PEG3-IP-DOTA exhibited high affinity (half-maximal inhibitory concentration, 1.6 nM) and good selectivity for FAP relative to its closest homologs, prolyl oligopeptidase (half-maximal inhibitory concentration, ∼14.0 nM) and dipeptidyl peptidase-IV (half-maximal inhibitory concentration, ∼860 nM). SPECT/CT scans exhibited high retention in 2 different solid tumor models and minimal uptake in healthy tissues. Quantitative biodistribution analyses revealed tumor-to-healthy-tissue ratios of more than 5 times for all major organs, and live animal studies demonstrated 65%-93% suppression of tumor growth in all 4 models tested, with minimal or no evidence of systemic toxicity. Conclusion: We conclude that [177Lu]Lu-FAP8-PEG3-IP-DOTA constitutes a promising and safe RPT candidate for FAPα-targeted radionuclide therapy of solid tumors.
    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
    在包括皮肤鳞状细胞癌(cSCC)的许多癌症类型中,癌相关成纤维细胞(CAF)通过各种信号传导途径与肿瘤细胞相互作用。在正常成纤维细胞(NF)亚型中,乳头状成纤维细胞(PFs)和网状成纤维细胞(RFs)对肿瘤细胞信号传导有明显的反应,最终实现了RF的差异化,而不是PF,进入CAF。成纤维细胞中亚型分化的调节仍未得到充分探索。在这项研究中,我们评估了PF之间的差异,RFs,和CAF,并检查了靶向TGFβ的小分子抑制剂的作用,PI3K/AKT/mTOR,和NOTCH通路对2D和3D培养中CAF和CAF重编程的肿瘤促进特性的影响。阻断TGFβ和PI3K强烈失活并同时诱导RF和CAF中的PF表型。3D共培养cSCC细胞系MET2与RF或CAF导致增强的肿瘤侵袭,“RF-CAF”转换和细胞因子产生,通过阻断TGFβ和PI3K/mTOR途径进一步抑制,但不是NOTCH途径。总之,这项研究确定了PFs的生物标志物,RF和CAF,并显示出阻断CAF和肿瘤细胞-CAF相互作用中关键信号通路的不同作用。这些发现促使了“CAF到PF”的治疗策略,并提供了在基于CAF的癌症治疗中使用包含的抑制剂的观点。
    Cancer-associated fibroblasts (CAFs) interact reciprocally with tumor cells through various signaling pathways in many cancer types including cutaneous squamous cell carcinoma (cSCC). Among normal fibroblast (NF) subtypes, papillary fibroblasts (PFs) and reticular fibroblasts (RFs) respond distinctly to tumor cell signaling, eventuating the differentiation of RFs, rather than PFs, into CAFs. The regulation of subtype differentiation in fibroblasts remains poorly explored. In this study, we assessed the differences between PFs, RFs, and CAFs, and examined the effects of small-molecule inhibitors targeting the TGFβ, PI3K/AKT/mTOR, and NOTCH pathways on the tumor-promoting property of CAFs and CAF reprogramming in 2D and 3D cultures. Blocking TGFβ and PI3K strongly deactivated and concurrently induced a PF phenotype in RFs and CAFs. 3D co-culturing a cSCC cell line MET2 with RFs or CAFs led to enhanced tumor invasion, \"RF-CAF\" transition and cytokine production, which were further repressed by blocking TGFβ and PI3K/mTOR pathways, but not NOTCH pathway. In conclusion, the study identified biomarkers for PFs, RFs and CAFs, and displayed different effects of blocking key signaling pathways in CAFs and tumor cell-CAF interplay. These findings prompted a \"CAF to PF\" therapeutic strategy, and provided perspectives of using included inhibitors in CAF-based cancer 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
    背景:化疗耐药是结直肠癌(CRC)患者治疗失败的关键原因。化疗耐药CRC的肿瘤微环境具有明显的免疫抑制作用,尽管潜在的机制尚不清楚.方法:从GEO数据库下载CRC数据集GSE69657和GSE62080,用Pearson方法分析TRPC5与FAP表达的相关性。通过免疫组织化学检查了CRC组织中瞬时受体电位通道5(TRPC5)和成纤维细胞激活蛋白(FAP)的原位表达。通过蛋白质印迹和RT-PCR分析了HCT8和HCT116细胞系以及相应的5-氟尿嘧啶(5-FU)抗性细胞系(HCT8R和HCT116R)中的TRPC5表达水平。从HCT8R和HCT116R细胞中分离外泌体,并与结直肠正常成纤维细胞(NFs)孵育,并检测癌相关成纤维细胞(CAFs)标志物。NFs还与从TRPC5敲低HCT8R细胞分离的外泌体一起孵育,并分析了细胞内Ca2水平和C-X-C基序趋化因子配体12(CXCL12)分泌的变化。结果:TRPC5和FAP在数据集中的表达呈正相关。CRC组织标本的免疫染色进一步显示,高TRPC5和FAP表达与较差的肿瘤消退显着相关。此外,与化学敏感细胞相比,化学抗性CRC细胞表达更高水平的TRPC5,和敲低TRPC5逆转化学抗性。源自CRC细胞的外来体诱导NF向CAF的转化。然而,来自化学抗性CRC细胞的TRPC5-外泌体可以促进CAF分泌更多的CXCL12。结论:化疗耐药的CRC细胞可通过外泌体TRPC5诱导CAFs活化并促进CXCL12分泌。
    Background: Chemoresistance is a key reason for treatment failure in colorectal cancer (CRC) patients. The tumor microenvironment of chemoresistant CRC is distinctly immunosuppressive, although the underlying mechanisms are unclear. Methods: The CRC data sets GSE69657 and GSE62080 were downloaded from the GEO database, and the correlation between TRPC5 and FAP expression was analyzed by Pearson method. The in-situ expression of transient receptor potential channel 5 (TRPC5) and fibroblast activation protein (FAP) in the CRC tissues was examined by immunohistochemistry. TRPC5 expression levels in the HCT8 and HCT116 cell lines and the corresponding 5-fluorouracil (5-FU)-resistant cell lines (HCT8R and HCT116R) were analyzed by western blotting and RT-PCR. Exosomes were isolated from the HCT8R and HCT116R cells and incubated with colorectal normal fibroblasts (NFs), and cancer-associated fibroblasts (CAFs)markers were detected. NFs were also incubated with exosomes isolated from TRPC5-knockdown HCT8R cells, and the changes in intracellular Ca2+ levels and C-X-C motif chemokine ligand 12 (CXCL12) secretion were analyzed. Results: TRPC5 and FAP expression showed positive correlation in the datasets. Immunostaining of CRC tissue specimens further revealed that high TRPC5 and FAP expressions were significantly associated with worse tumor regression. Furthermore, chemoresistant CRC cells expressed higher levels of TRPC5 compared to the chemosensitive cells, and knocking down TRPC5 reversed chemoresistance. Exosomes derived from CRC cells induced the transformation of NFs to CAFs. However, TRPC5-exosomes derived from chemoresistant CRC cells can promote CAFs to secrete more CXCL12. Conclusion: Chemoresistant CRC cells can induce CAFs activation and promote CXCL12 secretion through exosomal TRPC5.
    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
    胰腺导管腺癌(PDAC)由于其复杂的微环境而表现出巨大的临床挑战,其特征在于组织增生和复杂的肿瘤-基质相互作用。常规模型阻碍了研究用于治疗开发的细胞串扰。为了概括PDAC肿块的关键特征,这项研究创造了一种新的海岛PDAC肿瘤构建体(s&iPTC)。S&iPTC由位于由人类癌症相关成纤维细胞(CAF)填充的间质细胞外基质(ECM)内的3D打印的人类PDAC细胞岛组成。这种设计紧密地模拟了体内去纤维增生结构和营养贫乏的条件。该模型能够研究动态的肿瘤-基质串扰和信号相互作用,揭示了已知和尚未发现的多细胞代谢适应。使用该模型,我们发现了营养胁迫下CAFs的动态变化,类似于关键的体内人类肿瘤生态位,如促肿瘤炎症因子的分泌。此外,营养缺乏引起ECM组成的动态改变,并加剧了PDAC进展中公认的不良癌细胞分化特征。蛋白质组学分析揭示了与不良营养条件下侵袭性肿瘤行为和ECM重塑相关的蛋白质的富集。模拟无血管胰腺肿瘤核心经历的代谢应激。重要的是,通过S&IPTCs中生物标志物表达模式与PDAC患者生存率之间的负相关,模型与患者结局的相关性很明显.关键发现包括在营养丰富的条件下上调MMP和关键ECM蛋白(如胶原蛋白11和TGFβ)。已知受CAF监管,在营养剥夺的情况下,与低分化PDAC状态相关的E-cadherin表达伴随减少。此外,对营养剥夺的反应中透明质酸(HA)和整合素水平的升高强调了模型对PDAC微环境的保真度。我们还观察到在不良营养条件下IL-6增加和α-SMA表达减少。提示在类似于体内生态位的营养压力下,CAF从肌纤维母细胞向炎症表型的转变。总之,S&IPTC代表了PDAC质量的临床相关3D模型工程的重大进展。它为阐明肿瘤-基质相互作用和指导未来治疗策略以改善患者预后提供了一个有前途的平台。
    Pancreatic ductal adenocarcinoma (PDAC) presents a formidable clinical challenge due to its intricate microenvironment characterized by desmoplasia and complex tumor-stroma interactions. Conventional models hinder studying cellular crosstalk for therapeutic development. To recapitulate key features of PDAC masses, this study creates a novel sea-and-island PDAC tumor construct (s&i PTC). The s&i PTC consists of 3D-printed islands of human PDAC cells positioned within an interstitial extracellular matrix (ECM) populated by human cancer-associated fibroblasts (CAFs). This design closely mimics the in vivo desmoplastic architecture and nutrient-poor conditions. The model enables studying dynamic tumor-stroma crosstalk and signaling reciprocity, revealing both known and yet-to-be-discovered multicellular metabolic adaptations. Using the model, we discovered the orchestrated dynamic alterations of CAFs under nutrient stress, resembling critical in vivo human tumor niches, such as the secretion of pro-tumoral inflammatory factors. Additionally, nutrient scarcity induces dynamic alterations in the ECM composition and exacerbates poor cancer cell differentiation-features well-established in PDAC progression. Proteomic analysis unveiled the enrichment of proteins associated with aggressive tumor behavior and ECM remodeling in response to poor nutritional conditions, mimicking the metabolic stresses experienced by avascular pancreatic tumor cores. Importantly, the model\'s relevance to patient outcomes is evident through an inverse correlation between biomarker expression patterns in the s&i PTCs and PDAC patient survival rates. Key findings include upregulated MMPs and key ECM proteins (such as collagen 11 and TGFβ) under nutrient-avid conditions, known to be regulated by CAFs, alongside the concomitant reduction in E-cadherin expression associated with a poorly differentiated PDAC state under nutrient deprivation. Furthermore, elevated levels of hyaluronic acid (HA) and integrins in response to nutrient deprivation underscore the model\'s fidelity to the PDAC microenvironment. We also observed increased IL-6 and reduced α-SMA expression under poor nutritional conditions, suggesting a transition of CAFs from myofibroblastic to inflammatory phenotypes under a nutrient stress akin to in vivo niches. In conclusion, the s&i PTC represents a significant advancement in engineering clinically relevant 3D models of PDAC masses. It offers a promising platform for elucidating tumor-stroma interactions and guiding future therapeutic strategies to improve patient outcomes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号