cancer-associated fibroblast

癌症相关成纤维细胞
  • 文章类型: Journal Article
    背景:癌症相关成纤维细胞(CAF)是肿瘤微环境(TME)中突出的细胞类型,已经在各种肿瘤中鉴定了CAF亚群。然而,CAF如何在空间上协调肝脏TME内的其他细胞群体以促进癌症进展尚不清楚.
    方法:我们结合了多区域蛋白质组学(6例患者,24个样本),10X基因组学空间转录组学(11名患者,25个样品),和多重成像(92名患者,264个样本)破译表达异质性的技术,功能多样性,空间分布,共同定位,和成纤维细胞的相互作用。通过从5名肝癌患者中分离的细胞和体外功能测定来验证新鉴定的CAF亚群。
    结果:我们确定了肝脏CAF亚群,以COL1A2、COL4A1、COL4A2、CTGF、和FSTL1,并命名为F5-CAF。F5-CAF优先位于肿瘤巢内和周围,并与肝细胞癌(HCC)中具有较高干性的癌细胞共定位。92例患者的多重染色和371例患者的大量转录组表明,HCC中F5-CAFs的丰度与预后较差有关。进一步的体外实验表明,从肝癌患者中分离的F5-CAFs可以促进HCC细胞的增殖和干性。
    结论:我们确定了肝癌中的CAF亚群F5-CAF,这与癌症的干性和不良预后有关。我们的结果提供了TME中CAF亚群通过支持癌症干细胞的存活来促进肝癌发展的潜在机制。
    BACKGROUND: Cancer-associated fibroblasts (CAFs) are the prominent cell type in the tumor microenvironment (TME), and CAF subsets have been identified in various tumors. However, how CAFs spatially coordinate other cell populations within the liver TME to promote cancer progression remains unclear.
    METHODS: We combined multi-region proteomics (6 patients, 24 samples), 10X Genomics Visium spatial transcriptomics (11 patients, 25 samples), and multiplexed imaging (92 patients, 264 samples) technologies to decipher the expression heterogeneity, functional diversity, spatial distribution, colocalization, and interaction of fibroblasts. The newly identified CAF subpopulation was validated by cells isolated from 5 liver cancer patients and in vitro functional assays.
    RESULTS: We identified a liver CAF subpopulation, marked by the expression of COL1A2, COL4A1, COL4A2, CTGF, and FSTL1, and named F5-CAF. F5-CAF is preferentially located within and around tumor nests and colocalizes with cancer cells with higher stemness in hepatocellular carcinoma (HCC). Multiplexed staining of 92 patients and the bulk transcriptome of 371 patients demonstrated that the abundance of F5-CAFs in HCC was associated with a worse prognosis. Further in vitro experiments showed that F5-CAFs isolated from liver cancer patients can promote the proliferation and stemness of HCC cells.
    CONCLUSIONS: We identified a CAF subpopulation F5-CAF in liver cancer, which is associated with cancer stemness and unfavorable prognosis. Our results provide potential mechanisms by which the CAF subset in the TME promotes the development of liver cancer by supporting the survival of cancer stem cells.
    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)的治疗选择有限。中医药在TNBC的治疗中起着重要作用。草本植物对ScutellariabarbataD.Don和Scleromitriondiffusum(威尔德。)R.J.Wang(SH)因其抗肿瘤特性而在临床实践中常用。已被证明对肿瘤相关疾病有良好的治疗作用,但是潜在的分子机制尚未完全解释。
    目的:通过生物信息学,验证了主要来自癌症相关成纤维细胞(CAF)的IL6,与预后不良有关。此外,细胞和动物实验证实SH抑制肿瘤增殖,迁移,和通过抑制IL6/NFκB途径在原位肿瘤模型中的生长。
    方法:GEO,使用TCGA和HPA数据库分析CAFs和IL6的预后价值,然后检测IL6资源。在生物信息学之后,通过体外和体内实验验证了CAFs和CAFs衍生的IL6对TNBC的影响。细胞克隆形成试验,伤口愈合试验,用Transwell法检测体外对CAFs和CAFs来源的IL6的促进作用和对SH的抑制作用。使用小鼠中的TNBC模型来证明体内CAF和CAF衍生的IL6的促进和SH的抑制。通过检测独特的分子,通过蛋白质印迹法探索NF-κB的生物学途径。
    结果:生物信息学分析显示,高比例的CAFs和升高的IL6水平与TNBC的不良预后显著相关。同时,IL6被证明主要来源于CAF。在生物信息学的指示之后,体外实验表明,CAFs和IL6均能增强MDA-MD-231细胞的克隆形成和迁移能力(231),此外,CAFs的促进与IL6水平有关。基于这些数据,CAFs来源的IL6增强与NF-κB信号通路的激活密切相关,而SH可以减少活化。最后,体内实验证实了CAFs/CAFs衍生的IL6/NF-κB的促进作用和SH抑制的功效。
    结论:生物信息学数据表明,较高比例的CAFs和较高水平的CAFs衍生的IL6与TNBC较差的存活率显著相关。CAFs和CAFs衍生的IL6被证明在体外和体内都能促进TNBC的进展,其过程与NF-κB的激活密切相关。SH抑制了TNBC的进展,证明与CAFs/CAFs衍生的IL6/NF-κB密切相关。
    BACKGROUND: The treatment options for triple-negative breast cancer (TNBC) are limited. Traditional Chinese Medicine (TCM) plays an important role in the treatment of TNBC. The herb pair Scutellaria barbata D.Don and Scleromitrion diffusum (Willd.) R.J.Wang (SH) is commonly used in clinical practice for its anti-tumor properties. It has been proven to have good therapeutic effects on tumor-related diseases, but the underlying molecular mechanisms are not yet fully explained.
    OBJECTIVE: Through bioinformatics, it was validated that IL6, primarily derived from cancer-associated fibroblasts (CAFs), is associated with poor prognosis. Additionally, cell and animal experiments confirmed that SH inhibits tumor proliferation, migration, and growth in an orthotopic tumor model by suppressing the IL6/NF-κB pathway.
    METHODS: GEO, TCGA and HPA databases were used to analyze the prognostic value of CAFs and IL6, then IL6 resource was detected. After the bioinformatics, the influence of CAFs and CAFs-derived IL6 on TNBC was verified by experiments both in vitro and in vivo. Cell clone formation assay, wound-Healing assay, and Transwell assay were used to detect the promotion of CAFs and CAFs-derived IL6 and the inhibition of SH in vitro. TNBC model in mice was used to prove the promotion of CAFs and CAFs-derived IL6 and the inhibition of SH in vivo. The biological pathway of NF-κB was explored by western blotting through detecting unique molecules.
    RESULTS: Bioinformatics analysis revealed that higher proportion of CAFs and elevated level of IL6 were significantly associated with poor prognosis in TNBC. At the same time, IL6 was proved predominantly derived from CAFs. After the indication of bioinformatics, experiments in vitro demonstrated that both CAFs and IL6 could enhance the clone formation and migration ability of MDA-MD-231 cells (231), furthermore, the promotion of CAFs was related with the level of IL6. Based on these data, mechanism was detected that CAFs-derived IL6 enhancement was closely related to the activation of NF-κB signaling pathway, while the activation can be reduced by SH. In the end, the promotion of CAFs/CAFs-derived IL6/NF-κB and the efficacy of SH inhibition were both confirmed by experiments in vivo.
    CONCLUSIONS: Bioinformatics data indicates that higher proportion of CAFs and higher level of CAFs-derived IL6 are significantly related to poorer survival of TNBC. CAFs and CAFs-derived IL6 were proved to promote the progression of TNBC both in vitro and in vivo, and the process of which was significantly related to the activation of NF-κB. SH inhibited the progress of TNBC, which was proved to be closely related to CAFs/CAFs-derived IL6/NF-κB.
    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
    前列腺癌干细胞的功能异质性和生态位,它们是前列腺癌发展和治疗抵抗的主要驱动因素,已经引起了相当多的研究关注。癌症相关成纤维细胞(CAFs),它们是肿瘤微环境(TME)的关键组成部分,大大影响PCSC的干性。此外,CAF通过释放信号分子和改变周围环境来促进PCSC生长和存活。相反,PCSCs可能通过产生各种分子来影响CAFs的特性和行为。这种串扰机制对于前列腺癌的进展和治疗抗性的发展可能至关重要。使用类器官对TME进行建模,可以深入研究CAF-PCSC相互作用,提供了一个有价值的临床前工具来准确评估潜在的靶基因和设计新的前列腺癌治疗策略。这篇综述的目的是讨论当前关于CAF-PCSC相互作用和串扰的多水平和多目标调节机制的研究,旨在为解决前列腺癌治疗挑战的治疗方法提供信息。
    The functional heterogeneity and ecological niche of prostate cancer stem cells (PCSCs), which are major drivers of prostate cancer development and treatment resistance, have attracted considerable research attention. Cancer-associated fibroblasts (CAFs), which are crucial components of the tumor microenvironment (TME), substantially affect PCSC stemness. Additionally, CAFs promote PCSC growth and survival by releasing signaling molecules and modifying the surrounding environment. Conversely, PCSCs may affect the characteristics and behavior of CAFs by producing various molecules. This crosstalk mechanism is potentially crucial for prostate cancer progression and the development of treatment resistance. Using organoids to model the TME enables an in-depth study of CAF-PCSC interactions, providing a valuable preclinical tool to accurately evaluate potential target genes and design novel treatment strategies for prostate cancer. The objective of this review is to discuss the current research on the multilevel and multitarget regulatory mechanisms underlying CAF-PCSC interactions and crosstalk, aiming to inform therapeutic approaches that address challenges in prostate cancer treatment.
    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
    癌症相关成纤维细胞(CAFs),肿瘤微环境的组成部分,在肿瘤增殖中起关键作用,转移,和临床结果。然而,其在肾肾透明细胞癌(KIRC)中的具体作用尚不清楚。采用已建立的Seurat单细胞分析管道,我们鉴定了21个CAFs标记基因。随后,由6个CAFs标记基因组成的预后特征(RGS5,PGF,TPM2,GJA4,SEPT4和PLXDC1)是通过单变量和LASSOCox回归分析在队列中开发的。然后在外部队列中验证模型的功效,在1-,3-,和5年。高风险组的患者表现出明显较差的生存结果(p<0.001),风险评分是影响预后的独立因素(p<0.05)。在两个风险组之间观察到免疫细胞谱和药物敏感性的明显差异。在KIRC,PGF-VEGFR1信号通路显著增加。肿瘤组织中PGF表达显著升高,如定量实时聚合酶链反应所示。体外,transwell分析和CCK8显示重组PGF可以增强细胞增殖能力,迁移,和侵袭769P和786-O细胞。本研究首次建立了基于6个CAFs基因的KIRC预测模型。此外,PGF可能是增强KIRC治疗的潜在治疗靶标。
    Cancer-associated fibroblasts (CAFs), integral components of the tumor microenvironment, play a pivotal role in tumor proliferation, metastasis, and clinical outcomes. However, its specific roles in Kidney Renal Clear Cell Carcinoma (KIRC) remain poorly understood. Employing the established Seurat single-cell analysis pipeline, we identified 21 CAFs marker genes. Subsequently, a prognostic signature consisting of 6 CAFs marker genes (RGS5, PGF, TPM2, GJA4, SEPT4, and PLXDC1) was developed in a cohort through univariate and LASSO Cox regression analyses. The model\'s efficacy was then validated in an external cohort, with a remarkable predictive performance in 1-, 3-, and 5-year. Patients in the high-risk group exhibited significantly inferior survival outcomes (p < 0.001), and the risk score was an independent prognostic factor (p < 0.05). Distinct differences in immune cell profiles and drug susceptibility were observed between the two risk groups. In KIRC, the PGF-VEGFR1 signaling pathway displayed a notable increase. PGF expression was significantly elevated in tumor tissues, as demonstrated by quantitative real-time polymerase chain reaction. In vitro, transwell assays and CCK8 revealed that recombinant-PGF could enhance the capability of cell proliferation, migration, and invasion in 769P and 786-O cells. This study firstly developed a novel predictive model based on 6 CAFs genes for KIRC. Additionally, PGF may present a potential therapeutic target to enhance KIRC treatment.
    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
    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: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
    背景:骨膜素(POSTN)是各种肿瘤微环境中的关键细胞外基质蛋白。然而,POSTN在甲状腺癌进展中的作用目前尚不清楚.方法:使用Postn和Rag1敲除小鼠和原位小鼠模型来确定POSTN在甲状腺乳头状肿瘤进展中的作用。免疫荧光,细胞共培养,荧光原位杂交,染色质免疫沉淀测定,重组蛋白和抑制剂治疗探讨POSTN促进甲状腺乳头状瘤生长的潜在机制。结果:POSTN在甲状腺乳头状瘤中表达上调,与甲状腺癌患者的总体生存率呈负相关。癌症相关成纤维细胞(CAF)衍生的POSTN在体内和体外促进甲状腺乳头状肿瘤的生长。CAFs中的POSTN缺乏显着损害CAF促进的甲状腺乳头状肿瘤的生长。POSTN通过整合素-FAK-STAT3信号传导促进甲状腺乳头状瘤细胞增殖和IL-4表达.反过来,肿瘤细胞来源的IL-4通过激活STAT6诱导CAF的激活并刺激POSTN表达。我们揭示了CAF衍生的POSTN和肿瘤细胞衍生的IL-4在通过肿瘤细胞中的POSTN-整合素-FAK-STAT3-IL-4通路和CAF中的IL-4-STAT6-POSTN信号传导驱动乳头状甲状腺肿瘤发展中的关键作用。结论:我们的发现强调了POSTN和IL-4作为关键分子介质在CAFs和肿瘤细胞之间的动态相互作用中的意义。最终支持甲状腺乳头状肿瘤的生长。
    Background: Periostin (POSTN) is a critical extracellular matrix protein in various tumor microenvironments. However, the function of POSTN in thyroid cancer progression remains largely unknown. Methods: Postn and Rag1 knock-out mice and orthotopic mouse models were used to determine the role of POSTN on papillary thyroid tumor progression. Immunofluorescence, cell co-culture, fluorescence in situ hybridization, chromatin immunoprecipitation assay, recombinant protein and inhibitor treatment were performed to explore the underlying mechanisms of POSTN-promoted papillary thyroid tumor growth. Results: POSTN is up-regulated in papillary thyroid tumors and negatively correlates with the overall survival of patients with thyroid cancer. Cancer-associated fibroblast (CAF)-derived POSTN promotes papillary thyroid tumor growth in vivo and in vitro. POSTN deficiency in CAFs significantly impairs CAF-promoted papillary thyroid tumor growth. POSTN promotes papillary thyroid tumor cell proliferation and IL-4 expression through integrin-FAK-STAT3 signaling. In turn, tumor cell-derived IL-4 induces the activation of CAFs and stimulates POSTN expression by activating STAT6. We reveal the crucial role of CAF-derived POSTN and tumor cell-derived IL-4 in driving the development of papillary thyroid tumors through the POSTN-integrin-FAK-STAT3-IL-4 pathway in tumor cells and IL-4-STAT6-POSTN signaling in CAFs. Conclusion: Our findings underscore the significance of POSTN and IL-4 as critical molecular mediators in the dynamic interplay between CAFs and tumor cells, ultimately supporting the growth of papillary thyroid 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
    癌相关成纤维细胞(CAFs)表现出明显的表型异质性,与各种恶性肿瘤的免疫抑制有关的特定亚群。然而,它们是否以及如何减弱胃癌(GC)的抗肿瘤免疫仍然难以捉摸。CPT1C,一种独特的肉碱棕榈酰转移酶亚型,在调节脂肪酸氧化中起关键作用,简要表示为GC的肿瘤微环境(TME)中的原瘤代谢介质。在本研究中,我们最初确定了专门过表达CPT1C的成纤维细胞的特定亚群,在此将它们称为CPT1C+CAF。随后的研究结果表明,CPT1C+CAFs促进了基质富集和免疫抑制的TME,因为它们与细胞外基质相关的分子特征和两个免疫抑制亚群的富集相关。尤其是M2样巨噬细胞,和多种免疫相关途径。接下来,我们发现CPT1C+CAFs在体外促进巨噬细胞M2样表型.生物信息学分析揭示了CPT1C+CAFs和巨噬细胞M2样表型之间强大的IL-6信号传导,并将CPT1C+CAFs确定为IL-6的主要来源。同时,抑制CAFs中CPT1C的表达在体外显著降低IL-6的分泌。最后,我们证明了CPT1C+CAFs与治疗耐药的相关性.值得注意的是,在临床队列中,高CPT1C+CAFs浸润的GC患者对免疫治疗的反应较差。总的来说,我们的数据不仅提出了CPT1C+CAFs作为GC的TME中免疫抑制亚群的新鉴定,而且还揭示了CPT1CCAFs通过分泌IL-6诱导GC中巨噬细胞的免疫抑制M2样表型而损害肿瘤免疫的潜在机制。
    Cancer-associated fibroblasts (CAFs) exhibit remarkable phenotypic heterogeneity, with specific subsets implicated in immunosuppression in various malignancies. However, whether and how they attenuate anti-tumor immunity in gastric cancer (GC) remains elusive. CPT1C, a unique isoform of carnitine palmitoyltransferase pivotal in regulating fatty acid oxidation, is briefly indicated as a protumoral metabolic mediator in the tumor microenvironment (TME) of GC. In the present study, we initially identified specific subsets of fibroblasts exclusively overexpressing CPT1C, hereby termed them as CPT1C+CAFs. Subsequent findings indicated that CPT1C+CAFs fostered a stroma-enriched and immunosuppressive TME as they correlated with extracellular matrix-related molecular features and enrichment of both immunosuppressive subsets, especially M2-like macrophages, and multiple immune-related pathways. Next, we identified that CPT1C+CAFs promoted the M2-like phenotype of macrophage in vitro. Bioinformatic analyses unveiled the robust IL-6 signaling between CPT1C+CAFs and M2-like phenotype of macrophage and identified CPT1C+CAFs as the primary source of IL-6. Meanwhile, suppressing CPT1C expression in CAFs significantly decreased IL-6 secretion in vitro. Lastly, we demonstrated the association of CPT1C+CAFs with therapeutic resistance. Notably, GC patients with high CPT1C+CAFs infiltration responded poorly to immunotherapy in clinical cohort. Collectively, our data not only present the novel identification of CPT1C+CAFs as immunosuppressive subsets in TME of GC, but also reveal the underlying mechanism that CPT1C+CAFs impair tumor immunity by secreting IL-6 to induce the immunosuppressive M2-like phenotype of macrophage in GC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号