CAFs

CAF
  • 文章类型: Journal Article
    背景:肿瘤组织包括癌细胞和基质细胞,它们的相互作用形成了癌症微环境。因此,针对癌细胞以外的细胞的治疗也正在积极开发中,其中,针对PD-1的治疗,PD-1是一种在肿瘤免疫逃避中很重要的免疫检查点分子,也适用于头颈部癌症。PD-L1是PD-1的配体,在肿瘤细胞和基质细胞中均有表达,以及基于两种类型细胞的综合阳性率的评分系统,综合阳性评分(CPS),用于预测治疗效果。然而,关于PD-L1的表达尚不清楚。在这项研究中,我们通过组织病理学检查了控制PD-1/PD-L1表达的因素。这项研究包括在东京牙科学院Suidobashi医院口腔颌面外科接受舌鳞状细胞癌切除手术的37例患者。PD-L1、α-SMA的表达水平,通过免疫组织化学染色评估p53。
    结果:7名参与者CPS≥20,24名参与者1≤CPS<20,6名参与者CPS<1。α-SMA的总体阳性率,癌症相关成纤维细胞(CAFs)的标志物,27%(10/37名参与者),三个CPS组的α-SMA阳性率为85.7%(6/7),16.7%(4/24参与者),和0%(0/6参与者),分别。此外,p53的总体阳性率为37.8%(14/37),三个CPS组的p53阳性率为71.4%(5/7),37.5%(9/24参与者),和0%(0/6参与者),分别。
    结论:PD-L1的表达与α-SMA和p53阳性相关。此外,与p53的表达相比,α-SMA的表达与高CPS患者的PD-L1表达有更高的相关性。上述研究结果表明,CAF之间的相互作用,癌细胞,免疫活性细胞可以调节PD-L1的表达。
    BACKGROUND: Tumor tissues comprise cancer cells and stromal cells, and their interactions form the cancer microenvironment. Therefore, treatments targeting cells other than cancer cells are also actively being developed, and among them, treatment targeting PD-1, an immune checkpoint molecule that is important in tumor immune evasion, has also been indicated for head and neck cancer. PD-L1, a ligand of PD-1, is expressed in both tumor cells and stromal cells, and the scoring system based on the combined positivity rates of both types of cells, the combined positive score (CPS), is used for predicting treatment effect. However, much is unknown regarding the expression of PD-L1. In this study, we histopathologically examined factors controlling the expression of PD-1/PD-L1. This study included 37 patients who underwent resection surgery for tongue squamous cell carcinoma in the Department of Oral and Maxillofacial Surgery at Tokyo Dental College Suidobashi Hospital. The expression levels of PD-L1, α-SMA, and p53 were assessed by immunohistochemical staining.
    RESULTS: Seven participants had CPS ≥ 20, twenty-four participants had 1 ≤ CPS < 20, and six participants had CPS < 1. The overall positivity rate of α-SMA, a marker for cancer-associated fibroblasts (CAFs), was 27% (10/37 participants), and the positivity rates of α-SMA for the three CPS groups were 85.7% (6/7 participants), 16.7% (4/24 participants), and 0% (0/6 participants), respectively. In addition, the overall positivity rate of p53 was 37.8% (14/37 participants), and the positivity rates of p53 for the three CPS groups were 71.4% (5/7 participants), 37.5% (9/24 participants), and 0% (0/6 participants), respectively.
    CONCLUSIONS: The expression of PD-L1 demonstrated an association with α-SMA and p53 positivity. In addition, compared with the expression of p53, the expression of α-SMA demonstrated a higher association with PD-L1 expression in patients with a high CPS. The abovementioned findings suggest that the interactions between CAFs, cancer cells, and immunocompetent cells may regulate the expression of PD-L1.
    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
    光疗可以在杀死肿瘤细胞的同时引起自噬,导致肿瘤复发和转移。这里,我们构建了激光和酶双响应纳米药物递送系统Tf-Te@CTSL-HCQ(TT@CH),以与光疗协同作用精确调节自噬,从而抑制黑色素瘤的增殖和转移。首先,转铁蛋白(Tf)作为纳米反应器,通过生物模板矿化方法合成光疗剂Tf-Te。然后,以FAP-α反应肽(CAP)修饰的热敏脂质体为载体,将自噬抑制剂羟氯喹(HCQ)和Tf-Te,以获得智能TT@CH交付系统。一旦到达肿瘤部位,TT@CH可以通过在癌症相关成纤维细胞(CAF)上过度表达的FAP-α裂解,并释放Tf-Te和HCQ。然后Tf-Te可以靶向黑色素瘤细胞并发挥PTT/PDT抗肿瘤作用。更重要的是,PTT引起的高热可进一步促进TT@CH释放药物。同时,HCQ同时抑制CAFs和黑色素瘤细胞的自噬,并下调IL-6和HMGB1的分泌,从而有效抑制黑素瘤转移。药效学结果显示,TT@CH的抗肿瘤效果最好,抑瘤率最高,达91.3%。同时,与对照组相比,TT@CH治疗的小鼠的肺转移结节减少了124.33。总的来说,TT@CH为黑色素瘤提供了有效的治疗策略。
    Phototherapy can cause autophagy while killing tumour cells, leading to tumour recurrence and metastasis. Here, we constructed a laser and enzyme dual responsive nanodrug delivery system Tf-Te@CTSL-HCQ (TT@CH) to precisely regulate autophagy in synergy with phototherapy to inhibit the proliferation and metastasis of melanoma. Firstly, transferrin (Tf) was used as a nanoreactor to synthesise phototherapy agent Tf-Te by the biological template mineralisation method. Then, the thermosensitive liposome modified with FAP-α-responsive peptide (CAP) was used as a carrier to encapsulate autophagy inhibitor hydroxychloroquine (HCQ) and Tf-Te, to obtain an intelligent TT@CH delivery system. Once arriving at the tumour site, TT@CH can be cleaved by FAP-α overexpressed on cancer-associated fibroblasts (CAFs), and release Tf-Te and HCQ. Then Tf-Te can target melanoma cells and exert PTT/PDT anti-tumour effect. What\'s more, hyperpyrexia induced by PTT can further promote drugs release from TT@CH. Meanwhile, HCQ simultaneously inhibited autophagy of CAFs and melanoma cells, and down-regulated IL-6 and HMGB1 secretion, thus effectively inhibiting melanoma metastasis. Pharmacodynamic results exhibited the best anti-tumour effect of TT@CH with the highest tumour inhibition rate of 91.3%. Meanwhile, lung metastatic nodules of TT@CH treated mice reduced by 124.33 compared with that of mice in control group. Overall, TT@CH provided an effective therapy strategy for melanoma.
    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
    免疫检查点抑制剂(ICIs)有望突破三阴性乳腺癌(TNBC)的治疗和生存困境,然而,由于缺乏效应免疫细胞(内部问题)和由癌症相关成纤维细胞(CAFs)形成的物理屏障(外部限制),只有免疫调节亚型和≈5%的TNBC患者作为单药治疗应答.水凝胶药物递送平台,ALG@TBP-2/Pt(0)/nintedanib(ALG@TPN),旨在诱导强大的免疫功能和内部和外部肿瘤微环境(TME)的双重消除。被白光激活,通过I型和II型光动力疗法(PDT),TBP-2细胞内产生大量的活性氧(ROS),氧化线粒体DNA(mtDNA)。Pt(0)的独特过氧化氢酶活性将内源性H2O2转化为O2,减少缺氧限制性PDT并增强ROS产生功效。丰富的ROS可以将Pt(0)氧化为细胞毒性Pt(II),破坏核DNA(nDNA)。对mtDNA和nDNA的双重损伤可能双向激活cGAS/STING途径并增强免疫细胞应答。此外,尼达尼布对CAF有显著的抑制作用,削弱免疫屏障,加深免疫细胞浸润。总的来说,该研究提供了一种具有“PDT/化疗/抗CAFs”效果的自氧水凝胶,触发cGAS/STING途径重塑TME。内部和外部干预都会增加抗TNBC免疫反应。
    Immune checkpoint inhibitors (ICIs) offer promise in breaking through the treatment and survival dilemma of triple-negative breast cancer (TNBC), yet only immunomodulatory subtype and ≈5% TNBC patients respond as monotherapy due to lack of effector immune cells (internal problem) and physical barrier (external limitation) formed by cancer-associated fibroblasts (CAFs). A hydrogel drug-delivery platform, ALG@TBP-2/Pt(0)/nintedanib (ALG@TPN), is designed to induce strong immune functions and the dual elimination of the internal and external tumor microenvironment (TME). Activated by white light, through type I and II photodynamic therapy (PDT), TBP-2 generates large amounts of reactive oxygen species (ROS) intracellularly, oxidizing mitochondrial DNA (mtDNA). The unique catalase activity of Pt(0) converts endogenous H2O2 to O2, reducing the anoxia-limiting PDT and enhancing ROS generation efficacy. Abundant ROS can oxidize Pt(0) to cytotoxic Pt(II), damaging the nuclear DNA (nDNA). Dual damage to mtDNA and nDNA might bi-directionally activate the cGAS/STING pathway and enhance the immune cell response. Besides, nintedanib demonstrates a significant inhibitory effect on CAFs, weakening the immune barrier and deepening immune cell infiltration. Overall, the study provides a self-oxygenating hydrogel with the \"PDT/chemotherapy/anti-CAFs\" effect, triggering the cGAS/STING pathway to reshape the TME. Both internal and external interventions increase anti-TNBC immune responses.
    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(TME)和细胞外基质(ECM)在癌症研究中至关重要,因为它们对肿瘤进展的影响。Collagens,主要ECM组件,调节细胞信号和行为。在28个报告的胶原蛋白中,已知XII型胶原蛋白对ECM组织至关重要。由癌症相关成纤维细胞(CAF)过度产生,它的上调与各种癌症的低生存率相关。本研究旨在开发用于定量作为癌症生物标志物的循环XII型胶原的ELISA。开发了靶向XII型胶原蛋白的C末端的特异性ELISA,并用于分析来自癌症患者(n=203)和健康对照(n=33)的血清样品。此外,在来自不同组织的CAFs和正常成纤维细胞(NFs)中评估了XII型胶原蛋白的表达,在TGF-β刺激和非刺激条件下。nordicPRO-C12ELISA证明了对XII型胶原的稳健性和特异性。与健康对照相比,在患有各种癌症的患者中PRO-C12水平显著升高,并且在癌症患者和对照之间有效区分。使用基因表达数据验证结果。此外,蛋白质印迹分析显示,在TGF-β1刺激后,CAF和NFs中XII型胶原表达增加,提示TGF-β1在调节癌变和正常组织微环境中XII型胶原表达中的潜在作用。这项研究揭示了利用PRO-C12作为非侵入性血清生物标志物的有希望的途径,能够量化癌症患者的XII型胶原蛋白片段。有必要进行进一步的调查,以探索PRO-C12在不同癌症类型和疾病阶段的潜力。阐明其在癌症发展中的多方面作用。
    Understanding the tumor microenvironment (TME) and extracellular matrix (ECM) is crucial in cancer research due to their impact on tumor progression. Collagens, major ECM components, regulate cell signaling and behavior. Of the 28 reported collagens, type XII collagen is known to be vital for ECM organization. Over-produced by cancer-associated fibroblasts (CAFs), its upregulation correlates with poor survival in various cancers. This study aimed to develop an ELISA for quantifying circulating type XII collagen as a cancer biomarker. A specific ELISA targeting the C-terminal of type XII collagen was developed and used to analyze serum samples from cancer patients (n = 203) and healthy controls (n = 33). Additionally, type XII collagen expression was assessed in CAFs and normal fibroblasts (NFs) from different tissues, both under TGF-β stimulated and non-stimulated conditions. The nordicPRO-C12 ELISA demonstrated robustness and specificity for type XII collagen. PRO-C12 levels were significantly elevated in patients with various cancers compared to healthy controls and effectively distinguished between cancer patients and controls. Findings were validated using gene expression data. Furthermore, Western blot analysis revealed increased type XII collagen expression in both CAFs and NFs upon TGF-β1 stimulation, suggesting a potential role of TGF-β1 in modulating the expression of type XII collagen in cancerous and normal tissue microenvironments. This study unveils a promising avenue for harnessing PRO-C12 as a non-invasive serum biomarker, enabling the quantification of type XII collagen fragments in cancer patients. Further investigations are warranted to explore the potential of PRO-C12 across different cancer types and disease stages, shedding light on its multifaceted role in cancer development.
    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)的特征是免疫逃避,导致预后不良。癌症相关成纤维细胞(CAF)在协调PDAC肿瘤微环境中起关键作用。我们研究了CAF衍生的细胞外囊泡(EV)包装的长链非编码RNA(lncRNA)在免疫逃避中的作用,并探索了使用负载小干扰RNA(siRNA)的工程EV作为潜在治疗策略的基因治疗。我们的发现强调了来自CAF的EV包装的lncRNARP11-161H23.5在通过下调HLA-A表达促进PDAC免疫逃避中的意义,抗原呈递的关键成分。机械上,RP11-161H23.5与CNOT4形成复合物,CNOT4是mRNA死酶CCR4-NOT复合物的一个亚基,通过缩短其poly(A)尾巴来增强HLA-AmRNA的降解。这种免疫逃避机制损害了抗肿瘤免疫应答。为了解决这个问题,我们提出了一种创新的方法,该方法利用工程EV作为基于siRNA的基因治疗的天然和生物相容性纳米载体,该策略有望提高PDAC中免疫治疗的有效性.总的来说,我们的研究阐明了CAF衍生的EV包装的lncRNARP11-161H23.5/CNOT4/HLA-A轴在PDAC免疫逃避中的关键作用,并为治疗干预提供了新的途径.
    Pancreatic ductal adenocarcinoma (PDAC) is characterised by immune evasion that contribute to poor prognosis. Cancer-associated fibroblasts (CAFs) play a pivotal role in orchestrating the PDAC tumour microenvironment. We investigated the role of CAF-derived extracellular vesicle (EV)-packaged long non-coding RNAs (lncRNAs) in immune evasion and explored gene therapy using engineered EVs loading small interfering RNAs (siRNAs) as a potential therapeutic strategy. Our findings highlight the significance of EV-packaged lncRNA RP11-161H23.5 from CAF in promoting PDAC immune evasion by downregulating HLA-A expression, a key component of antigen presentation. Mechanistically, RP11-161H23.5 forms a complex with CNOT4, a subunit of the mRNA deadenylase CCR4-NOT complex, enhancing the degradation of HLA-A mRNA by shortening its poly(A) tail. This immune evasion mechanism compromises the anti-tumour immune response. To combat this, we propose an innovative approach utilising engineered EVs as natural and biocompatible nanocarriers for siRNA-based gene therapy and this strategy holds promise for enhancing the effectiveness of immunotherapy in PDAC. Overall, our study sheds light on the critical role of CAF-derived EV-packaged lncRNA RP11-161H23.5/CNOT4/HLA-A axis in PDAC immune evasion and presents a novel avenue for therapeutic intervention.
    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:探讨癌相关成纤维细胞(CAFs)影响肺癌细胞生长和免疫逃避的机制。
    方法:最初,从GEO数据集GSE48397下载比较CAFs与正常成纤维细胞的数据集.选择具有最显著差异表达的基因并使用临床数据进行验证。随后,CAF被隔离,选择的基因在CAF中被敲低。用H1299或A549细胞进行共培养实验,分析肺癌细胞生长的变化,迁移,和体内外免疫逃避。为了进一步阐明上游调控机制,相关的ChIP-seq数据是从GEO数据库下载的,并通过ChIP-qPCR和荧光素酶报告基因测定验证了调控关系。
    结果:OLR1在CAFs中显著过表达,并且与肺癌患者的不良预后密切相关。OLR1的敲除在体外和体内显着抑制了CAFs对肺癌细胞生长和免疫逃避的支持。ChIP-seq结果表明,PRRX1可以通过募集H3K27ac和H3K4me3促进OLR1表达,从而激活CAF。PRRX1的敲减显着抑制CAFs的功能,而OLR1的进一步过度表达恢复了CAFs对肺癌细胞生长的支持,迁移,和免疫逃避。
    结论:PRRX1通过招募H3K27ac和H3K4me3促进OLR1表达,激活CAFs,从而促进增长,迁移,和肺癌细胞的免疫逃避。
    OBJECTIVE: To investigate the mechanism by which cancer-associated fibroblasts (CAFs) affect the growth and immune evasion of lung cancer cells.
    METHODS: Initially, datasets comparing CAFs with normal fibroblasts were downloaded from the GEO dataset GSE48397. Genes with the most significant differential expression were selected and validated using clinical data. Subsequently, CAFs were isolated, and the selected genes were knocked down in CAFs. Co-culture experiments were conducted with H1299 or A549 cells to analyze changes in lung cancer cell growth, migration, and immune evasion in vitro and in vivo. To further elucidate the upstream regulatory mechanism, relevant ChIP-seq data were downloaded from the GEO database, and the regulatory relationships were validated through ChIP-qPCR and luciferase reporter assays.
    RESULTS: OLR1 was significantly overexpressed in CAFs and strongly correlated with adverse prognosis in lung cancer patients. Knockdown of OLR1 markedly inhibited CAFs\' support for the growth and immune evasion of lung cancer cells in vitro and in vivo. ChIP-seq results demonstrated that PRRX1 can promote OLR1 expression by recruiting H3K27ac and H3K4me3, thereby activating CAFs. Knockdown of PRRX1 significantly inhibited CAFs\' function, while further overexpression of OLR1 restored CAFs\' support for lung cancer cell growth, migration, and immune evasion.
    CONCLUSIONS: PRRX1 promotes OLR1 expression by recruiting H3K27ac and H3K4me3, activating CAFs, and thereby promoting the growth, migration, and immune evasion of lung cancer 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
    胰腺腺癌(PAAD),被称为最致命的癌症之一,以复杂的肿瘤微环境为特征,主要由细胞外基质中的癌症相关成纤维细胞(CAF)组成。这些CAFs通过与透明质酸(HA)和透明质酸酶相互作用显着改变基质,它降解HA-癌症进展和扩散的重要过程。尽管这种互动的关键作用,CAFs和透明质酸酶在PAAD发育中的具体功能尚不完全清楚。我们的研究调查了这种相互作用,并评估了针对透明质酸酶的新型抗癌化合物NSC777201。这项研究利用计算方法来分析基因表达综合(GEO)数据库中的基因表达数据,特别是GSE172096,比较了癌症相关和正常成纤维细胞的基因表达谱。我们对用NSC777201处理的胰腺癌细胞进行了内部测序,以鉴定差异表达基因(DEGs),并进行了功能富集和途径分析。使用TCGA-PAAD和人类蛋白质图谱(HPA)数据库进一步验证了鉴定的DEGs的诊断,预后,和生存影响,伴随着灵巧路径分析(IPA)和NSC777201的分子对接,体外,和临床前体内验证。结果显示与CAF相关的416DEGs和与NSC777201治疗相关的570DEGs,有九个重叠的DEG。一个关键的发现是跨膜蛋白TMEM2,它与FAP密切相关,CAF标记,并且与PAAD的高危人群相关。NSC777201处理显示TMEM2的抑制,通过拯救试验验证,表明靶向TMEM2的重要性。进一步分析,包括IPA,证明NSC777201调节CAF细胞衰老,增强其治疗潜力。体外和体内研究均证实了NSC777201对TMEM2表达的抑制作用,加强其在靶向PAAD中的作用。因此,TMEM2已被确定为PAAD中的一种与热无关的生物标志物,受CAF活性和HA积累的影响。NSC777201在靶向和潜在逆转PAAD进展中的关键过程方面表现出巨大的潜力,证明其作为有前途的治疗剂的功效。
    Pancreatic adenocarcinoma (PAAD), known as one of the deadliest cancers, is characterized by a complex tumor microenvironment, primarily comprised of cancer-associated fibroblasts (CAFs) in the extracellular matrix. These CAFs significantly alter the matrix by interacting with hyaluronic acid (HA) and the enzyme hyaluronidase, which degrades HA - an essential process for cancer progression and spread. Despite the critical role of this interaction, the specific functions of CAFs and hyaluronidase in PAAD development are not fully understood. Our study investigates this interaction and assesses NSC777201, a new anti-cancer compound targeting hyaluronidase. This research utilized computational methods to analyze gene expression data from the Gene Expression Omnibus (GEO) database, specifically GSE172096, comparing gene expression profiles of cancer-associated and normal fibroblasts. We conducted in-house sequencing of pancreatic cancer cells treated with NSC777201 to identify differentially expressed genes (DEGs) and performed functional enrichment and pathway analysis. The identified DEGs were further validated using the TCGA-PAAD and Human Protein Atlas (HPA) databases for their diagnostic, prognostic, and survival implications, accompanied by Ingenuity Pathway Analysis (IPA) and molecular docking of NSC777201, in-vitro, and preclinical in-vivo validations. The result revealed 416 DEGs associated with CAFs and 570 DEGs related to NSC777201 treatment, with nine overlapping DEGs. A key finding was the transmembrane protein TMEM2, which strongly correlated with FAP, a CAF marker, and was associated with higher-risk groups in PAAD. NSC777201 treatment showed inhibition of TMEM2, validated by rescue assay, indicating the importance of targeting TMEM2. Further analyses, including IPA, demonstrated that NSC777201 regulates CAF cell senescence, enhancing its therapeutic potential. Both in-vitro and in-vivo studies confirmed the inhibitory effect of NSC777201 on TMEM2 expression, reinforcing its role in targeting PAAD. Therefore, TMEM2 has been identified as a theragnostic biomarker in PAAD, influenced by CAF activity and HA accumulation. NSC777201 exhibits significant potential in targeting and potentially reversing critical processes in PAAD progression, demonstrating its efficacy as a promising therapeutic agent.
    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
    细胞外基质(ECM)是肿瘤环境的主要组成部分。促进建立亲入侵行为。这种环境由肿瘤和基质衍生的代谢物支持,特别是乳酸。在前列腺癌(PCa)中,癌症相关成纤维细胞(CAFs)是分泌乳酸的主要贡献者,能够影响癌细胞的代谢和转录调控。这里,我们描述了CAF分泌的乳酸在PCa细胞中促进编码胶原蛋白家族的基因表达的机制。利用乳酸的PCa细胞依赖于增加的α-酮戊二酸(α-KG),该α-酮戊二酸激活依赖α-KG的胶原原氨酰-4-羟化酶(P4HA1)以支持胶原羟基化。从头合成的胶原蛋白通过激活盘状结构域受体1(DDR1)发挥信号作用,支持PCa细胞的干细胞样和侵袭性特征。乳酸诱导的胶原羟基化和DDRl活化的抑制减少了PCa细胞的转移性定植。总的来说,这些结果为胶原蛋白重塑/信号传导与PCa利用的营养环境之间的联系提供了新的理解.
    Extracellular matrix (ECM) is a major component of the tumor environment, promoting the establishment of a pro-invasive behavior. Such environment is supported by both tumor- and stromal-derived metabolites, particularly lactate. In prostate cancer (PCa), cancer-associated fibroblasts (CAFs) are major contributors of secreted lactate, able to impact on metabolic and transcriptional regulation in cancer cells. Here, we describe a mechanism by which CAF-secreted lactate promotes in PCa cells the expression of genes coding for the collagen family. Lactate-exploiting PCa cells rely on increased α-ketoglutarate (α-KG) which activates the α-KG-dependent collagen prolyl-4-hydroxylase (P4HA1) to support collagen hydroxylation. De novo synthetized collagen plays a signaling role by activating discoidin domain receptor 1 (DDR1), supporting stem-like and invasive features of PCa cells. Inhibition of lactate-induced collagen hydroxylation and DDR1 activation reduces the metastatic colonization of PCa cells. Overall, these results provide a new understanding of the link between collagen remodeling/signaling and the nutrient environment exploited by PCa.
    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)是最具侵袭性的癌症,具有明显的纤维化,其死亡率在人类癌症中排名第二。癌相关成纤维细胞(CAFs)在PDAC进展中起关键作用,我们回顾了目前对PDACCAFs的分子理解和新的治疗潜力。CAF相关基因(CAFG)根据代表基质/上皮表达比(SE比)的基质特异性初步分为三类。最近使用单细胞转录组技术进行的分类阐明,CAFs由肌成纤维细胞(myCAFs)组成,炎性CAFs(iCAFs),和其他次要的(例如,POSTN-CAFs和抗原呈递CAFs,apCAFs)。LRRC15是myCAFs标记,白喉毒素和myCAF的消耗诱导细胞毒性T淋巴细胞(CTL)的快速积累,因此增强了PDL1抗体治疗。这一发现表明,就PDAC进展而言,myCAFs可能是肿瘤免疫的关键调节因子。myCAFs位于肿瘤细胞附近的CAFs中,虽然PDPN和/或COL14A1标记的iCAF远离肿瘤细胞,假定由于血液供应不足而导致iCAF中的低氧和酸性环境与HIF1A和GPR68表达一致。在衰老CAF中,iCAF可以与SASP(分泌相关表型)共享。myCAFs的经典特征是由TGFB1诱导的CAFGs,而具有SASP的化学抗性CAFs可能依赖于IL6表达并伴有STAT3激活。最近,发现CAFs的独特代谢可以靶向预防PDAC进展,PDAC细胞利用葡萄糖,而CAF反过来利用乳酸,可能是表观遗传调节的,由其靶基因包括CXCR4介导。总之,CAFs具有独特的分子特性,已被严格阐明为PDAC进展的新治疗靶标。
    Pancreatic ductal adenocarcinoma (PDAC) is the most aggressive cancer with striking fibrosis, and its mortality rate is ranked second across human cancers. Cancer-associated fibroblasts (CAFs) play a critical role in PDAC progression, and we reviewed the molecular understanding of PDAC CAFs and novel therapeutic potential at present. CAFs-associated genes (CAFGs) were tentatively classified into three categories by stroma specificity representing stroma/epithelia expression ratios (SE ratios). The recent classification using single cell transcriptome technology clarified that CAFs were composed of myofibroblasts (myCAFs), inflammatory CAFs (iCAFs), and other minor ones (e.g., POSTN-CAFs and antigen presenting CAFs, apCAFs). LRRC15 is a myCAFs marker, and myCAFs depletion by diphtheria toxin induces the rapid accumulation of cytotoxic T lymphocytes (CTLs) and therefore augment PDL1 antibody treatments. This finding proposes that myCAFs may be a critical regulator of tumor immunity in terms of PDAC progression. myCAFs are located in CAFs adjacent to tumor cells, while iCAFs marked by PDPN and/or COL14A1 are distant from tumor cells, where hypoxic and acidic environments being located in iCAFs putatively due to poor blood supply is consistent with HIF1A and GPR68 expressions. iCAFs may be shared with SASP (secretion-associated phenotypes) in senescent CAFs. myCAFs are classically characterized by CAFGs induced by TGFB1, while chemoresistant CAFs with SASP may dependent on IL6 expression and accompanied by STAT3 activation. Recently, it was found that the unique metabolism of CAFs can be targeted to prevent PDAC progression, where PDAC cells utilize glucose, whereas CAFs in turn utilize lactate, which may be epigenetically regulated, mediated by its target genes including CXCR4. In summary, CAFs have unique molecular characteristics, which have been rigorously clarified as novel therapeutic targets of PDAC progression.
    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
    胶质母细胞瘤(GBM)是成人中枢神经系统最常见的恶性肿瘤。GBM具有高水平的治疗失败,其预后通常是令人沮丧的。肿瘤细胞的表型异质性,GBM肿瘤微环境(TME)内非肿瘤细胞群体的动态复杂性,和他们的双向串扰有助于当前治疗方法的挑战。在这里,我们讨论了GBM的病因,并描述了TME中几种主要类型的非肿瘤细胞,它们对GBM发病机制的影响,以及这种影响的分子机制。我们还讨论了它们作为潜在治疗靶标或预后生物标志物的价值,参考关于这个主题的最新作品。我们得出的结论是,除非考虑TME内所有非肿瘤细胞的“关键参与者”群体,在开发GBM治疗方面没有突破。
    Glioblastoma (GBM) is the most common malignancy of the central nervous system in adults. GBM has high levels of therapy failure and its prognosis is usually dismal. The phenotypic heterogeneity of the tumor cells, dynamic complexity of non-tumor cell populations within the GBM tumor microenvironment (TME), and their bi-directional cross-talk contribute to the challenges of current therapeutic approaches. Herein, we discuss the etiology of GBM, and describe several major types of non-tumor cells within its TME, their impact on GBM pathogenesis, and molecular mechanisms of such an impact. We also discuss their value as potential therapeutic targets or prognostic biomarkers, with reference to the most recent works on this subject. We conclude that unless all \"key player\" populations of non-tumor cells within the TME are considered, no breakthrough in developing treatment for GBM can be achieved.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号