molecular oncology

  • 文章类型: Journal Article
    目的:胃癌的基因组研究已经确定了影响RHO信号的高度复发的基因组改变,尤其是在弥漫性胃癌(DGC)组织学亚型中。这些改变包括染色体间翻译,导致粘附蛋白CLDN18和RHO调节因子ARHGAP26融合。尚不清楚这些融合结构如何影响RHO途径的活性以及它们对胃癌发展的更广泛影响。在这里,我们开发了一个模型,使我们能够研究这种融合蛋白在DGC发病机理中的功能,并确定具有这些改变的DGC肿瘤的潜在治疗靶标。
    方法:我们构建了一个转基因小鼠模型,将LSL-CLDN18-ARHGAP26融合物工程化到Col1A1基因座中,其表达可以通过Cre重组酶诱导。利用这个模型产生的类器官,我们评估了其致癌活性和融合蛋白对RHOA途径的生化作用及其在DGC发病机理中的下游细胞生物学效应。
    结果:我们证明了CLDN18-ARHGAP26在胃类器官中的表达诱导了印戒细胞的形成,DGC的特征性特征,当与肿瘤抑制基因Trp53的丢失相结合时,能够协同转化胃细胞。CLDN18-ARHGAP26促进RHOA和下游效应子信号的激活。分子上,融合促进粘着斑激酶(FAK)的激活和YAP途径的诱导。FAK和YAP/TEAD抑制的组合可以显著阻断肿瘤生长。
    结论:这些结果表明CLDN18-ARHGAP26融合是一种功能获得的DGC癌基因,可导致RHOA的激活以及FAK和YAP信号的激活。这些结果为进一步评估这些致命癌症的新兴FAK和YAP-TEAD抑制剂提供了依据。
    Genomic studies of gastric cancer have identified highly recurrent genomic alterations impacting RHO signalling, especially in the diffuse gastric cancer (DGC) histological subtype. Among these alterations are interchromosomal translations leading to the fusion of the adhesion protein CLDN18 and RHO regulator ARHGAP26. It remains unclear how these fusion constructs impact the activity of the RHO pathway and what is their broader impact on gastric cancer development. Herein, we developed a model to allow us to study the function of this fusion protein in the pathogenesis of DGC and to identify potential therapeutic targets for DGC tumours with these alterations.
    We built a transgenic mouse model with LSL-CLDN18-ARHGAP26 fusion engineered into the Col1A1 locus where its expression can be induced by Cre recombinase. Using organoids generated from this model, we evaluated its oncogenic activity and the biochemical effects of the fusion protein on the RHOA pathway and its downstream cell biological effects in the pathogenesis of DGC.
    We demonstrated that induction of CLDN18-ARHGAP26 expression in gastric organoids induced the formation of signet ring cells, characteristic features of DGC and was able to cooperatively transform gastric cells when combined with the loss of the tumour suppressor geneTrp53. CLDN18-ARHGAP26 promotes the activation of RHOA and downstream effector signalling. Molecularly, the fusion promotes activation of the focal adhesion kinase (FAK) and induction of the YAP pathway. A combination of FAK and YAP/TEAD inhibition can significantly block tumour growth.
    These results indicate that the CLDN18-ARHGAP26 fusion is a gain-of-function DGC oncogene that leads to activation of RHOA and activation of FAK and YAP signalling. These results argue for further evaluation of emerging FAK and YAP-TEAD inhibitors for these deadly cancers.
    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:PI3K-AKT通路是否以及如何,代谢稳态的中心节点,高脂诱导的非酒精性脂肪性肝炎(NASH)和肝细胞癌(HCC)仍然是一个谜。在这种情况下,AKT调节的特征将为抗击HCC提供新的策略。
    方法:代谢文库筛选揭示棕榈酸(PA)可以激活AKT。采用体内和体外棕榈酰化测定法检测AKT棕榈酰化。不同的细胞和小鼠模型,包括AKT1C77S和AKT1C224S敲入细胞的产生,使用Zdhhc17和Zdhhc24敲除小鼠和Akt1C224S敲入小鼠。来自NASH和HCC患者的人类肝组织,流体动力学转染小鼠模型,我们还进一步探索了高脂/高胆固醇饮食(HFHCD)诱导的NASH/HCC小鼠模型和高脂和蛋氨酸/胆碱缺乏饮食(HFMCD)诱导的NASH小鼠模型进行机制研究。
    结果:通过筛选代谢物库,PA已被定义为通过促进其棕榈酰修饰来激活AKT,生长因子诱导的AKT激活的必要步骤。生物学,高脂肪饮食可以促进AKT激酶的活性,从而促进NASH和肝癌。机械上,棕榈酰结合以不依赖PIP3的方式将AKT锚定到细胞膜上,部分通过防止AKT组装成非活性聚合物。棕榈酰转移酶ZDHHC17/24被表征为棕榈酰化AKT以发挥致癌作用。有趣的是,抗肥胖药物奥利司他或特异性穿透肽可以通过限制PA合成或抑制AKT修饰来有效减弱AKT棕榈酰化和激活,分别,从而拮抗肝脏肿瘤发生。
    结论:我们的发现阐明了PA-ZDHHC17/24介导的棕榈酰化对AKT的新型微调调节,并通过服用PA限制饮食来强调肿瘤治疗策略,限制PA合成,或直接靶向AKT棕榈酰化。
    Whether and how the PI3K-AKT pathway, a central node of metabolic homeostasis, is responsible for high-fat-induced non-alcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC) remain a mystery. Characterisation of AKT regulation in this setting will provide new strategies to combat HCC.
    Metabolite library screening disclosed that palmitic acid (PA) could activate AKT. In vivo and in vitro palmitoylation assay were employed to detect AKT palmitoylation. Diverse cell and mouse models, including generation of AKT1C77S and AKT1C224S knock-in cells, Zdhhc17 and Zdhhc24 knockout mice and Akt1C224S knock-in mice were employed. Human liver tissues from patients with NASH and HCC, hydrodynamic transfection mouse model, high-fat/high-cholesterol diet (HFHCD)-induced NASH/HCC mouse model and high-fat and methionine/choline-deficient diet (HFMCD)-induced NASH mouse model were also further explored for our mechanism studies.
    By screening a metabolite library, PA has been defined to activate AKT by promoting its palmitoyl modification, an essential step for growth factor-induced AKT activation. Biologically, a high-fat diet could promote AKT kinase activity, thereby promoting NASH and liver cancer. Mechanistically, palmitoyl binding anchors AKT to the cell membrane in a PIP3-independent manner, in part by preventing AKT from assembling into an inactive polymer. The palmitoyltransferases ZDHHC17/24 were characterised to palmitoylate AKT to exert oncogenic effects. Interestingly, the anti-obesity drug orlistat or specific penetrating peptides can effectively attenuate AKT palmitoylation and activation by restricting PA synthesis or repressing AKT modification, respectively, thereby antagonising liver tumorigenesis.
    Our findings elucidate a novel fine-tuned regulation of AKT by PA-ZDHHC17/24-mediated palmitoylation, and highlight tumour therapeutic strategies by taking PA-restricted diets, limiting PA synthesis, or directly targeting AKT palmitoylation.
    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:胆管癌(CCA)是一种异质性恶性肿瘤,死亡率高,预后差,以及临床上对新疗法的迫切需求。CCA表观基因组的知识主要限于异常DNA甲基化。已确定增强子活性的失调会影响致癌作用并用于新疗法,但在CCA中尚未进行研究。我们的目的是通过增强子谱分析来确定CCA不同亚型中的潜在治疗靶标。
    方法:对多种CCA进行了整合的多组学增强子活性分析。一组不同的CCA细胞系,患者来源和细胞系来源的异种移植物被用于研究确定的富集途径和脆弱性.NanoString,多重免疫组织化学染色和单细胞空间转录组学用于探索不同CCA的免疫原性。
    结果:我们确定了三个不同的组,与不同的病因和独特的途径有关。已鉴定途径的药物抑制剂在体外和体内模型中降低了肿瘤生长。第一组(ESTRO)主要是侥幸阳性的CCA,在雌激素信号中显示激活,对MTOR抑制剂敏感.另一组(OXPHO),主要是BAP1和IDH突变的CCA,显示激活的氧化磷酸化途径,并且对氧化磷酸化抑制剂敏感。在最后一组(IMMUN)中激活了免疫相关途径,由免疫原性CCA亚型和具有马兜铃酸(AA)突变特征的CCA组成。AA突变负荷的瘤内差异与不同免疫细胞群体的瘤内变化相关。
    结论:我们的研究阐明了增强子失调的潜在机制,并加深了对不同CCA亚型中不同肿瘤发生过程的理解,具有潜在的显着治疗和临床益处。
    Cholangiocarcinoma (CCA) is a heterogeneous malignancy with high mortality and dismal prognosis, and an urgent clinical need for new therapies. Knowledge of the CCA epigenome is largely limited to aberrant DNA methylation. Dysregulation of enhancer activities has been identified to affect carcinogenesis and leveraged for new therapies but is uninvestigated in CCA. Our aim is to identify potential therapeutic targets in different subtypes of CCA through enhancer profiling.
    Integrative multiomics enhancer activity profiling of diverse CCA was performed. A panel of diverse CCA cell lines, patient-derived and cell line-derived xenografts were used to study identified enriched pathways and vulnerabilities. NanoString, multiplex immunohistochemistry staining and single-cell spatial transcriptomics were used to explore the immunogenicity of diverse CCA.
    We identified three distinct groups, associated with different etiologies and unique pathways. Drug inhibitors of identified pathways reduced tumour growth in in vitro and in vivo models. The first group (ESTRO), with mostly fluke-positive CCAs, displayed activation in estrogen signalling and were sensitive to MTOR inhibitors. Another group (OXPHO), with mostly BAP1 and IDH-mutant CCAs, displayed activated oxidative phosphorylation pathways, and were sensitive to oxidative phosphorylation inhibitors. Immune-related pathways were activated in the final group (IMMUN), made up of an immunogenic CCA subtype and CCA with aristolochic acid (AA) mutational signatures. Intratumour differences in AA mutation load were correlated to intratumour variation of different immune cell populations.
    Our study elucidates the mechanisms underlying enhancer dysregulation and deepens understanding of different tumourigenesis processes in distinct CCA subtypes, with potential significant therapeutics and clinical benefits.
    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:越来越多的证据表明肿瘤细胞表现出与其谱系祖细胞相似的特征。我们发现S100钙结合蛋白A10(S100A10)表现出与肝脏祖细胞基因相似的表达模式。然而,S100A10在肝细胞癌(HCC)进展中的作用尚不清楚。此外,细胞外囊泡(EV)是肿瘤发生和转移的关键介质,但是S100A10的细胞外功能,特别是与电动汽车(EV-S100A10)相关的功能,是未知的。
    方法:在体外和体内研究了S100A10和EV-S100A10在HCC进展中的功能和机制。使用针对S100A10的中和抗体(NA)来评估EV-S100A10的显著性。
    结果:功能上,S100A10促进HCC起始,自我更新,体外和体内的化学抗性和转移。意义重大,我们发现S100A10由HCC细胞在体外和HCC患者血浆中分泌到EV中。富含S100A10的EV增强了HCC细胞的干性和转移能力,上调表皮生长因子受体(EGFR),AKT和ERK信号,促进上皮-间质转化。EV-S100A10还在HCC细胞运动中充当化学引诱物。意义重大,S100A10控制电动汽车中的蛋白质负载,并通过与整合素αⅤ的物理结合介导MMP2,纤连蛋白和EGF与电动汽车膜的结合。重要的是,用S100A10-NA阻断EV-S100A10可显着消除这些增强作用。
    结论:总而言之,我们的结果发现,S100A10通过其在EV中的转移和调节EV的蛋白质载物显著促进HCC进展.EV-S100A10可能是HCC进展的潜在治疗靶标和生物标志物。
    Growing evidence indicates that tumour cells exhibit characteristics similar to their lineage progenitor cells. We found that S100 calcium binding protein A10 (S100A10) exhibited an expression pattern similar to that of liver progenitor genes. However, the role of S100A10 in hepatocellular carcinoma (HCC) progression is unclear. Furthermore, extracellular vesicles (EVs) are critical mediators of tumourigenesis and metastasis, but the extracellular functions of S100A10, particularly those related to EVs (EV-S100A10), are unknown.
    The functions and mechanisms of S100A10 and EV-S100A10 in HCC progression were investigated in vitro and in vivo. Neutralising antibody (NA) to S100A10 was used to evaluate the significance of EV-S100A10.
    Functionally, S100A10 promoted HCC initiation, self-renewal, chemoresistance and metastasis in vitro and in vivo. Of significance, we found that S100A10 was secreted by HCC cells into EVs both in vitro and in the plasma of patients with HCC. S100A10-enriched EVs enhanced the stemness and metastatic ability of HCC cells, upregulated epidermal growth factor receptor (EGFR), AKT and ERK signalling, and promoted epithelial-mesenchymal transition. EV-S100A10 also functioned as a chemoattractant in HCC cell motility. Of significance, S100A10 governed the protein cargos in EVs and mediated the binding of MMP2, fibronectin and EGF to EV membranes through physical binding with integrin αⅤ. Importantly, blockage of EV-S100A10 with S100A10-NA significantly abrogated these enhancing effects.
    Altogether, our results uncovered that S100A10 promotes HCC progression significantly via its transfer in EVs and regulating the protein cargoes of EVs. EV-S100A10 may be a potential therapeutic target and biomarker for HCC 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
    目的:N6-甲基腺苷(m6A)与胰腺导管腺癌(PDAC)的进展密切相关。PDAC的另一个突出特点是代谢重编程,通过肿瘤微环境提供足够的营养来支持细胞的快速生长。然而,m6A相关代谢基因对PDAC微环境的潜在影响仍然知之甚少。因此,我们试图使用m6A相关基因构建生存预测模型,以阐明PDAC的分子特征.
    方法:在本研究中,从癌症基因组图谱(TCGA)胰腺腺癌数据集获得m6A相关代谢基因,并进行共表达分析。一致聚类根据m6A相关代谢基因的表达识别具有不同免疫细胞浸润模式的两个不同亚组。采用多变量Cox回归分析和最小绝对收缩和选择算子(LASSO)分析来创建m6A相关代谢模型。构建了包括临床特征和基于m6A相关代谢调节因子表达的风险评分的列线图。
    结果:包含ATP8B2,GMPS,建立LDHA和SDR39U1来预测PDAC患者的总生存期(OS)。该签名还可靠地预测了来自国际癌症基因组联盟(ICGC)和ArrayExpress(E-MTAB-6134)的两个独立验证队列中的存活。四基因标签将患者分为高风险和低风险组,具有不同的OS值,如对数秩检验所验证的。在这四个基因中,LDHA在PDAC组织和细胞系中均上调。
    结论:总的来说,我们分析了免疫微环境,预测药物敏感性,并根据m6A调节因子和代谢重新布线之间的串扰评估突变景观的含义,我们还构建了一个基于m6A相关代谢基因的新特征来预测PDAC预后。
    OBJECTIVE: N6-methyladenosine (m6A) is tightly associated with the progression of pancreatic ductal adenocarcinoma (PDAC). Another prominent feature of PDAC is metabolic reprogramming, which provides sufficient nutrients to support rapid cell growth via the tumor microenvironment. However, the underlying influences of m6A-associated metabolic genes on the PDAC microenvironment remain poorly understood. Therefore, we sought to construct a survival prediction model using m6A-related genes to clarify the molecular characteristics of PDAC.
    METHODS: In the present study, m6A-related metabolic genes were obtained from The Cancer Genome Atlas (TCGA) pancreatic adenocarcinoma dataset and subjected to coexpression analysis. Consensus clustering recognized two distinct subgroups with different immune cell infiltration patterns according to the expression of m6A-associated metabolic genes. Multivariate Cox regression analyses and least absolute shrinkage and selection operator (LASSO) analysis were adopted to create an m6A-related metabolism model. A nomogram including clinical features and the risk score based on the expression of m6A-related metabolism regulators was constructed.
    RESULTS: A four-gene signature comprising ATP8B2, GMPS, LDHA and SDR39U1 was built to predict the overall survival (OS) of PDAC patients. This signature also robustly predicted survival in two independent validation cohorts from the International Cancer Genome Consortium (ICGC) and ArrayExpress (E-MTAB-6134). The four-gene signature divided patients into high- and low-risk groups with distinct OS values as verified by the log-rank test. Among the four genes, LDHA was upregulated in both PDAC tissues and cell lines.
    CONCLUSIONS: Collectively, we analyzed the immune microenvironment, predicted drug sensitivity and assessed the implications of the mutation landscape based on the crosstalk between m6A regulators and metabolic rewiring, and we also constructed a novel signature based on m6A-associated metabolic genes to predict PDAC 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
    CircRNAs是近年来研究的热点。它们在各种人类肿瘤中差异表达,并可以通过各种机制调节癌基因和抑癌基因的表达。多样性,稳定性,circRNAs的进化保守性和细胞或组织特异性表达模式也赋予它们在促进或抑制肿瘤细胞恶性生物学行为进展中的重要调节作用。更有趣的是,新兴研究还发现,circRNAs不仅可以调节其他基因的表达,而且它们的亲本基因表达,从而影响肿瘤的发展。除了一些传统的特点,circRNAs在调控亲本基因表达方面具有一定的特异性,影响亲本基因转录的比例更高,更容易翻译成蛋白质来调节亲本基因表达。CircRNAs通常被认为不能产生蛋白质,因此CircRNAs在调节亲本基因表达中表现出的蛋白质编码能力是独特的,并且表明CircRNAs对亲本基因表达的调节作用不仅是竞争性结合关系,而且circRNAs和亲本基因之间更复杂的分子关系,这值得进一步研究。本文综述了circRNAs调控亲本基因表达的分子机制及其在肿瘤发生发展中的生物学作用。旨在为circRNAs在肿瘤靶向治疗中的临床应用提供新的思路。
    CircRNAs have been the focus of research in recent years. They are differentially expressed in various human tumors and can regulate oncogenes and tumor suppressor genes expression through various mechanisms. The diversity, stability, evolutionary conservatism and cell- or tissue-specific expression patterns of circRNAs also endow them with important regulatory roles in promoting or inhibiting tumor cells malignant biological behaviors progression. More interestingly, emerging studies also found that circRNAs can regulate not only other genes expression, but also their parental gene expression and thus influence tumors development. Apart from some conventional features, circRNAs have a certain specificity in the regulation of parental gene expression, with a higher proportion affecting parental gene transcription and easier translation into protein to regulate parental gene expression. CircRNAs are generally thought to be unable to produce proteins and therefore the protein-coding ability exhibited by circRNAs in regulating parental gene expression is unique and indicates that the regulatory effects of parental gene expression by circRNAs are not only a competitive binding relationship, but also a more complex molecular relationship between circRNAs and parental gene, which deserves further study. This review summarizes the molecular mechanisms of circRNAs regulating parental gene expression and their biological roles in tumorigenesis and development, aiming to provide new ideas for the clinical application of circRNAs in tumor-targeted therapy.
    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
    Many cancers engage embryonic genes for rapid growth and evading the immune system. SOX9 has been upregulated in many tumours, yet the role of SOX9 in mediating immunosuppressive tumour microenvironment is unclear. Here, we aim to dissect the role of SOX9-mediated cancer stemness attributes and immunosuppressive microenvironment in advanced gastric adenocarcinoma (GAC) for novel therapeutic discoveries.
    Bulk RNAseq/scRNA-seq, patient-derived cells/models and extensive functional studies were used to identify the expression and functions of SOX9 and its target genes in vitro and in vivo. Immune responses were studied in PBMCs or CD45+ immune cells cocultured with tumour cells with SOX9high or knockout and the KP-Luc2 syngeneic models were used for efficacy of combinations.
    SOX9 is one of the most upregulated SOX genes in GAC and highly expressed in primary and metastatic tissues and associated with poor prognosis. Depletion of SOX9 in patient-derived GAC cells significantly decreased cancer stemness attributes, tumour formation and metastases and consistently increased CD8+ T cell responses when cocultured with PBMCs/CD45+ cells from GAC patients. RNA sequencing identified the leukaemia inhibitory factor (LIF) as the top secreted molecule regulated by SOX9 in tumour cells and was enriched in malignant ascites and mediated SOX9-induced M2 macrophage repolarisation and inhibited T cell function.
    Epithelial SOX9 is critical in suppressing CD8+ T cell responses and modified macrophage function in GAC through the paracrine LIF factor. Cotargeting LIF/LIFR and CSF1R has great potential in targeting SOX9-mediated cancer stemness, T cell immunosuppression and metastases suggesting the novel combination therapy against advanced GAC.
    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:赖氨酰氧化酶样蛋白2(LOXL2)有助于不同肿瘤实体的肿瘤进展和转移,但其在胰腺导管腺癌(PDAC)中的作用尚未在免疫活性体内PDAC模型中进行评估。
    方法:为此,我们使用PDAC患者数据集,患者来源的异种移植物体内和体外模型,和四个条件基因工程小鼠模型(GEMMS)来剖析LOXL2在PDAC中的作用。对于基于GEMM的研究,K-Ras+/LSL-G12D;Trp53LSL-R172H;Pdx1-Cre小鼠(KPC)和K-Ras+/LSL-G12D;Pdx1-Cre小鼠(KC)与Loxl2等位基因Floxed小鼠(Loxl2Exon2fl/fl)或条件性Loxl2过度表达KCL2PCKI小鼠(用于研究总体生存率;肿瘤发病率,负荷和分化;转移;上皮间质转化(EMT);干性和细胞外胶原基质(ECM)组织。
    结果:使用这些PDAC小鼠模型,我们表明,虽然Loxl2消融对原发性肿瘤的发展和生长几乎没有影响,其丢失显著减少了转移并增加了总生存率。我们将这种效应归因于非细胞自主因素,主要是ECM重塑。Loxl2过表达,另一方面,促进原发性和转移性肿瘤生长,降低总生存率,这可能与EMT和干性的增加有关。我们还确定了肿瘤相关的巨噬细胞分泌的制瘤素M(OSM)作为LOXL2表达的诱导剂,并显示体内靶向巨噬细胞会影响Osm和Loxl2表达和胶原纤维排列。
    结论:综合来看,我们的发现建立了PDAC中LOXL2的新病理生理作用和功能,有可能被用来治疗转移性疾病。
    The lysyl oxidase-like protein 2 (LOXL2) contributes to tumour progression and metastasis in different tumour entities, but its role in pancreatic ductal adenocarcinoma (PDAC) has not been evaluated in immunocompetent in vivo PDAC models.
    Towards this end, we used PDAC patient data sets, patient-derived xenograft in vivo and in vitro models, and four conditional genetically-engineered mouse models (GEMMS) to dissect the role of LOXL2 in PDAC. For GEMM-based studies, K-Ras +/LSL-G12D;Trp53 LSL-R172H;Pdx1-Cre mice (KPC) and the K-Ras +/LSL-G12D;Pdx1-Cre mice (KC) were crossed with Loxl2 allele floxed mice (Loxl2Exon2 fl/fl) or conditional Loxl2 overexpressing mice (R26Loxl2 KI/KI) to generate KPCL2KO or KCL2KO and KPCL2KI or KCL2KI mice, which were used to study overall survival; tumour incidence, burden and differentiation; metastases; epithelial to mesenchymal transition (EMT); stemness and extracellular collagen matrix (ECM) organisation.
    Using these PDAC mouse models, we show that while Loxl2 ablation had little effect on primary tumour development and growth, its loss significantly decreased metastasis and increased overall survival. We attribute this effect to non-cell autonomous factors, primarily ECM remodelling. Loxl2 overexpression, on the other hand, promoted primary and metastatic tumour growth and decreased overall survival, which could be linked to increased EMT and stemness. We also identified tumour-associated macrophage-secreted oncostatin M (OSM) as an inducer of LOXL2 expression, and show that targeting macrophages in vivo affects Osm and Loxl2 expression and collagen fibre alignment.
    Taken together, our findings establish novel pathophysiological roles and functions for LOXL2 in PDAC, which could be potentially exploited to treat metastatic disease.
    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
    OBJECTIVE: Pancreatic ductal adenocarcinoma (PDAC) shows a remarkable predilection for liver metastasis. Pro-oncogenic secretome delivery and trafficking via exosomes are crucial for pre-metastatic microenvironment formation and metastasis. This study aimed to explore the underlying mechanisms of how PDAC-derived exosomes (Pex) modulate the liver microenvironment and promote metastasis.
    METHODS: C57BL/6 mice were \'educated\' by tail vein Pex injection. The intrasplenic injection liver metastasis and PDAC orthotopic transplantation models were used to evaluate liver metastasis. Stable cell lines CD44v6 (CD44 variant isoform 6) or C1QBP (complement C1q binding protein) knockdown or overexpression was established using lentivirus transfection or gateway systems. A total of 142 patients with PDAC in Huashan Hospital were retrospectively enrolled. Prognosis and liver metastasis were predicted using Kaplan-Meier survival curves and logistic regression models.
    RESULTS: Pex tail vein injection induced the deposition of liver fibrotic extracellular matrix, which promoted PDAC liver metastasis. Specifically, the exosomal CD44v6/C1QBP complex was delivered to the plasma membrane of hepatic satellite cells (HSCs), leading to phosphorylation of insulin-like growth factor 1 signalling molecules, which resulted in HSC activation and liver fibrosis. Expression of Pex CD44v6 and C1QBP in PDAC patients with liver metastasis was significantly higher than in PDAC patients without liver metastasis, and simultaneous high expression of exosomal CD44v6 and C1QBP correlated with a worse prognosis and a higher risk of postoperative PDAC liver metastasis.
    CONCLUSIONS: The Pex-derived CD44v6/C1QBP complex is essential for the formation of a fibrotic liver microenvironment and PDAC liver metastasis. Highly expressed exosomal CD44v6 and C1QBP are promising biomarkers for predicting prognosis and liver metastasis in patients with 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
    BACKGROUND: High tumor mutation burden (TMB) has gradually become a sensitive biomarker for predicting the response to immunotherapy in many cancers, including lung, bladder and head and neck cancers. However, whether high TMB predicts the response to immunotherapy and prognosis in pancreatic ductal adenocarcinoma (PDAC) remained obscure. Hence, it is significant to investigate the role of genes related to TMB (TRGs) in PDAC.
    METHODS: The transcriptome and mutation data of PDAC was downloaded from The Cancer Genome Atlas-Pancreatic Adenocarcinoma (TCGA). Five independent external datasets of PDAC were chosen to validate parts of our results. qRT-PCR and immunohistochemical staining were also performed to promote the reliability of this study.
    RESULTS: The median overall survival (OS) was significantly increased in TMB_low group compared with the counterpart with higher TMB score after tumor purity adjusted (P = 0.03). 718 differentially expressed TRGs were identified and functionally enriched in some oncogenic pathways. 67 TRGs were associated with OS in PDAC. A prognostic model for the OS was constructed and showed a high predictive accuracy (AUC = 0.849). We also found TMB score was associated with multiple immune components and signatures in tumor microenvironment. In addition, we identified a PDAC subgroup featured with TMBlowMicrosatellite instabilityhigh (MSIhigh) was associated with prolonged OS and a key molecule, ANKRD55, potentially mediating the survival benefits.
    CONCLUSIONS: This study analyzed the biological function, prognosis value, implications for mutation landscape and potential influence on immune microenvironment of TRGs in PDAC, which contributed to get aware of the role of TMB in PDAC. Future studies are expected to investigate how these TRGs regulate the initiation, development or repression of PDAC.
    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