Tumour progression

肿瘤进展
  • 文章类型: Journal Article
    背景:大多数关于肿瘤从前体病变向胆囊腺癌进展的研究都是从不同患者取样的病变,提供致病级联的总体视图。这是否反映了个体患者的致瘤过程仍未充分探索。基因组和表观基因组研究表明,胆囊癌的一部分起源于胆管上皮内瘤变(BilIN)前体病变,而其他人独立于BilIN形成。缺少支持这些结论的空间转录组数据。此外,可以在同一病理样本中检测到多个具有前体或腺癌病变的区域。然而,缺乏有关此类病变的患者内部变异性的知识。
    方法:为了表征个别患者胆囊癌肿瘤发生的空间转录组学,我们选择了两名具有不同病因的癌症患者,其样本同时显示多个正常上皮区域,BilIN和腺癌。使用GeoMx数字空间剖面,我们表征了两个患者中每个样本的大量感兴趣区域(ROI)的整个转录组(分别为24和32个ROI),每个ROI覆盖大约200个正常上皮细胞,低品位Bilin,高级别Bilin或腺癌。使用人胆囊类器官和细胞系衍生的肿瘤来研究基因的肿瘤促进作用。
    结果:空间转录组学显示每种类型的病变都显示出有限的患者内部转录组变异性。我们的数据进一步表明,一名患者的腺癌来自高级别BilIN,另一名患者的低级别BilIN,在后一种情况下,共存的高品位Bilin通过一个独特的过程进化。两名患者在肿瘤进展过程中表现出不同的信号通路激活序列,但两个患者的信号蛋白4A(SEMA4A)表达均受到抑制。使用人类胆囊来源的类器官和细胞系来源的肿瘤,我们提供的证据表明,抑制SEMA4A可促进上皮的假分层,并增强细胞迁移和存活.
    结论:胆囊腺癌可以根据患者的特定过程发展,并且注意到前体病变和癌症病变的患者内部变异性有限。我们的数据表明,抑制SEMA4A可以促进肿瘤进展。他们还强调,除了组织学信息外,还需要获得基因表达数据,以避免低估低度癌前病变的风险。
    BACKGROUND: Most studies on tumour progression from precursor lesion toward gallbladder adenocarcinoma investigate lesions sampled from distinct patients, providing an overarching view of pathogenic cascades. Whether this reflects the tumourigenic process in individual patients remains insufficiently explored. Genomic and epigenomic studies suggest that a subset of gallbladder cancers originate from biliary intraepithelial neoplasia (BilIN) precursor lesions, whereas others form independently from BilINs. Spatial transcriptomic data supporting these conclusions are missing. Moreover, multiple areas with precursor or adenocarcinoma lesions can be detected within the same pathological sample. Yet, knowledge about intra-patient variability of such lesions is lacking.
    METHODS: To characterise the spatial transcriptomics of gallbladder cancer tumourigenesis in individual patients, we selected two patients with distinct cancer aetiology and whose samples simultaneously displayed multiple areas of normal epithelium, BilINs and adenocarcinoma. Using GeoMx digital spatial profiling, we characterised the whole transcriptome of a high number of regions of interest (ROIs) per sample in the two patients (24 and 32 ROIs respectively), with each ROI covering approximately 200 cells of normal epithelium, low-grade BilIN, high-grade BilIN or adenocarcinoma. Human gallbladder organoids and cell line-derived tumours were used to investigate the tumour-promoting role of genes.
    RESULTS: Spatial transcriptomics revealed that each type of lesion displayed limited intra-patient transcriptomic variability. Our data further suggest that adenocarcinoma derived from high-grade BilIN in one patient and from low-grade BilIN in the other patient, with co-existing high-grade BilIN evolving via a distinct process in the latter case. The two patients displayed distinct sequences of signalling pathway activation during tumour progression, but Semaphorin 4 A (SEMA4A) expression was repressed in both patients. Using human gallbladder-derived organoids and cell line-derived tumours, we provide evidence that repression of SEMA4A promotes pseudostratification of the epithelium and enhances cell migration and survival.
    CONCLUSIONS: Gallbladder adenocarcinoma can develop according to patient-specific processes, and limited intra-patient variability of precursor and cancer lesions was noticed. Our data suggest that repression of SEMA4A can promote tumour progression. They also highlight the need to gain gene expression data in addition to histological information to avoid understimating the risk of low-grade preneoplastic lesions.
    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:一系列研究表明转移RNA(tRNA)加工在肿瘤进展过程中的出现。然而,tRNA加工基因在神经母细胞瘤(NB)中的作用和调控机制,儿童脑外常见的恶性肿瘤,还未知。
    方法:对多组学结果进行分析,以确定下游tRNA加工基因的关键调节因子。使用免疫共沉淀和质谱方法来测量蛋白质之间的相互作用。通过双荧光素酶报告基因检测转录调节因子对下游基因表达的影响,染色质免疫沉淀,蛋白质印迹和实时定量逆转录聚合酶链反应(RT-PCR)方法。已经进行了研究以揭示转录调节因子对NB生物学过程的影响和机制。使用对数秩检验分析生存差异。
    结果:c-Myc被鉴定为在NB细胞中驱动tRNA加工基因表达和随后的苹果酸-天冬氨酸穿梭(MAS)的转录因子。机械上,c-Myc直接促进谷氨酰-氨酰-tRNA合成酶(EPRS)和亮氨酰-tRNA合成酶(LARS)的表达,通过抑制一般控制未抑制的2或激活雷帕霉素信号的机制靶标,导致谷氨酸-草酰乙酸转氨酶1(GOT1)和苹果酸脱氢酶1(MDH1)的翻译上调。同时,层粘连蛋白A(LMNA)通过物理相互作用抑制c-Myc反式激活,导致对MAS的压制,有氧糖酵解,肿瘤发生和侵略性。临床前,洛贝林被发现是一种LMNA结合化合物,以促进其与c-Myc的相互作用,抑制氨酰tRNA合成酶的表达,MAS与NB的肿瘤进展,以及来自c-Myc敲入小鼠的类器官的生长。低水平的LMNA或c-Myc表达升高,EPRS,LARS,GOT1或MDH1与临床NB患者的预后较差和生存时间较短有关。
    结论:这些结果表明,LMNA靶向c-Myc转录激活抑制了MAS和肿瘤进展所必需的tRNA加工。
    BACKGROUND: A series of studies have demonstrated the emerging involvement of transfer RNA (tRNA) processing during the progression of tumours. Nevertheless, the roles and regulating mechanisms of tRNA processing genes in neuroblastoma (NB), the prevalent malignant tumour outside the brain in children, are yet unknown.
    METHODS: Analysis of multi-omics results was conducted to identify crucial regulators of downstream tRNA processing genes. Co-immunoprecipitation and mass spectrometry methods were utilised to measure interaction between proteins. The impact of transcriptional regulators on expression of downstream genes was measured by dual-luciferase reporter, chromatin immunoprecipitation, western blotting and real-time quantitative reverse transcription-polymerase chain reaction (RT-PCR) methods. Studies have been conducted to reveal impact and mechanisms of transcriptional regulators on biological processes of NB. Survival differences were analysed using the log-rank test.
    RESULTS: c-Myc was identified as a transcription factor driving tRNA processing gene expression and subsequent malate-aspartate shuttle (MAS) in NB cells. Mechanistically, c-Myc directly promoted the expression of glutamyl-prolyl-tRNA synthetase (EPRS) and leucyl-tRNA synthetase (LARS), resulting in translational up-regulation of glutamic-oxaloacetic transaminase 1 (GOT1) as well as malate dehydrogenase 1 (MDH1) via inhibiting general control nonrepressed 2 or activating mechanistic target of rapamycin signalling. Meanwhile, lamin A (LMNA) inhibited c-Myc transactivation via physical interaction, leading to suppression of MAS, aerobic glycolysis, tumourigenesis and aggressiveness. Pre-clinically, lobeline was discovered as a LMNA-binding compound to facilitate its interaction with c-Myc, which inhibited aminoacyl-tRNA synthetase expression, MAS and tumour progression of NB, as well as growth of organoid derived from c-Myc knock-in mice. Low levels of LMNA or elevated expression of c-Myc, EPRS, LARS, GOT1 or MDH1 were linked to a worse outcome and a shorter survival time of clinical NB patients.
    CONCLUSIONS: These results suggest that targeting c-Myc transactivation by LMNA inhibits tRNA processing essential for MAS and tumour 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
    血管生成素(ANG)是一种特殊的分泌型核糖核酸酶,也称为RNase5,在脊椎动物中广泛表达。ANG失调与乳房发育密切相关,鼻咽,和肺癌。近年来,研究发现,ANG不仅通过激活内皮细胞诱导新生血管形成,而且在癌细胞的可塑性中也起着调节作用。细胞可塑性在癌症发生中起关键作用,programming,迁移,治疗抗性,和复发。因此,它是癌症诊断的一种有前途的生物标志物,预后评估,和治疗。这篇综述总结了关于ANG在癌症发生发展中的作用和临床应用的最新知识。
    Angiogenin (ANG) is a specialised secreted ribonuclease, also known as RNase5, that is widely expressed in vertebrates. ANG dysregulation is closely associated with the development of breast, nasopharyngeal, and lung cancers. In recent years, studies have found that ANG not only induces neovascularisation by activating endothelial cells, but also plays a regulatory role in the plasticity of cancer cells. Cellular plasticity plays pivotal roles in cancer initiation, progression, migration, therapeutic resistance, and relapse. Therefore, it is a promising biomarker for cancer diagnosis, prognostic evaluation, and therapy. This review summarises the current knowledge regarding the roles and clinical applications of ANG in cancer development and 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
    类固醇受体共激活因子-1(SRC-1,也称为NCOA1)通常通过直接与转录因子结合并募集到靶基因启动子以通过增加染色质可及性和促进转录复合物的形成来促进基因转录而发挥转录共激活因子的作用。近几十年来,已经报道了SRC-1的各种生物学和病理学功能,特别是在肿瘤发生的背景下。SRC-1是多种癌症进展的促进者,包括乳腺癌,前列腺癌,胃肠道癌,神经癌症,女性生殖系统癌症.SRC-1的新出现的多器官致癌作用仍在研究中,可能不仅限于产生类固醇激素的组织。越来越多的证据表明,SRC-1通过直接结合转录因子促进靶基因表达,这可能构成一种独立于AR或ER的新型共激活模式。此外,已经报道了用包括各种小分子或天然活性化合物的药物抑制SRC-1的抗肿瘤作用,但是它们在临床癌症治疗中的实际应用非常有限。对于这篇评论,我们收集了SRC-1致癌作用的典型证据,强调了它的主要合作者和调控基因,并绘制了SRC-1促进原发性肿瘤进展的潜在机制。
    Steroid receptor coactivator-1 (SRC-1, also known as NCOA1) frequently functions as a transcriptional coactivator by directly binding to transcription factors and recruiting to the target gene promoters to promote gene transcription by increasing chromatin accessibility and promoting the formation of transcriptional complexes. In recent decades, various biological and pathological functions of SRC-1 have been reported, especially in the context of tumorigenesis. SRC-1 is a facilitator of the progression of multiple cancers, including breast cancer, prostate cancer, gastrointestinal cancer, neurological cancer, and female genital system cancer. The emerging multiorgan oncogenic role of SRC-1 is still being studied and may not be limited to only steroid hormone-producing tissues. Growing evidence suggests that SRC-1 promotes target gene expression by directly binding to transcription factors, which may constitute a novel coactivation pattern independent of AR or ER. In addition, the antitumour effect of pharmacological inhibition of SRC-1 with agents including various small molecules or naturally active compounds has been reported, but their practical application in clinical cancer therapy is very limited. For this review, we gathered typical evidence on the oncogenic role of SRC-1, highlighted its major collaborators and regulatory genes, and mapped the potential mechanisms by which SRC-1 promotes primary tumour 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: Preprint
    实体肿瘤通常在进展过程中忍受营养不足。肿瘤细胞如何适应时间和空间营养不足尚不清楚。我们先前通过转录组筛选将STC2鉴定为暴露于营养不足的细胞中最上调的基因之一。表明STC2在细胞适应营养不足方面的潜力。然而,营养不足和随后的适应引起STC2诱导的分子机制仍然难以捉摸。这里,我们报道STC2蛋白显著增加并通过Gln-/Glc-剥夺分泌到培养基中。STC2启动子含有ATF4和p65/RelA激活的顺式元件,两种转录因子被多种细胞应激激活。生物学,STC2诱导和分泌促进细胞存活,但在营养不足时减弱细胞增殖,从而将癌细胞的优先级从增殖转变为存活。STC2的缺失通过诱导小鼠异种移植物中的细胞凋亡和坏死而损害肿瘤生长。机械上,在营养不足的条件下,细胞的活性氧(ROS)水平升高,STC2的缺乏进一步升高了导致细胞凋亡增加的ROS水平。RNA-Seq分析显示STC2诱导抑制单胺氧化酶B(MAOB)的表达,产生ROS的线粒体膜酶。此外,在人肿瘤样品中还鉴定出STC2和MAOB水平之间的负相关。重要的是,重组STC2对培养基的给药有效地抑制MAOB表达以及细胞凋亡,提示STC2以自分泌/旁分泌方式起作用。一起来看,我们的发现表明营养不足诱导STC2表达,反过来通过维持氧化还原稳态来控制癌细胞对营养不足的适应,强调STC2作为癌症治疗的治疗靶标的潜力。
    Solid tumours often endure nutrient insufficiency during progression. How tumour cells adapt to temporal and spatial nutrient insufficiency remains unclear. We previously identified STC2 as one of the most upregulated genes in cells exposed to nutrient insufficiency by transcriptome screening, indicating the potential of STC2 in cellular adaptation to nutrient insufficiency. However, the molecular mechanisms underlying STC2 induction by nutrient insufficiency and subsequent adaptation remain elusive. Here, we report that STC2 protein is dramatically increased and secreted into the culture media by Gln-/Glc-deprivation. STC2 promoter contains cis-elements that are activated by ATF4 and p65/RelA, two transcription factors activated by a variety of cellular stress. Biologically, STC2 induction and secretion promote cell survival but attenuate cell proliferation during nutrient insufficiency, thus switching the priority of cancer cells from proliferation to survival. Loss of STC2 impairs tumour growth by inducing both apoptosis and necrosis in mouse xenografts. Mechanistically, under nutrient insufficient conditions, cells have increased levels of reactive oxygen species (ROS), and lack of STC2 further elevates ROS levels that lead to increased apoptosis. RNA-Seq analyses reveal STC2 induction suppresses the expression of monoamine oxidase B (MAOB), a mitochondrial membrane enzyme that produces ROS. Moreover, a negative correlation between STC2 and MAOB levels is also identified in human tumour samples. Importantly, the administration of recombinant STC2 to the culture media effectively suppresses MAOB expression as well as apoptosis, suggesting STC2 functions in an autocrine/paracrine manner. Taken together, our findings indicate that nutrient insufficiency induces STC2 expression, which in turn governs the adaptation of cancer cells to nutrient insufficiency through the maintenance of redox homeostasis, highlighting the potential of STC2 as a therapeutic target for 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
    背景:细胞外囊泡(EV)通过细胞与细胞的通讯参与癌症的发展。长链非编码RNA(lncRNA),电动汽车的一个组成部分,可以通过EV介导的递送在非小细胞肺癌(NSCLC)中发挥重要作用。
    方法:通过lncRNA微阵列分析鉴定了EV中NSCLC相关的lncRNAAL139294.1。在体外和体内检查了AL139294.1在NSCLC中的作用。使用共聚焦显微镜观察AL139294.1在EV中的包封及其向受体细胞的转运。使用共培养装置来检查运输的AL139294.1对受体细胞的致癌行为的影响。进行双荧光素酶报告基因测定以验证miR-204-5p与AL139294.1和含溴结构域蛋白4(BRD4)的直接相互作用。使用定量聚合酶链反应对电动汽车中的AL139294.1和miR-204-5p进行定量。进行了接收器工作特性分析以评估诊断效率。
    结果:EV中的lncRNAAL139294.1在体外和体内促进NSCLC进展。将AL139294.1封装到EV中并转运到受体细胞后,它促进了细胞的增殖,迁移,和侵袭能力通过与miR-204-5p竞争性结合来调节BRD4,导致Wnt和NF-κB2途径的激活。此外,血清lncRNAAL139294.1在电动汽车中的表达增加,而miR-204-5p在EV中在NSCLC中降低。EV中高水平的lncRNAAL139294.1和低水平的miR-204-5p与晚期病理分期相关,淋巴结转移,和远处转移,强调了它们在区分疾病的严重表现方面的有希望的效用。
    结论:这项研究揭示了与NSCLC相关的EV中的一种新型lncRNA,即,AL139294.1,为NSCLC的发展提供了有价值的见解,并为NSCLC引入了潜在的诊断生物标志物。
    BACKGROUND: Extracellular vesicles (EVs) participate in cancer development via cell-to-cell communication. Long non-coding RNAs (lncRNAs), one component of EVs, can play an essential role in non-small-cell lung cancer (NSCLC) through EV-mediated delivery.
    METHODS: The NSCLC-associated lncRNA AL139294.1 in EVs was identified via lncRNA microarray analysis. The role of AL139294.1 in NSCLC was examined in vitro and in vivo. Confocal microscopy was used to observe the encapsulation of AL139294.1 into EVs and its transport to recipient cells. A co-culture device was used to examine the effects of transported AL139294.1 on the oncogenic behaviour of recipient cells. Dual-luciferase reporter assay was performed to verify the direct interaction of miR-204-5p with AL139294.1 and bromodomain-containing protein 4 (BRD4). AL139294.1 and miR-204-5p in EVs were quantified using quantitative polymerase chain reaction. Receiver operating characteristic analyses were conducted to evaluate the diagnostic efficiency.
    RESULTS: The lncRNA AL139294.1 in EVs promoted NSCLC progression in vitro and in vivo. After AL139294.1 was encapsulated into EVs and transported to recipient cells, it promoted the cells\' proliferation, migration, and invasion abilities by competitively binding with miR-204-5p to regulate BRD4, leading to the activation of the Wnt and NF-κB2 pathways. Additionally, the expression of serum lncRNA AL139294.1 in EVs was increased, whereas miR-204-5p in EVs was decreased in NSCLC. High levels of lncRNA AL139294.1 and low levels of miR-204-5p in EVs were associated with advanced pathological staging, lymph node metastasis, and distant metastasis, underscoring their promising utility for distinguishing between more and less severe manifestations of the disease.
    CONCLUSIONS: This study reveals a novel lncRNA in EVs associated with NSCLC, namely, AL139294.1, providing valuable insights into the development of NSCLC and introducing potential diagnostic biomarkers for NSCLC.
    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:Tousled样激酶1(TLK1)在胃癌(GC)中的作用尚不清楚。
    目的:为了研究表达,生物学功能,以及TLK1在GC中的潜在机制。
    方法:我们通过westernblot和免疫荧光检测了TLK1蛋白在GC细胞和组织中的表达水平和定位TLK1,分别。我们用慢病毒转染各种GC细胞,以创建TLK1过表达和敲低系,并通过体外集落形成建立TLK1的功能作用,5-乙炔基-2'-脱氧尿苷,和Transwell分析以及流式细胞术。我们应用生物信息学来阐明与TLK1相关的信号通路。我们通过在裸鼠中诱导皮下异种移植肿瘤来进行TLK1功能的体内验证。
    结果:TLK1在GC细胞和组织中与正常细胞和组织相比显著上调,并且主要定位于细胞核。TLK1敲低显著降低集落形成,扩散,入侵,GC细胞的迁移和凋亡增加。TLK1过表达具有相反的作用。生物信息学揭示,随后的实验验证了,肿瘤生长因子-β信号通路与TLK1介导的GC进展有关。体内测定证实TLK1促进GC中的肿瘤发生。
    结论:本研究的结果表明,TLK1在GC进展中起着至关重要的作用,因此,有望作为这种疾病的治疗靶标。
    BACKGROUND: The role of Tousled-like kinase 1 (TLK1) in in gastric cancer (GC) remains unclear.
    OBJECTIVE: To investigate the expression, biological function, and underlying mechanisms of TLK1 in GC.
    METHODS: We measured TLK1 protein expression levels and localized TLK1 in GC cells and tissues by western blot and immunofluorescence, respectively. We transfected various GC cells with lentiviruses to create TLK1 overexpression and knockdown lines and established the functional roles of TLK1 through in vitro colony formation, 5-ethynyl-2`-deoxyuridine, and Transwell assays as well as flow cytometry. We applied bioinformatics to elucidate the signaling pathways associated with TLK1. We performed in vivo validation of TLK1 functions by inducing subcutaneous xenograft tumors in nude mice.
    RESULTS: TLK1 was significantly upregulated in GC cells and tissues compared to their normal counterparts and was localized mainly to the nucleus. TLK1 knockdown significantly decreased colony formation, proliferation, invasion, and migration but increased apoptosis in GC cells. TLK1 overexpression had the opposite effects. Bioinformatics revealed, and subsequent experiments verified, that the tumor growth factor-beta signaling pathway was implicated in TLK1-mediated GC progression. The in vivo assays confirmed that TLK1 promotes tumorigenesis in GC.
    CONCLUSIONS: The findings of the present study indicated that TLK1 plays a crucial role in GC progression and is, therefore, promising as a therapeutic target against this 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: Meta-Analysis
    作为最突出的RNA修饰,N6-甲基腺苷(m6A)参与肿瘤发生和发展的调节。环状RNA(circularRNAs)在无处不在的生命过程中也起着至关重要的作用。肾细胞癌(RCC)中m6A调节是否需要circRNAs仍不清楚。Meta分析和生物信息学分析发现IGF2BP3在RCC中表达上调,预后较差。IGF2BP3在体外和体内均能显著促进RCC的进展。机械上,circRARS与IGF2BP3的KH1-KH2结构域结合以增强m6A修饰识别。一个12-nt序列(GUCUUCCAGCAA)被证明是circRARS的IGF2BP3结合位点。此外,CAPN15,CD44,HMGA2,TNRC6A和ZMIZ2被筛选为由IGF2BP3/circRARS复合物以m6A依赖性方式调节的靶基因。稳定剂蛋白质,包括HuR,Matrin3和pAbPC1被circRARS招募,从而提高上述5个靶基因的mRNA稳定性。因此,IGF2BP3/circRARS复合物促进RCC细胞的脂质积累,并通过靶基因促进舒尼替尼耐药.circRARS与IGF2BP3协同促进m6A识别,从而促进RCC进展。因此,IGF2BP3可能是RCC诊断和预后的潜在生物标志物和治疗靶标。
    As the most prominent RNA modification, N6-methyladenosine (m6 A) participates in the regulation of tumour initiation and progression. Circular RNAs (circRNAs) also play crucial roles in ubiquitous life processes. Whether circRNAs are required for m6 A regulation in renal cell carcinoma (RCC) remains unclear. Meta-analysis and bioinformatics identified that IGF2BP3 was upregulated in RCC and indicated a worse prognosis. IGF2BP3 significantly promoted RCC progression in vitro and in vivo. Mechanistically, circRARS bound to KH1-KH2 domains of IGF2BP3 to enhance m6 A modification recognition. A 12-nt sequence (GUCUUCCAGCAA) was proven to be the IGF2BP3-binding site of circRARS. Additionally, CAPN15, CD44, HMGA2, TNRC6A and ZMIZ2 were screened to be the target genes regulated by the IGF2BP3/circRARS complex in an m6 A-dependent manner. Stabiliser proteins, including HuR, Matrin3 and pAbPC1, were recruited by circRARS, thereby increasing the mRNA stability of the forementioned five target genes. Consequently, the IGF2BP3/circRARS complex facilitated the lipid accumulation of RCC cells and promoted sunitinib resistance via target genes. circRARS synergised with IGF2BP3 to facilitate m6 A recognition, thereby promoting RCC progression. Thus, IGF2BP3 could be a potential biomarker for RCC diagnosis and prognosis and a therapeutic target.
    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
    食管鳞状细胞癌(ESCC)是侵袭性最强的恶性肿瘤之一,具有很高的发病率和死亡率。虽然手术,放疗和化疗是食管癌的常见治疗选择,治疗后5年生存率仍然很低。一方面,许多食道癌是在晚期发现的,另一方面,治疗抵抗是治疗局部晚期ESCC的主要障碍。癌症相关成纤维细胞(CAFs),肿瘤微环境中基质细胞的主要类型,增强肿瘤进展和治疗耐药性,并已成为食道癌靶向治疗研究的主要焦点。为了提供潜力,改善治疗效果的前瞻性目标,这篇综述总结了CAFs的起源和激活及其在ESCC中调节肿瘤进展和治疗抵抗中的具体作用。我们还强调了ESCCCAF靶向治疗的临床潜力和新兴趋势。
    Oesophageal squamous cell carcinoma (ESCC) is one of the most aggressive malignant tumours with high morbidity and mortality. Although surgery, radiotherapy and chemotherapy are common treatment options available for oesophageal cancer, the 5-year survival rate remains low after treatment. On the one hand, many oesophageal cancers are are discovered at an advanced stage and, on the other hand, treatment resistance is a major obstacle to treating locally advanced ESCC. Cancer-associated fibroblasts (CAFs), the main type of stromal cell in the tumour microenvironment, enhance tumour progression and treatment resistance and have emerged as a major focus of study on targeted therapy of oesophageal cancer.With the aim of providing potential, prospective targets for improving therapeutic efficacy, this review summarises the origin and activation of CAFs and their specific role in regulating tumour progression and treatment resistance in ESCC. We also emphasize the clinical potential and emerging trends of ESCC CAFs-targeted treatments.
    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
    赖氨酰氧化酶样2(LOXL2)属于赖氨酰氧化酶(LOX)家族,它包含人类中的五种赖氨酸酪氨酸醌(LTQ)依赖性铜胺氧化酶。2003年,LOXL2首次被确定为肿瘤进展的启动子,在二十年的过程中,许多研究已经确定其参与多种癌症。对大量人类肿瘤样品的广泛研究表明,L0XL2表达失调与患者的不良预后密切相关。此外,研究表明,LOXL2与影响肿瘤进展各个方面的各种靶标之间存在关联.此外,发现一个复杂的信号因子网络在转录中起作用,转录后,翻译后水平为L0XL2在肿瘤中异常表达的潜在机制提供了见解。此外,L0XL2沉默或过表达的基因修饰小鼠模型的开发使得能够深入探索其在各种癌症模型中的体内作用.鉴于LOXL2在许多癌症中的重要作用,目前正在进行大量努力,以确定可能改善患者预后的特异性抑制剂.在这次审查中,我们旨在对LOXL2在癌症中作用的20年研究进行全面概述.
    Lysyl Oxidase Like 2 (LOXL2) belongs to the lysyl oxidase (LOX) family, which comprises five lysine tyrosylquinone (LTQ)-dependent copper amine oxidases in humans. In 2003, LOXL2 was first identified as a promoter of tumour progression and, over the course of two decades, numerous studies have firmly established its involvement in multiple cancers. Extensive research with large cohorts of human tumour samples has demonstrated that dysregulated LOXL2 expression is strongly associated with poor prognosis in patients. Moreover, investigations have revealed the association of LOXL2 with various targets affecting diverse aspects of tumour progression. Additionally, the discovery of a complex network of signalling factors acting at the transcriptional, post-transcriptional, and post-translational levels has provided insights into the mechanisms underlying the aberrant expression of LOXL2 in tumours. Furthermore, the development of genetically modified mouse models with silenced or overexpressed LOXL2 has enabled in-depth exploration of its in vivo role in various cancer models. Given the significant role of LOXL2 in numerous cancers, extensive efforts are underway to identify specific inhibitors that could potentially improve patient prognosis. In this review, we aim to provide a comprehensive overview of two decades of research on the role of LOXL2 in cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号