RAS/MAPK

RAS / MAPK
  • 文章类型: Journal Article
    肺腺癌(LUAC)是最常见的肺癌,其发病率正在增加。补体因子B(CFB)是替代补体途径中的重要因子。据报道,CFB参与了许多癌症的进展,包括胰腺癌,皮肤鳞状细胞癌,还有鼻咽癌,但CFB在LUAC中的作用和分子机制尚不清楚。本研究旨在探讨CFB在LUAC恶性进展中的作用。在我们之前的研究中,我们发现CFB在LUAC临床样本中表达下调.这里,我们首先在体外检测细胞功能。细胞增殖和迁移增加,CFB敲低后细胞凋亡和细胞周期阻滞被抑制。CFB的过表达在体外抑制了LUAC的恶性进展。此外,体内实验表明,CFB的上调抑制了肿瘤的生长和Ki67的表达。此外,我们的数据表明,CFB负调节Ras/丝裂原活化蛋白激酶(MAPK)信号通路。此外,CFB的上调抑制LUAC的进展被Ras/MAPK途径激活剂(ML-098或C16-PAF)逆转。我们的研究发现CFB作为肿瘤抑制因子通过抑制Ras/MAPK通路抑制LUAC的肿瘤发生,提示CFB可能是LUAC的潜在生物标志物和治疗靶点。
    Lung adenocarcinoma (LUAC) as the most common lung cancer, and its incidence is increasing. Complement factor B (CFB) is an important factor in the alternative complement pathway. CFB has been reported to be involved in the progression of many cancers, including in pancreatic cancer, cutaneous squamous cell carcinoma, and nasopharyngeal carcinoma, but the function and molecular mechanism of CFB in LUAC remains unclear. The present study aimed to explore the role of CFB in LUAC malignant progression. In our previous study, we found that CFB was downregulated expression in LUAC clinical samples. Here, we firstly detected the cell function in vitro. Cell proliferation and migration were increased, while cell apoptosis and cell cycle arrest were suppressed after CFB knockdown. Overexpression of CFB repressed the malignant progression of LUAC in vitro. Besides, in vivo experiments revealed that upregulation of CFB inhibited tumor growth and Ki67 expression. Additionally, our data indicated that CFB negatively regulated Ras/mitogen-activated protein kinase (MAPK) signaling pathway. Furthermore, upregulation of CFB inhibited the progression of LUAC was reversed by Ras/MAPK pathway activators (ML-098 or C16-PAF). Our study uncovered that CFB acts as a tumor suppressor repressed tumorigenesis of LUAC through inhibiting the Ras/MAPK pathway, suggesting that CFB may be a potential biomarker and therapeutic target for LUAC.
    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
    室间隔缺损(VSD)被认为是最常见的先天性心脏病(CHD)之一。占所有心脏畸形的40%,并在个别患者和家庭中以孤立的CHD以及其他心脏和心外先天性畸形发生。VSD的遗传病因复杂且异常异质性。据报道,染色体异常,例如非整倍性和结构变异以及各种基因中的罕见点突变与这种心脏缺陷有关。这包括具有已知遗传原因的明确定义的综合征(例如,DiGeorge综合征和Holt-Oram综合征)以及迄今为止尚未定义的以非特异性症状为特征的综合征形式。编码心脏转录因子的基因突变(例如,NKX2-5和GATA4)和信号分子(例如,CFC1)在VSD病例中最常见。此外,新的高分辨率方法,如比较基因组杂交,能够发现大量不同的拷贝数变异,导致通常包含多个基因的染色体区域的增加或丢失,VSD患者。在这一章中,我们将描述在VSD患者中观察到的广泛遗传异质性,并考虑该领域的最新进展.
    Ventricular septal defects (VSDs) are recognized as one of the commonest congenital heart diseases (CHD), accounting for up to 40% of all cardiac malformations, and occur as isolated CHDs as well as together with other cardiac and extracardiac congenital malformations in individual patients and families. The genetic etiology of VSD is complex and extraordinarily heterogeneous. Chromosomal abnormalities such as aneuploidy and structural variations as well as rare point mutations in various genes have been reported to be associated with this cardiac defect. This includes both well-defined syndromes with known genetic cause (e.g., DiGeorge syndrome and Holt-Oram syndrome) and so far undefined syndromic forms characterized by unspecific symptoms. Mutations in genes encoding cardiac transcription factors (e.g., NKX2-5 and GATA4) and signaling molecules (e.g., CFC1) have been most frequently found in VSD cases. Moreover, new high-resolution methods such as comparative genomic hybridization enabled the discovery of a high number of different copy number variations, leading to gain or loss of chromosomal regions often containing multiple genes, in patients with VSD. In this chapter, we will describe the broad genetic heterogeneity observed in VSD patients considering recent advances in this field.
    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: Case Reports
    Noonan综合征是一种所谓的“放射病”,“这是以身材矮小为特征的,独特的面部特征,先天性心脏缺陷,和发育迟缓。在临床诊断为努南综合征的个体中,80%-90%在与该疾病有关的已知基因中有致病变异,但其余病例的分子机制尚不清楚。ETS2抑制因子(ERF)的杂合致病变体,作为RAS/MAPK信号通路的阻遏物,引起综合征性颅骨融合。这里,我们报道了一个ERF移码变异体,其与Noonan综合征样表型在一个家族中共分离.先证者是一名3岁女性,面部特征畸形,包括突起,超端粒,稍微向下倾斜的睑裂,低设置后旋转的耳朵,鼻梁凹陷,身材矮小,和发育迟缓。先证者的外显子组测序鉴定了杂合ERF变体[NM_006494.4:c.185delp.(Glu62Glyfs*15)]。她的母亲和姐姐表现出相似的表型,并且具有相同的杂合ERF变体。先前报道的大部分患有综合征性颅骨融合和致病性ERF变异的患者也显示出与Noonan综合征重叠的特征性特征。本发现支持杂合ERF变体与Noonan综合征样表型之间的关联。
    Noonan syndrome is a so-called \"RASopathy,\" that is characterized by short stature, distinctive facial features, congenital heart defects, and developmental delay. Of individuals with a clinical diagnosis of Noonan syndrome, 80%-90% have pathogenic variants in the known genes implicated in the disorder, but the molecular mechanism is unknown in the remaining cases. Heterozygous pathogenic variants of ETS2 repressor factor (ERF), which functions as a repressor in the RAS/MAPK signaling pathway, cause syndromic craniosynostosis. Here, we report an ERF frameshift variant cosegregating with a Noonan syndrome-like phenotype in a family. The proband was a 3-year-old female who presented with dysmorphic facial features, including proptosis, hypertelorism, slightly down slanted palpebral fissures, low-set posteriorly rotated ears, depressed nasal bridge, short stature, and developmental delay. Exome sequencing of the proband identified a heterozygous ERF variant [NM_006494.4: c.185del p.(Glu62Glyfs*15)]. Her mother and sister showed a similar phenotype and had the same heterozygous ERF variant. A large proportion of the previously reported patients with syndromic craniosynostosis and pathogenic ERF variants also showed characteristic features that overlap with those of Noonan syndrome. The present finding supports an association between heterozygous ERF variants and a Noonan syndrome-like phenotype.
    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
    人类催乳素及其受体(PRLr)与乳腺癌的发病机制密切相关。人PRLr(hPRLrI)的中间形式通过可变剪接产生,并且具有新的13个氨基酸的尾部(\"I-tail\")增益。当与长形式hPRLr(hPRLrL)共表达时,hPRLrI在体外诱导正常乳腺上皮的显着增殖和不依赖锚定的生长。hPRLrL和hPRLrI共表达是体内诱导乳腺上皮转化所必需的。I-tail与泛素样蛋白神经前体细胞表达的发育下调蛋白8相关。用神经前体细胞表达的发育下调的蛋白8激活酶抑制剂pevonedistat处理导致hPRLrL增加和乳腺癌细胞死亡。这项研究的目的是确定hPRLrII-tail在hPRLrL/hPRLrI介导的乳腺转化中的功能。将hPRLrL/hPRLrI和hPRLrL/hPRLrIΔ13(I-尾部去除突变体)递送至MCF10AT细胞。当去除hPRLrI-tail时,细胞增殖降低。I尾缺失降低了锚定非依赖性生长并减弱了细胞迁移。I-tail参与Ras/MAPK信号传导,但不参与PI3K/Akt信号传导途径,如通过蛋白质印迹所示。I-tail去除导致hPRLrI稳定性降低。RNA测序数据显示,I尾去除导致催乳素诱导的差异基因表达。独创性途径分析表明ERK的活性减弱。用ERK1/2抑制剂ulixertinib治疗乳腺癌细胞导致集落形成能力降低和增殖减少。这些研究表明,hPRLrII-tail有助于乳腺癌的发生,可能是开发新乳腺癌疗法的有希望的靶标。
    Prolactin and its receptor (PRLr) in humans are significantly involved in breast cancer pathogenesis. The intermediate form of human PRLr (hPRLrI) is produced by alternative splicing and has a novel 13 amino acid tail (\"I-tail\") gain. hPRLrI induces significant proliferation and anchorage-independent growth of normal mammary epithelia in vitro when coexpressed with the long form hPRLr (hPRLrL). hPRLrL and hPRLrI coexpression is necessary to induce the transformation of mammary epithelia in vivo. The I-tail is associated with the ubiquitin-like protein neural precursor cell expressed developmentally downregulated protein 8. Treatment with the neural precursor cell expressed developmentally downregulated protein 8-activating enzyme inhibitor pevonedistat resulted in increased hPRLrL and the death of breast cancer cells. The goal of this study was to determine the function of the hPRLrI I-tail in hPRLrL/hPRLrI-mediated mammary transformation. hPRLrL/hPRLrI and hPRLrL/hPRLrIΔ13 (I-tail removal mutant) were delivered to MCF10AT cells. Cell proliferation was decreased when hPRLrI I-tail was removed. I-tail deletion decreased anchorage-independent growth and attenuated cell migration. The I-tail was involved in Ras/MAPK signaling but not PI3K/Akt signaling pathway as shown by western blot. I-tail removal resulted in decreased hPRLrI stability. RNA-sequencing data revealed that I-tail removal resulted in differential gene expression induced by prolactin. Ingenuity Pathway Analysis revealed that the activity of ERK was attenuated. Treatment of breast cancer cells with ERK1/2 inhibitor ulixertinib resulted in decreased colony-forming ability and less proliferation. These studies suggest that the hPRLrI I-tail contributed to breast oncogenesis and may be a promising target for the development of new breast cancer therapies.
    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
    从昆虫到人类,卵子发生与营养输入密切相关,然而,关于整个生物体生理学如何将饮食变化与卵母细胞发育相匹配,人们知之甚少。考虑到饮食引起的脂肪组织功能障碍与生育问题的风险增加有关,和其他肥胖相关的病理生理,破译将脂肪营养感知与卵巢和其他组织的远程控制联系起来的细胞和分子机制至关重要。我们以前对果蝇的研究表明,氨基酸感应,通过氨基酸反应途径和mTOR介导的信号传导功能在脂肪细胞内控制生殖干细胞维持和排卵,分别。此外,我们证明了脂肪细胞内的胰岛素/胰岛素样生长因子信号采用不同的效应轴,PI3K/Akt1依赖和独立,胰岛素受体活性的下游介导脂肪与卵巢的交流。这里,我们报道,Ras/MAPK信号轴在脂肪细胞中发挥功能,调节成熟卵母细胞的早期种系囊肿存活和排卵,但对于种系干细胞维持或通过卵黄发生的进展并不重要.因此,这些研究揭示了介导器官间通讯的信号通路活动的复杂性.
    From insects to humans, oogenesis is tightly linked to nutritional input, yet little is known about how whole organism physiology matches dietary changes with oocyte development. Considering that diet-induced adipose tissue dysfunction is associated with an increased risk for fertility problems, and other obesity-associated pathophysiologies, it is critical to decipher the cellular and molecular mechanisms linking adipose nutrient sensing to remote control of the ovary and other tissues. Our previous studies in Drosophila melanogaster have shown that amino acid sensing, via the amino acid response pathway and mTOR-mediated signaling function within adipocytes to control germline stem cell maintenance and ovulation, respectively. Additionally, we demonstrated that insulin/insulin-like growth factor signaling within adipocytes employs distinct effector axes, PI3K/Akt1-dependent and -independent, downstream of insulin receptor activity to mediate fat-to-ovary communication. Here, we report that the Ras/MAPK signaling axis functions in adipocytes to regulate early germline cyst survival and ovulation of mature oocytes but is not important for germline stem cell maintenance or the progression through vitellogenesis. Thus, these studies uncover the complexity of signaling pathway activity that mediates inter-organ communication.
    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: Review
    背景:儿童系统性红斑狼疮(cSLE)被认为是一种多基因自身免疫性疾病;然而,随着最近对几种相关的新型高外显率遗传变异的认识,单基因狼疮样表型正在出现。放射病,一组由RAS/MAPK通路突变引起的疾病,最近被描述为单基因狼疮的原因。
    方法:我们介绍了一个13岁男孩,患有Noonan样综合征,头发松弛,患有单基因狼疮。肾活检证实了III类狼疮性肾炎,并确定了斑马体的存在。
    结论:放射病是单基因狼疮的一个原因。我们报告了一例患有Noonan样综合征并伴有毛发松弛的儿童单基因狼疮的新病例。在这种情况下从未描述过的狼疮性肾炎,可能是演示的一部分。SLE或RASopathies中斑马体的存在不清楚,但是没有其他已知的疾病(法布里病或药物)被确定为我们患者斑马体的原因。
    BACKGROUND: Childhood systemic lupus erythematosus (cSLE) has been considered as a polygenic autoimmune disease; however, a monogenic lupus-like phenotype is emerging with the recent recognition of several related novel high-penetrance genetic variants. RASopathies, a group of disorders caused by mutations in the RAS/MAPK pathway, have been recently described as a cause of monogenic lupus.
    METHODS: We present a 13-year-old boy with Noonan-like syndrome with loose anagen hair who developed a monogenic lupus. The renal biopsy confirmed a class III lupus nephritis and identified the presence of zebra bodies.
    CONCLUSIONS: RASopathies represent a cause of monogenic lupus. We report a new case of monogenic lupus in a child with Noonan-like syndrome with loose anagen hair. Lupus nephritis which has never been described in this context, may be part of the presentation. The presence of zebra bodies in SLE or RASopathies in unclear, but no other known conditions (Fabry disease or drugs) were identified as the cause of zebra bodies in our patient.
    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
    作为二进制开关,RAS蛋白在信号传导过程中切换到ON/OFF状态,并且在正常条件下处于皮带状态。然而,在RAS相关疾病,如癌症和放射病,调节RAS信号传导的基因或RAS本身的突变会永久激活RAS蛋白。这种转换的结构基础是众所周知的;然而,RAS蛋白调控的确切机制尚不清楚.RAS/MAPK综合征是由RAS/丝裂原活化蛋白激酶通路相关基因的种系突变引起的多系统发育障碍,影响1/1-2,000儿童。这些包括各种疾病,如努南综合征(NS)和NS相关疾病(NSRD),例如心脏皮肤(CFC)综合征,科斯特洛综合征(CS),和具有多个腹水的NS(NSML,也称为LEOPARD综合征)。与放射病相关的心肌病(CM)和肥厚型心肌病的频繁表现表明,放射病可能是CM的潜在致病因素。然而,目前的支持证据是零星和不清楚的。RASopathy患者还表现出广泛的先天性心脏病(CHD)。超过15个基因编码RAS/MAPK信号通路的成分,这些成分对细胞周期至关重要,并在增殖中起调节作用。分化,增长,和新陈代谢。这些基因与这些综合征的分子遗传发病机制有关。然而,一方面是给定综合征的遗传异质性,另一方面是多个综合征的等位基因,这使得分类难以诊断RAS/MAPK相关疾病。尽管在大多数拉丝菌中存在一些遗传同质性,几种放射病是等位基因疾病。这种等位基因指向关键信号节点的作用,并揭示了这些相关综合征之间的重叠。尽管通过识别因果突变和对其病理生理后果的功能分析,在理解RASpathy的病理生理学方面取得了相当大的进展,对于许多被诊断为放射病的患者,仍然存在未知的因果基因.
    As binary switches, RAS proteins switch to an ON/OFF state during signaling and are on a leash under normal conditions. However, in RAS-related diseases such as cancer and RASopathies, mutations in the genes that regulate RAS signaling or the RAS itself permanently activate the RAS protein. The structural basis of this switch is well understood; however, the exact mechanisms by which RAS proteins are regulated are less clear. RAS/MAPK syndromes are multisystem developmental disorders caused by germline mutations in genes associated with the RAS/mitogen-activated protein kinase pathway, impacting 1 in 1,000-2,500 children. These include a variety of disorders such as Noonan syndrome (NS) and NS-related disorders (NSRD), such as cardio facio cutaneous (CFC) syndrome, Costello syndrome (CS), and NS with multiple lentigines (NSML, also known as LEOPARD syndrome). A frequent manifestation of cardiomyopathy (CM) and hypertrophic cardiomyopathy associated with RASopathies suggest that RASopathies could be a potential causative factor for CM. However, the current supporting evidence is sporadic and unclear. RASopathy-patients also display a broad spectrum of congenital heart disease (CHD). More than 15 genes encode components of the RAS/MAPK signaling pathway that are essential for the cell cycle and play regulatory roles in proliferation, differentiation, growth, and metabolism. These genes are linked to the molecular genetic pathogenesis of these syndromes. However, genetic heterogeneity for a given syndrome on the one hand and alleles for multiple syndromes on the other make classification difficult in diagnosing RAS/MAPK-related diseases. Although there is some genetic homogeneity in most RASopathies, several RASopathies are allelic diseases. This allelism points to the role of critical signaling nodes and sheds light on the overlap between these related syndromes. Even though considerable progress has been made in understanding the pathophysiology of RASopathy with the identification of causal mutations and the functional analysis of their pathophysiological consequences, there are still unidentified causal genes for many patients diagnosed with RASopathies.
    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:环状RNA(circularRNAs)是非编码RNA。越来越多的证据表明,circRNAs在人类生物过程中起着关键作用,尤其是肿瘤发生,和发展。然而,circRNAs在肝细胞癌(HCC)中的确切作用机制尚不清楚.
    方法:使用生物信息学工具和RT-qPCR来鉴定circdhPR的作用,来自二氢蝶啶还原酶(DHPR)基因座的circRNA,在HCC和癌旁组织中。采用Kaplan-Meier分析和Cox比例风险模型分析circdhPR表达与患者预后的相关性。慢病毒载体用于建立稳定的circdhpr过表达细胞。体外和体内研究表明,circDHPR会影响肿瘤的增殖和转移。机械测定,包括西方印迹,免疫组织化学,双荧光素酶报告分析,荧光原位杂交,和RNA免疫沉淀,已经证明了circDHPR的分子机制。
    结果:CircDHPR在HCC中下调,低circDHPR表达与低总生存率和无病生存率相关。CircDHPR过表达在体外和体内抑制肿瘤生长和转移。进一步的系统研究表明,circdhpr结合miR-3194-5p,RASGEF1B的上游调节器。这种内源性竞争抑制了miR-3194-5p的沉默效应。我们证实,circdhpr过表达抑制肝癌的生长和转移,通过海绵miR-3194-5p上调RASGEF1B的表达,被认为是Ras/MAPK信号通路的抑制因子。
    结论:环状DHPR表达异常导致细胞增殖失控,肿瘤发生,和转移。CircDHPR可作为HCC的生物标志物和治疗靶标。
    BACKGROUND: Circular RNAs (circRNAs) are noncoding RNAs. Accumulating evidence suggests that circRNAs play a critical role in human biological processes, especially tumorigenesis, and development. However, the exact mechanisms of action of circRNAs in hepatocellular carcinoma (HCC) remain unclear.
    METHODS: Bioinformatic tools and RT-qPCR were used to identify the role of circDHPR, a circRNA derived from the dihydropteridine reductase (DHPR) locus, in HCC and para-carcinoma tissues. Kaplan-Meier analysis and the Cox proportional hazard model were used to analyze the correlation between circDHPR expression and patient prognosis. Lentiviral vectors were used to establish stable circDHPR-overexpressing cells. In vitro and in vivo studies have shown that tumor proliferation and metastasis are affected by circDHPR. Mechanistic assays, including Western blotting, immunohistochemistry, dual-luciferase reporter assays, fluorescence in situ hybridization, and RNA immunoprecipitation, have demonstrated the molecular mechanism underlying circDHPR.
    RESULTS: CircDHPR was downregulated in HCC, and low circDHPR expression was associated with poor overall survival and disease-free survival rates. CircDHPR overexpression inhibits tumor growth and metastasis in vitro and in vivo. Further systematic studies revealed that circDHPR binds to miR-3194-5p, an upstream regulator of RASGEF1B. This endogenous competition suppresses the silencing effect of miR-3194-5p. We confirmed that circDHPR overexpression inhibited HCC growth and metastasis by sponging miR-3194-5p to upregulate the expression of RASGEF1B, which is regarded as a suppressor of the Ras/MAPK signaling pathway.
    CONCLUSIONS: Aberrant circDHPR expression leads to uncontrolled cell proliferation, tumorigenesis, and metastasis. CircDHPR may serve as a biomarker and therapeutic target for HCC.
    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
    暂无摘要。
    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:乳腺癌是全世界女性最常见的肿瘤。糖尿病是一种全球性的慢性代谢性疾病,发病率逐年上升。据报道,糖尿病可以积极调节许多肿瘤的发展。然而,高血糖环境调节乳腺癌的具体机制尚不清楚。PFKFB3(6-磷酸果糖-2-激酶/果糖-2,6-双磷酸酶3)是糖尿病糖酵解过程的关键调节因子,以及乳腺癌的启动子.所以,我们希望在本研究中探讨PFKFB3与伴有高血糖的乳腺癌患者的不良预后之间的潜在联系.
    方法:细胞培养物用于构建不同葡萄糖的乳腺癌细胞系。采用免疫组织化学方法分析良性乳腺组织中PFKFB3蛋白水平,浸润性导管癌伴糖尿病和浸润性导管癌不伴糖尿病。采用Kaplan-Meier绘图仪数据库和GEO数据库(GSE61304)分析不同PFKFB3表达的乳腺癌患者的生存期。采用Westernblot方法分析乳腺癌细胞中PFKFB3、上皮间质转化(EMT)相关蛋白和细胞外调节蛋白激酶(ERK)的蛋白水平。利用基因集癌症分析(GSCA)来研究PFKFB3的潜在下游信号传导途径。TargetScan和OncomiR用于探索高血糖导致PFKFB3过度表达的潜在机制。利用转染(包括siRNA和miRNA转染引物)来抑制或模拟相应RNA的表达。细胞功能测定(包括细胞计数,MTT,菌落形成,伤口愈合,和细胞迁移测定)用于探索乳腺癌细胞的增殖和迁移。
    结果:在这项研究中,我们通过体外细胞实验和组织学实验证明,PFKFB3在合并高血糖的乳腺癌中的表达高于正常血糖的乳腺癌。PFKFB3的过表达降低了乳腺癌患者的生存期,并与乳腺癌合并糖尿病的许多临床病理参数相关。PFKFB3在高血糖环境中促进乳腺癌的增殖和迁移,可能受miR-26的调控。此外,PFKFB3在高血糖环境中刺激乳腺癌的上皮-间质转化。在下游机制探索方面,我们预测并验证了PFKFB3通过RAS/MAPK通路对乳腺癌合并高血糖的促癌作用。
    结论:结论:PFKFB3可在高血糖状态下过度表达,可能成为乳腺癌合并糖尿病的潜在治疗靶点。
    BACKGROUND: Breast cancer is the most common tumor in women worldwide. Diabetes mellitus is a global chronic metabolic disease with increasing incidence. Diabetes mellitus has been reported to positively regulate the development of many tumors. However, the specific mechanism of hyperglycemic environment regulating breast cancer remains unclear. PFKFB3 (6-phosphofructose-2-kinase/fructose-2, 6-bisphosphatase 3) is a key regulatory factor of the glycolysis process in diabetes mellitus, as well as a promoter of breast cancer. So, we want to explore the potential link between PFKFB3 and the poor prognosis of breast cancer patients with hyperglycemia in this study.
    METHODS: Cell culture was utilized to construct different-glucose breast cancer cell lines. Immunohistochemistry was adopted to analyze the protein level of PFKFB3 in benign breast tissues, invasive ductal carcinoma with diabetes and invasive ductal carcinoma without diabetes. The Kaplan-Meier plotter database and GEO database (GSE61304) was adopted to analyze the survival of breast cancer patients with different PFKFB3 expression. Western blot was adopted to analyze the protein level of PFKFB3, epithelial-mesenchymal transition (EMT)-related protein and extracellular regulated protein kinases (ERK) in breast cancer cells. Gene Set Cancer Analysis (GSCA) was utilized to investigate the potential downstream signaling pathways of PFKFB3. TargetScan and OncomiR were utilized to explore the potential mechanism of PFKFB3 overexpression by hyperglycemia. Transfections (including siRNAs and miRNA transfection premiers) was utilized to restrain or mimic the expression of the corresponding RNA. Cell functional assays (including cell counting, MTT, colony formation, wound-healing, and cell migration assays) were utilized to explore the proliferation and migration of breast cancer cells.
    RESULTS: In this study, we demonstrated that the expression of PFKFB3 in breast cancer complicated with hyperglycemia was higher than that in breast cancer with euglycemia through cell experiment in vitro and histological experiment. PFKFB3 overexpression decreased the survival period of breast cancer patients and was correlated with a number of clinicopathological parameters of breast cancer complicated with diabetes. PFKFB3 promoted the proliferation and migration of breast cancer in a hyperglycemic environment and might be regulated by miR-26. In addition, PFKFB3 stimulated epithelial-mesenchymal transition of breast cancer in a hyperglycemic environment. In terms of downstream mechanism exploration, we predicted and verified the cancer-promoting effect of PFKFB3 in breast cancer complicated with hyperglycemia through RAS/MAPK pathway.
    CONCLUSIONS: In conclusion, PFKFB3 could be overexpressed by hyperglycemia and might be a potential therapeutic target for breast cancer complicated with diabetes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号