PFKFB3

PFKFB3
  • 文章类型: Journal Article
    急性肺损伤(ALI)是一种以肺泡结构破坏和过度炎症反应为特征的破坏性呼吸系统疾病。巨噬细胞的有氧糖酵解在ALI的病理生理中起着至关重要的作用。先前的研究表明,炎症细胞中关键限速酶6-磷酸果糖-2-激酶/果糖-2,6-双磷酸酶3(PFKFB3)的表达显着增加,促进炎症细胞糖酵解速率的增加。然而,对PFKFB3在巨噬细胞炎症和ALI中的生物学功能知之甚少。在这项研究中,我们发现PFKFB3在脂多糖(LPS)诱导的ALI小鼠和巨噬细胞中显著增加。敲除pfkfb3减弱LPS诱导的糖酵解通量,减少了促炎细胞因子的释放,和失活的NF-κB信号通路在巨噬细胞中。随后,我们发现脱氢莫来石内酯(DL),一种天然的倍半萜内酯,PFKFB3的mRNA和蛋白水平均显著降低。此外,它减少了体外炎性细胞因子的释放和失活的NF-κB通路。因此,DL减轻了LPS诱导的肺水肿,减少了小鼠肺组织中炎性细胞的浸润。总之,我们的研究揭示了PFKFB3在LPS诱导的炎症中的重要作用,并发现了DL对ALI的保护作用的新分子机制。
    Acute lung injury (ALI) is a destructive respiratory disease characterized by alveolar structural destruction and excessive inflammation responses. Aerobic glycolysis of macrophages plays a crucial role in the pathophysiology of ALI. Previous studies have shown that the expression of the key rate-limiting enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) in inflammatory cells is significantly increased, which promotes an increase in the rate of glycolysis in inflammatory cells. However, little is known about the biological functions of PFKFB3 in macrophage inflammation and ALI. In this study, we identified that PFKFB3 is markedly increased in lipopolysaccharide (LPS)-induced ALI mice and macrophages. Knockdown of pfkfb3 attenuated LPS-induced glycolytic flux, decreased the release of pro-inflammatory cytokines, and inactivated NF-κB signaling pathway in macrophages. Subsequently, we found that dehydrocostus lactone (DL), a natural sesquiterpene lactone, significantly decreased both the mRNA and protein levels of PFKFB3. Furthermore, it reduced the release of inflammatory cytokines and inactivated NF-κB pathways in vitro. Accordingly, DL alleviated LPS-induced pulmonary edema and reduced the infiltration of inflammatory cells in mouse lung tissue. In summary, our study reveals the vital role of PFKFB3 in LPS-induced inflammation and discovers a novel molecular mechanism underlying DL\'s protective effects on ALI.
    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
    动脉粥样硬化的发生和发展与巨噬细胞的参与密切相关。虽然NLRP3炎症小体激活对局部高度炎症微环境的创建的贡献是公认的,确切的触发因素仍不清楚。在这项研究中,我们旨在研究NLRP3炎性体激活对缺氧诱导的PFKFB3参与的糖酵解在动脉粥样硬化发展中的调控机制。建立动脉粥样硬化模型,我们选择了用高脂肪西方饮食治疗的ApoE基因敲除小鼠。然后我们量化了HIF-1α的表达,PFKFB3和NLRP3。此外,我们在动脉粥样硬化建模过程中使用了PFKFB3抑制剂PFK158.随后通过18F-FDGmicro-PET/CT测定糖酵解活性,离体葡萄糖摄取,和ECAR分析。此外,我们使用脂多糖(LPS)和TNF-α诱导骨髓源性巨噬细胞(BMDMs)在低氧和常氧条件下分化为M1样表型。我们的组织学分析揭示了PFKFB3在人动脉粥样硬化斑块中的积累,证明与NLRP3表达和巨噬细胞的共定位。用PFK158治疗可降低糖酵解活性和NLRP3炎性体活化,从而减轻动脉粥样硬化的发生。机械上,缺氧促进糖酵解重编程和NLRP3炎症小体激活。随后阻断HIF-1α或PFKFB3下调缺氧BMDMs中的NLRP3/Caspase-1/IL-1β通路。我们的研究表明,HIF-1α/PFKFB3/NLRrexP3是动脉粥样硬化中巨噬细胞炎症激活的关键机制。PFKFB3抑制的治疗潜力可能代表了一种有希望的动脉粥样硬化保护策略。
    The onset and progression of atherosclerosis are closely linked to the involvement of macrophages. While the contribution of NLRP3 inflammasome activation to the creation of a local highly inflammatory microenvironment is well recognized, the precise triggers remain unclear. In this study, we aimed to investigate the regulatory mechanism of NLRP3 inflammasome activation in response to hypoxia-induced glycolysis involving PFKFB3 in the development of atherosclerosis. To develop an atherosclerosis model, we selected ApoE knockout mice treated with a high-fat western diet. We then quantified the expression of HIF-1α, PFKFB3, and NLRP3. In addition, we administered the PFKFB3 inhibitor PFK158 during atherosclerosis modeling. The glycolytic activity was subsequently determined through 18F-FDG micro-PET/CT, ex vivo glucose uptake, and ECAR analysis. Furthermore, we employed lipopolysaccharide (LPS) and TNF-α to induce the differentiation of bone marrow-derived macrophages (BMDMs) into M1-like phenotypes under both hypoxic and normoxic conditions. Our histological analyses revealed the accumulation of PFKFB3 in human atherosclerotic plaques, demonstrating colocalization with NLRP3 expression and macrophages. Treatment with PFK158 reduced glycolytic activity and NLRP3 inflammasome activation, thereby mitigating the occurrence of atherosclerosis. Mechanistically, hypoxia promoted glycolytic reprogramming and NLRP3 inflammasome activation in BMDMs. Subsequent blocking of either HIF-1α or PFKFB3 downregulated the NLRP3/Caspase-1/IL-1β pathway in hypoxic BMDMs. Our study demonstrated that the HIF-1α/PFKFB3/NLRP3 axis serves as a crucial mechanism for macrophage inflammation activation in the emergence of atherosclerosis. The therapeutic potential of PFKFB3 inhibition may represent a promising strategy for atheroprotection.
    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
    地黄,牛膝Bl.(A.bidentata),山药,和菊花拉马特(C.桑草)在中国被称为“四大怀庆中药”,用作功能性食品的材料。在本文中,采用UPLC-Q-TOF-MS/MS对4种怀庆中药的成分进行鉴定,黄酮和芳香化合物是这些草药的主要原因。此外,梭菌在氯化钴诱导的HUVECs损伤中表现出最显著的作用,可以减少细胞凋亡和ROS的过量产生,乳酸脱氢酶(LD)和丙酮酸,增加细胞的迁移能力。同时,A.bidentata在异丙肾上腺素诱导的H9C2细胞损伤中表现出最显著的作用,这可以降低ROS的生产过剩水平,BNP,NO,LD和丙酮酸。Westernblot显示,梭菌和A.bidentata也可以降低bax/bcl-2比率的水平,裂解的caspase-3,细胞色素c,HIF-1α,分别为GLUT1、HKII和PFKFB3。
    Rehmannia glutinosa Libosch, Achyranthes bidentata Bl. (A. bidentata), Dioscorea opposita Thunb, and Chrysanthemum morifolium Ramat (C. morifolium) are known as the \'Four Huaiqing Chinese Medicine\' in China, which are used as materials for functional foods. In this paper, the constituents of Four Huaiqing Chinese Medicine were identified by UPLC-Q-TOF-MS/MS, and flavones and aromatic compounds are mainly responsible for these herbs. Moreover, C. morifolium exhibited the most significant effect in cobalt chloride-induced HUVECs injury, which could decrease cell apoptosis and the overproduction of ROS, lactic dehydrogenase (LD) and pyruvic acid, and increase the migration capacity of cells. Meanwhile, A. bidentata exhibited the most significant effect in isoproterenol-induced H9C2 cell injury, which could decrease the levels of ROS overproduction, BNP, NO, LD and pyruvic acid. Western blot revealed that C. morifolium and A. bidentata also could decrease the levels of bax/bcl-2 ratio, cleaved caspase-3, cytochrome c, HIF-1ɑ, GLUT1, HKII and PFKFB3, respectively.
    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:Rho相关的含卷曲螺旋的蛋白激酶(ROCK)的失调与各种恶性肿瘤的转移和进展有关。然而,其中一种异构体,ROCK1在肿瘤细胞中调节糖酵解尚未完全了解。这里,我们试图阐明ROCK1如何通过调节糖酵解活性影响胰腺癌(PC)的进展.
    方法:通过建立沉默细胞模型,体外分析ROCK1的生物学功能。免疫共沉淀证实了ROCK1和c-MYC之间的直接结合,荧光素酶报告基因分析揭示了c-MYC与PFKFB3基因启动子的结合。这些结果在动物实验中得到证实。
    结果:ROCK1在PC组织中高表达,并在细胞质中富集,高表达与预后不良有关。沉默ROCK1可抑制PC细胞的增殖和迁移,促进其凋亡。机械上,ROCK1直接与c-MYC相互作用,促进其磷酸化(Ser62)并抑制其降解,从而增加关键糖酵解调节因子PFKFB3的转录,增强糖酵解活性并促进PC生长。沉默ROCK1增加了吉西他滨(GEM)在体内和体外的敏感性。
    结论:ROCK1通过c-MYC/PFKFB3信号通路促进PC细胞糖酵解活性并促进PC肿瘤生长。ROCK1敲低可以抑制体内PC肿瘤的生长,增加PC肿瘤的GEM敏感性,为PC提供了至关重要的临床治疗策略。
    BACKGROUND: Dysregulation of Rho-associated coiled coil-containing protein kinases (ROCKs) is involved in the metastasis and progression of various malignant tumors. However, how one of the isomers, ROCK1, regulates glycolysis in tumor cells is incompletely understood. Here, we attempted to elucidate how ROCK1 influences pancreatic cancer (PC) progression by regulating glycolytic activity.
    METHODS: The biological function of ROCK1 was analyzed in vitro by establishing a silenced cell model. Coimmunoprecipitation confirmed the direct binding between ROCK1 and c-MYC, and a luciferase reporter assay revealed the binding of c-MYC to the promoter of the PFKFB3 gene. These results were verified in animal experiments.
    RESULTS: ROCK1 was highly expressed in PC tissues and enriched in the cytoplasm, and its high expression was associated with a poor prognosis. Silencing ROCK1 inhibited the proliferation and migration of PC cells and promoted their apoptosis. Mechanistically, ROCK1 directly interacted with c-MYC, promoted its phosphorylation (Ser 62) and suppressed its degradation, thereby increasing the transcription of the key glycolysis regulatory factor PFKFB3, enhancing glycolytic activity and promoting PC growth. Silencing ROCK1 increased gemcitabine (GEM) sensitivity in vivo and in vitro.
    CONCLUSIONS: ROCK1 promotes glycolytic activity in PC cells and promotes PC tumor growth through the c-MYC/PFKFB3 signaling pathway. ROCK1 knockdown can inhibit PC tumor growth in vivo and increase the GEM sensitivity of PC tumors, providing a crucial clinical therapeutic strategy for PC.
    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
    金黄色葡萄球菌(S。金黄色葡萄球菌)在乳腺上皮细胞中持续存在延长的时间,利用宿主代谢资源促进复制。这项研究揭示了细胞内金黄色葡萄球菌重编程宿主代谢的机制,PFKFB3在此过程中起着至关重要的作用。机械上,金黄色葡萄球菌诱导的线粒体损伤,导致线粒体活性氧(mROS)水平升高和电子传递链(ETC)功能障碍。此外,金黄色葡萄球菌将线粒体动力学的平衡从融合转变为裂变,随后激活PINK1-PRKN依赖性线粒体自噬,导致沉默蛋白3(SIRT3)的丢失,以稳定低氧诱导因子1α(HIF1α),并将宿主代谢向增强的糖酵解转移。PFKFB3的抑制逆转了金黄色葡萄球菌诱导的线粒体损伤和SIRT3的降解。总的来说,我们的研究结果阐明了金黄色葡萄球菌重编程宿主代谢的机制,并为金黄色葡萄球菌感染的治疗提供了见解.
    Staphylococcus aureus (S. aureus) persists within mammary epithelial cells for an extended duration, exploiting the host metabolic resources to facilitate replication. This study revealed a mechanism by which intracellular S. aureus reprograms host metabolism, with PFKFB3 playing a crucial role in this process. Mechanistically, S. aureus induced mitochondrial damage, leading to increased levels of mitochondrial reactive oxygen species (mROS) and dysfunction in electron transport chain (ETC). Moreover, S. aureus shifted the balance of mitochondrial dynamics from fusion to fission, subsequently activating PINK1-PRKN-dependent mitophagy, causing loss of the sirtuin 3 (SIRT3) to stabilize hypoxic inducible factor 1α (HIF1α), and shifting the host metabolism toward enhanced glycolysis. The inhibition of PFKFB3 reversed the mitochondrial damage and degradation of SIRT3 induced by S. aureus. Overall, our findings elucidate the mechanism by which S. aureus reprograms host metabolism and offer insights into the treatment of S. aureus infection.
    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
    6-磷酸果糖-2-激酶/果糖-2,6-双磷酸酶-3(PFKFB3)通过参与肿瘤有氧糖酵解影响癌症进展。在这项研究中,我们旨在通过分析公开数据库的组合来评估PFKFB3在膀胱癌(BLCA)患者中的预后意义,临床患者数据,还有我们医院的膀胱肿瘤样本.膀胱癌的单细胞和大量RNA-seq数据,从ENA获得,GEO,和TCGA数据库,用于我们的分析。结果表明,与配对的正常组织相比,膀胱癌中的PFKFB3mRNA表达显着升高。此外,PFKFB3高表达的BLCA患者预后差(P<0.05)。为了验证这些发现,我们对来自青岛大学附属医院或北京协和医院接受根治性膀胱切除术的89例BLCA患者的标本进行了临床资料和免疫组织化学染色.该验证过程的发现证实,PFKFB3的高表达可作为预测BLCA患者预后较差的生物标志物(OR:2.462,95%CI:1.202-5.042,P=0.012)。为了便于临床应用,我们开发了一个基于四个变量的列线图,包括PFKFB3表达式,预测BLCA患者的生存率。重要的是,该列线图显示出0.03的低平均预测误差。一起来看,我们的研究结果表明,PFKFB3有可能作为BLCA患者的预后生物标志物和治疗靶点.
    6-phosphofructo-2-kinase/fructose-2,6-biphosphatase-3 (PFKFB3) influences cancer progression via participating in tumor aerobic glycolysis. In this study, we aimed to evaluate the prognostic significance of PFKFB3 in bladder cancer (BLCA) patients by analyzing a combination of publicly available databases, clinical patient data, and bladder tumor samples from our hospital. Single-cell and bulk RNA-seq data of bladder cancer, obtained from ENA, GEO, and TCGA databases, were utilized for our analysis. The results indicated that PFKFB3 mRNA expression was markedly elevated in bladder cancer compared to paired normal tissue. Furthermore, BLCA patients with high PFKFB3 expression exhibited a significantly worse prognosis (P < 0.05). To validate these findings, clinical data and immunohistochemistry staining were performed on specimens obtained from 89 BLCA patients who underwent radical cystectomy at either Qingdao University Affiliated Hospital or Peking Union Medical College Hospital. The findings from this verification process confirmed that high expression of PFKFB3 serves as a biomarker for predicting worse prognosis in BLCA patients (OR: 2.462, 95 % CI: 1.202-5.042, P = 0.012). To facilitate clinical application, we developed a nomogram based on four variables, including PFKFB3 expression, to predict the survival of BLCA patients. Importantly, this nomogram demonstrated a low mean prediction error of 0.03. Taken together, our findings suggest that PFKFB3 has the potential to serve as both a prognostic biomarker and a therapeutic target for BLCA patients.
    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
    已证明肾脏中持续升高的糖酵解可促进慢性肾脏疾病(CKD)。然而,潜在的机制仍不清楚.这里,我们观察到6-磷酸果糖-2-激酶/果糖-2,6-双磷酸酶3(PFKFB3),一种关键的糖酵解酶,在小鼠缺血再灌注损伤(IRI)后,肾近端肾小管细胞(PTCs)被显著诱导,以及CKD患者的多种病因。PFKFB3的表达与肾脏纤维化的严重程度呈正相关。此外,CKD患者和小鼠表现出增加的尿乳酸/肌酸水平和肾乳酸,分别。PFKFB3的PTCs特异性缺失显著降低了肾乳酸水平,减轻炎症和纤维化,并在IRI小鼠模型中保留肾功能。在具有PFKFB3杂合缺陷的小鼠或用PFKFB3抑制剂治疗的小鼠中观察到类似的保护作用。机械上,来自PFKFB3介导的管状糖酵解重编程的乳酸显着增强了组蛋白的乳酸,尤其是H4K12la,富含NF-κB信号基因如Ikbkb的启动子,瑞拉,Relb,激活它们的转录并促进炎症反应。Further,PFKFB3的PTCs特异性缺失抑制IKKβ的活化,IκBα,和IRI肾脏中的p65。此外,CKD患者H4K12la水平升高与肾脏炎症和纤维化呈正相关。这些发现表明,管状PFKFB3可能通过促进H4K12la介导的基因转录及其激活在增强NF-κB信号传导中起双重作用。因此,在肾小管细胞中靶向PFKFB3介导的NF-κB信号通路可能是CKD治疗的新策略.
    Persistently elevated glycolysis in kidney has been demonstrated to promote chronic kidney disease (CKD). However, the underlying mechanism remains largely unclear. Here, we observed that 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), a key glycolytic enzyme, was remarkably induced in kidney proximal tubular cells (PTCs) following ischemia-reperfusion injury (IRI) in mice, as well as in multiple etiologies of patients with CKD. PFKFB3 expression was positively correlated with the severity of kidney fibrosis. Moreover, patients with CKD and mice exhibited increased urinary lactate/creatine levels and kidney lactate, respectively. PTC-specific deletion of PFKFB3 significantly reduced kidney lactate levels, mitigated inflammation and fibrosis, and preserved kidney function in the IRI mouse model. Similar protective effects were observed in mice with heterozygous deficiency of PFKFB3 or those treated with a PFKFB3 inhibitor. Mechanistically, lactate derived from PFKFB3-mediated tubular glycolytic reprogramming markedly enhanced histone lactylation, particularly H4K12la, which was enriched at the promoter of NF-κB signaling genes like Ikbkb, Rela, and Relb, activating their transcription and facilitating the inflammatory response. Further, PTC-specific deletion of PFKFB3 inhibited the activation of IKKβ, I κ B α, and p65 in the IRI kidneys. Moreover, increased H4K12la levels were positively correlated with kidney inflammation and fibrosis in patients with CKD. These findings suggest that tubular PFKFB3 may play a dual role in enhancing NF-κB signaling by promoting both H4K12la-mediated gene transcription and its activation. Thus, targeting the PFKFB3-mediated NF-κB signaling pathway in kidney tubular cells could be a novel strategy for CKD therapy.
    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
    最近在弥漫性中线胶质瘤(DMG)中的研究表明,表观基因组失调与代谢重新布线之间存在很强的联系。这里,我们评估了在H3.3K27MDMG中靶向名为磷酸果糖-2-激酶/果糖-2,6-双磷酸酶3(PFKFB3)的糖酵解蛋白的价值。我们观察到H3.3K27M细胞的活力被PFK15(一种有效的PFKFB3抑制剂)显著降低。此外,PFKFB3抑制诱导细胞凋亡和G2/M期阻滞。有趣的是,K27M突变等位基因的CRISPR敲除对观察到的表型具有协同作用。总之,我们确定PFKFB3是H3.3K27MDMG的新靶标,使PFK15成为未来动物研究和临床试验的潜在候选者。
    Recent studies in Diffuse Midline Gliomas (DMG) demonstrated a strong connection between epigenome dysregulation and metabolic rewiring. Here, we evaluated the value of targeting a glycolytic protein named Phosphofructo-2-kinase/Fructose-2,6-bisphosphatase 3 (PFKFB3) in H3.3K27M DMG. We observed that the viability of H3.3K27M cells is dramatically reduced by PFK15, a potent inhibitor of PFKFB3. Furthermore, PFKFB3 inhibition induced apoptosis and G2/M arrest. Interestingly, CRISPR-Knockout of the K27M mutant allele has a synergistic effect on the observed phenotype. Altogether, we identified PFKFB3 as a new target for H3.3K27M DMG, making PFK15 a potential candidate for future animal studies and clinical trials.
    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
    酶6-磷酸果糖-2-激酶/果糖-2,6-双磷酸酶同工型3(PFKFB3)是糖酵解的关键调节剂,在调节炎症反应中起关键作用,从而促进炎性疾病如脓毒症的发展。尽管它很重要,在脓毒症的背景下,制定靶向PFKFB3的策略仍然具有挑战性.在这项研究中,我们采用基于microRNA的方法降低PFKFB3的表达。通过多个荟萃分析,我们观察到临床脓毒症样本中miR-106a-5p表达下调和PFKFB3表达上调.这些变化也在早期败血症患者和脂多糖(LPS)诱导的败血症小鼠模型的血液单核细胞中得到证实。miR-106a-5p的过表达显著降低了LPS诱导的糖酵解能力的增加,炎症反应,和巨噬细胞的焦亡。机械上,我们将PFKFB3鉴定为miR-106a-5p的直接靶蛋白,并证明其在LPS诱导的巨噬细胞焦凋亡和炎症反应中的重要作用.此外,用agomir-miR-106a-5p治疗在LPS小鼠脓毒症模型中具有保护作用,但这种作用在骨髓特异性Pfkfb3敲除小鼠中减弱。这些发现表明miR-106a-5p通过调节PFKFB3介导的葡萄糖代谢抑制脓毒症巨噬细胞的焦亡和炎症反应,代表了治疗脓毒症的潜在治疗选择。
    The enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoform 3 (PFKFB3) is a critical regulator of glycolysis and plays a key role in modulating the inflammatory response, thereby contributing to the development of inflammatory diseases such as sepsis. Despite its importance, the development of strategies to target PFKFB3 in the context of sepsis remains challenging. In this study, we employed a miRNA-based approach to decrease PFKFB3 expression. Through multiple meta-analyses, we observed a downregulation of miR-106a-5p expression and an upregulation of PFKFB3 expression in clinical sepsis samples. These changes were also confirmed in blood monocytes from patients with early sepsis and from a mouse model of lipopolysaccharide (LPS)-induced sepsis. Overexpression of miR-106a-5p significantly decreased the LPS-induced increase in glycolytic capacity, inflammatory response, and pyroptosis in macrophages. Mechanistically, we identified PFKFB3 as a direct target protein of miR-106a-5p and demonstrated its essential role in LPS-induced pyroptosis and inflammatory response in macrophages. Furthermore, treatment with agomir-miR-106a-5p conferred a protective effect in an LPS mouse model of sepsis, but this effect was attenuated in myeloid-specific Pfkfb3 KO mice. These findings indicate that miR-106a-5p inhibits macrophage pyroptosis and inflammatory response in sepsis by regulating PFKFB3-mediated glucose metabolism, representing a potential therapeutic option for the treatment of sepsis.
    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
    雌激素受体α(ERα)在大约70%的乳腺癌病例中表达,并决定了内分泌治疗的敏感性和有效性。6-磷酸果糖-2-激酶/果糖-2,6-二磷酸酶3(PFKFB3)是一种糖酵解酶,在许多人类肿瘤中高度表达,最近的研究表明,它在提高药物敏感性方面发挥着重要作用。然而,PFKFB3在调节ERα表达中的作用及其机制尚不清楚。这里,我们通过免疫组织化学(IHC)发现,PFKFB3在ER阳性乳腺癌中升高,而PFKFB3的高表达导致预后更差.体外和体内实验证实PFKFB3促进ER阳性乳腺癌细胞增殖。PFKFB3的过度表达促进雌激素非依赖性ER阳性乳腺癌的生长。在无雌激素的情况下,用siPFKFB3处理的MCF7细胞的RNA测序数据显示雌激素信号通路的富集,荧光素酶分析表明,PFKFB3的敲低抑制了ERα的转录活性。机械上,PFKFB3的下调促进STUB1与ERα的结合,通过基于K48的泛素键加速ERα降解。最后,与单独使用每种药物相比,氟维司群联合PFKFB3抑制剂PFK158更有效地抑制了体内ER阳性乳腺癌细胞的生长.总之,这些数据表明,PFKFB3被确定为ER阳性乳腺癌的不良预后因素,并且在ERα稳定性和活性的调节中起着以前未被认识到的作用.我们的结果进一步探索了通过早期与PFKFB3抑制剂联合使用来提高氟维司群敏感性的有效方法。
    Estrogen receptor alpha (ERα) is expressed in approximately 70% of breast cancer cases and determines the sensitivity and effectiveness of endocrine therapy. 6-phosphofructo-2-kinase/fructose-2, 6-biphosphatase3 (PFKFB3) is a glycolytic enzyme that is highly expressed in a great many human tumors, and recent studies have shown that it plays a significant role in improving drug sensitivity. However, the role of PFKFB3 in regulating ERα expression and the underlying mechanism remains unclear. Here, we find by using immunohistochemistry (IHC) that PFKFB3 is elevated in ER-positive breast cancer and high expression of PFKFB3 resulted in a worse prognosis. In vitro and in vivo experiments verify that PFKFB3 promotes ER-positive breast cancer cell proliferation. The overexpression of PFKFB3 promotes the estrogen-independent ER-positive breast cancer growth. In an estrogen-free condition, RNA-sequencing data from MCF7 cells treated with siPFKFB3 showed enrichment of the estrogen signaling pathway, and a luciferase assay demonstrated that knockdown of PFKFB3 inhibited the ERα transcriptional activity. Mechanistically, down-regulation of PFKFB3 promotes STUB1 binding to ERα, which accelerates ERα degradation by K48-based ubiquitin linkage. Finally, growth of ER-positive breast cancer cells in vivo was more potently inhibited by fulvestrant combined with the PFKFB3 inhibitor PFK158 than for each drug alone. In conclusion, these data suggest that PFKFB3 is identified as an adverse prognosis factor for ER-positive breast cancer and plays a previously unrecognized role in the regulation of ERα stability and activity. Our results further explores an effective approach to improve fulvestrant sensitivity through the early combination with a PFKFB3 inhibitor.
    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