PFKP

PFKP
  • 文章类型: Journal Article
    背景:磷酸果糖激酶P(PFKP)是糖酵解中的关键限速酶,在各种病理生理过程中起着至关重要的作用。然而,其在肿瘤中的具体功能尚不清楚。本研究旨在评估PFKP在多种肿瘤类型(泛癌)中的表达和特异性作用,并探讨其作为癌症治疗治疗靶点的潜在临床意义。
    方法:我们分析了PFKP的表达,免疫细胞浸润,使用来自癌症基因组图谱(TCGA)和基因表达综合(GEO)数据库的数据,以及各种癌症的患者预后。此外,我们在肺癌细胞中进行了一系列实验,包括蛋白质印迹,CCK-8测定,集落形成试验,transwell迁移试验,划痕伤口愈合试验,LDH释放试验,和流式细胞术,评价PFKP对肿瘤细胞的影响。
    结果:发现PFKP在大多数癌症中高表达,并被确定为预后危险因素。PFKP表达升高与较差的临床结果相关,特别是在肺腺癌(LUAD)。受试者工作特征(ROC)曲线分析表明,PFKP可以有效区分癌组织和正常组织。PFKP在大多数肿瘤中的表达与肿瘤突变负荷(TMB)显着相关,微卫星不稳定性(MSI),免疫评分,和免疫细胞浸润。体外实验证明PFKP过表达在抑制细胞凋亡的同时促进肺癌细胞增殖和迁移,而PFKP缺乏导致相反的效果。
    结论:PFKP是参与肿瘤发生的癌基因,可能影响肿瘤内的免疫微环境。我们的研究结果表明,PFKP可以作为预测肿瘤预后和免疫治疗疗效的潜在生物标志物。
    BACKGROUND: Phosphofructokinase P (PFKP) is a key rate-limiting enzyme in glycolysis, playing a crucial role in various pathophysiological processes. However, its specific function in tumors remains unclear. This study aims to evaluate the expression and specific role of PFKP across multiple tumor types (Pan-cancer) and to explore its potential clinical significance as a therapeutic target in cancer treatment.
    METHODS: We analyzed the expression of PFKP, immune cell infiltration, and patient prognosis across various cancers using data from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Additionally, we conducted a series of experiments in lung cancer cells, including Western blot, CCK-8 assay, colony formation assay, transwell migration assay, scratch wound healing assay, LDH release assay, and flow cytometry, to evaluate the impact of PFKP on tumor cells.
    RESULTS: PFKP was found to be highly expressed in most cancers and identified as a prognostic risk factor. Elevated PFKP expression is associated with poorer clinical outcomes, particularly in lung adenocarcinoma (LUAD). Receiver operating characteristic (ROC) curve analysis indicated that PFKP can effectively differentiate between cancerous and normal tissues. The expression of PFKP in most tumors showed significant correlations with tumor mutational burden (TMB), microsatellite instability (MSI), immune score, and immune cell infiltration. In vitro experiments demonstrated that PFKP overexpression promotes lung cancer cell proliferation and migration while inhibiting apoptosis, whereas PFKP deficiency results in the opposite effects.
    CONCLUSIONS: PFKP acts as an oncogene involved in tumorigenesis and may influence the immune microenvironment within the tumor. Our findings suggest that PFKP could serve as a potential biomarker for predicting prognosis and the efficacy of immunotherapy in tumors.
    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:磷酸果糖激酶-血小板(PFKP)的异常表达通过修饰多种生物学功能在多种人类癌症的发展中起着至关重要的作用。然而,PFKP在头颈部鳞状细胞癌(HNSCC)中的作用的确切分子机制尚未完全阐明。
    方法:我们评估了120例HNSCC患者的肿瘤和邻近正常组织中PFKP和c-Myc的表达水平。进行了一系列体外和体内实验以探索PFKP和c-Myc之间的反馈回路对HNSCC进展的影响。此外,我们探索了使用患者来源的类器官(PDO)在HNSCC中靶向PFKP和c-Myc的治疗效果,细胞系来源的异种移植物,和患者来源的异种移植物。
    结果:我们的发现表明,PFKP在HNSCC组织和细胞系中经常上调,与预后不良有关。我们的体外和体内实验表明,升高的PFKP促进细胞增殖,血管生成,和HNSCC的转移。机械上,PFKP增加了ERK介导的c-Myc的稳定性,从而推动HNSCC的进展。此外,c-Myc在转录水平刺激PFKP表达,从而在PFKP和c-Myc之间形成正反馈回路。此外,我们的多个模型表明,共同靶向PFKP和c-Myc在HNSCC中触发协同抗肿瘤作用。
    结论:我们的研究证明了PFKP/c-Myc正反馈回路在驱动HNSCC进展中的关键作用,并提示同时靶向PFKP和c-Myc可能是HNSCC的一种新颖有效的治疗策略。
    BACKGROUND: The aberrant expression of phosphofructokinase-platelet (PFKP) plays a crucial role in the development of various human cancers by modifying diverse biological functions. However, the precise molecular mechanisms underlying the role of PFKP in head and neck squamous cell carcinoma (HNSCC) are not fully elucidated.
    METHODS: We assessed the expression levels of PFKP and c-Myc in tumor and adjacent normal tissues from 120 HNSCC patients. A series of in vitro and in vivo experiments were performed to explore the impact of the feedback loop between PFKP and c-Myc on HNSCC progression. Additionally, we explored the therapeutic effects of targeting PFKP and c-Myc in HNSCC using Patient-Derived Organoids (PDO), Cell Line-Derived Xenografts, and Patients-Derived Xenografts.
    RESULTS: Our findings indicated that PFKP is frequently upregulated in HNSCC tissues and cell lines, correlating with poor prognosis. Our in vitro and in vivo experiments demonstrate that elevated PFKP facilitates cell proliferation, angiogenesis, and metastasis in HNSCC. Mechanistically, PFKP increases the ERK-mediated stability of c-Myc, thereby driving progression of HNSCC. Moreover, c-Myc stimulates PFKP expression at the transcriptional level, thus forming a positive feedback loop between PFKP and c-Myc. Additionally, our multiple models demonstrate that co-targeting PFKP and c-Myc triggers synergistic anti-tumor effects in HNSCC.
    CONCLUSIONS: Our study demonstrates the critical role of the PFKP/c-Myc positive feedback loop in driving HNSCC progression and suggests that simultaneously targeting PFKP and c-Myc may be a novel and effective therapeutic strategy for HNSCC.
    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:肺癌是全球死亡的主要原因之一。最近的研究表明,环状RNA在多种癌症中失调,但是肺癌的机制仍然不清楚。在我们的工作中,我们探讨了circLDLRAD3在肺癌中的作用机制。
    方法:通过实时定量聚合酶链反应(RT-qPCR)检测肺癌中circLDLRAD3,microRNA-497-5p(miR-497-5p)和血小板型PFK(PFKP)的丰度。同时,通过蛋白质印迹定量PFKP水平.细胞计数试剂盒-8(CCK-8),5-乙炔基-2'-脱氧尿苷(EdU)测定,transwell分析,伤口愈合试验,流式细胞术,westernblot,进行免疫组织化学(IHC)测定和糖酵解代谢分析以进行功能分析。此外,miR-497-5p与circLDLRAD3或FKPF之间的相互作用通过双荧光素酶报告基因和RNA免疫沉淀(RIP)检测.最终,体内实验用于测量circLDLRAD3的作用。
    结果:circLDLRAD3和PFKP水平升高。沉默circLDLRAD3抑制细胞活力,扩散,迁移,肺癌细胞的侵袭和糖酵解代谢及促进细胞凋亡。在机制上,circLDLRAD3作为miR-497-5p海绵调节PFKP水平。MiR-497-5p通过抑制PFKP抑制肺癌的进展。此外,circLDLRAD3敲低也抑制体内肿瘤生长。
    结论:CircLDLRAD3通过调节miR-497-5p增加PFKP的表达促进肺癌的发展,这也为肺癌治疗提供了潜在的靶向治疗。
    OBJECTIVE: Lung cancer is one of the leading causes of death worldwide. Recent studies have shown that circular RNAs are dysregulated in a variety of cancers, but the mechanism in lung cancer is still indistinct. In our work, we explored the action mechanism of circLDLRAD3 in lung cancer.
    METHODS: The abundance of circLDLRAD3, microRNA-497-5p (miR-497-5p) and platelet-type PFK (PFKP) was measured by real-time quantitative polymerase chain reaction (RT-qPCR) in lung cancer. Meanwhile, the level of PFKP was quantified by western blot. Cell counting kit-8 (CCK-8), 5-Ethynyl-2\'-deoxyuridine (EdU) assay, transwell assay, wound healing assay, flow cytometry, western blot, immunohistochemical (IHC) assay and glycolysis metabolism analysis were performed for functional analyses. Furthermore, the interplay between miR-497-5p and circLDLRAD3 or FKPF was detected by the dual-luciferase reporter and RNA Immunoprecipitation (RIP) assays. Eventually, the in vivo experiments were applied to measure the role of circLDLRAD3.
    RESULTS: The levels of circLDLRAD3 and PFKP were increased. Silencing circLDLRAD3 inhibited cell viability, proliferation, migration, invasion and glycolysis metabolism and promoted cell apoptosis in lung cancer cells. In mechanism, circLDLRAD3 regulated PFKP level as a miR-497-5p sponge. MiR-497-5p suppressed the progression of lung cancer by inhibiting PFKP. In addition, circLDLRAD3 knockdown also inhibited tumor growth in vivo.
    CONCLUSIONS: CircLDLRAD3 promoted the development of lung cancer through increasing PFKP expression by regulating miR-497-5p, which also provided a potential targeted therapy for lung cancer treatment.
    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:三阴性乳腺癌(TNBC)仍然是最具挑战性的乳腺癌亚型,缺乏明确的治疗靶点。有氧糖酵解是促进癌症进展的代谢重编程的标志。PFKP是一种参与有氧糖酵解的限速酶,在各种类型的癌症中过度表达。然而,在TNBC中PFKP翻译后修饰的潜在机制和作用尚不清楚.
    方法:探讨PFKP蛋白是否在TNBC的进展中具有潜在的作用,通过CPTAC数据库分析检查TNBC和正常乳腺组织中的PFKP蛋白水平,免疫组化染色(IHC),和蛋白质印迹分析。进一步的CCK-8测定,集落形成试验,EDU掺入测定,和肿瘤异种移植实验用于检测PFKP对TNBC进展的影响。为了阐明USP5-PFKP通路在TNBC进展中的作用,泛素测定,免疫共沉淀(Co-IP),基于质谱的蛋白质鉴定,蛋白质印迹试验,免疫荧光显微镜,体外结合测定,并进行糖酵解试验。
    结果:这里,我们表明PFKP蛋白在TNBC中高表达,这与患者的TNBC进展和不良预后有关。此外,我们证明了PFKP耗竭在体外和体内显着抑制了TNBC的进展。重要的是,我们发现PFKP是去泛素化酶USP5的真正靶标,而USP5介导的去泛素化和PFKP的稳定对癌细胞有氧糖酵解和TNBC进展至关重要.此外,我们发现TNBC样本中USP5和PFKP的表达之间存在很强的正相关。值得注意的是,USP5和PFKP的高表达与不良临床结局显著相关.
    结论:我们的研究将USP5-PFKP轴确立为TNBC进展的重要调节机制,并为未来TNBC治疗的治疗干预提供了理论基础。
    Triple-negative breast cancer (TNBC) remains the most challenging subtype of breast cancer and lacks definite treatment targets. Aerobic glycolysis is a hallmark of metabolic reprogramming that contributes to cancer progression. PFKP is a rate-limiting enzyme involved in aerobic glycolysis, which is overexpressed in various types of cancers. However, the underlying mechanisms and roles of the posttranslational modification of PFKP in TNBC remain unknown.
    To explore whether PFKP protein has a potential role in the progression of TNBC, protein levels of PFKP in TNBC and normal breast tissues were examined by CPTAC database analysis, immunohistochemistry staining (IHC), and western blotting assay. Further CCK-8 assay, colony formation assay, EDU incorporation assay, and tumor xenograft experiments were used to detect the effect of PFKP on TNBC progression. To clarify the role of the USP5-PFKP pathway in TNBC progression, ubiquitin assay, co-immunoprecipitation (Co-IP), mass spectrometry-based protein identification, western blotting assay, immunofluorescence microscopy, in vitro binding assay, and glycolysis assay were conducted.
    Herein, we showed that PFKP protein was highly expressed in TNBC, which was associated with TNBC progression and poor prognosis of patients. In addition, we demonstrated that PFKP depletion significantly inhibited the TNBC progression in vitro and in vivo. Importantly, we identified that PFKP was a bona fide target of deubiquitinase USP5, and the USP5-mediated deubiquitination and stabilization of PFKP were essential for cancer cell aerobic glycolysis and TNBC progression. Moreover, we found a strong positive correlation between the expression of USP5 and PFKP in TNBC samples. Notably, the high expression of USP5 and PFKP was significantly correlated with poor clinical outcomes.
    Our study established the USP5-PFKP axis as an important regulatory mechanism of TNBC progression and provided a rationale for future therapeutic interventions in the treatment of TNBC.
    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
    作为一种历史悠久的化疗药物,5-氟尿嘧啶(5-FU)广泛用于临床治疗结直肠癌(CRC)。然而,相当一部分患者在某个阶段出现5-FU耐药性,这是一个巨大的挑战。因此,需要揭示可以指导制定有效策略来克服5-FU抗性的机制。这里,我们报道PFKP在HCT116/5-FUCRC中表达较高。此外,PFKP的遗传抑制抑制糖酵解,NF-κB激活,GLUT1和HK2在HCT116/5-FU细胞中的表达。PFKP过表达通过NF-κB信号通路促进HCT116细胞糖酵解和GLUT1和HK2的表达。我们的功能测定表明,PFKP沉默可以使HCT116/5-FU细胞对5-FU敏感,凋亡细胞数量增加。相比之下,PFKP的强制表达在HCT116细胞中赋予5-FU抗性。此外,PFKP沉默显著抑制CRC异种移植肿瘤生长。值得注意的是,PFKP沉默和5-FU的组合抑制肿瘤生长。因此,我们的结果表明,PFKP通过促进糖酵解增强5-FU抗性,这表明PFKP可能是5-FU耐药CRC靶向治疗的新候选药物。
    As a long-established chemotherapy drug, 5-fluorouracil (5-FU) is widely used to clinically manage colorectal cancer (CRC). However, a substantial portion of patients develop 5-FU resistance at some stage, which poses a great challenge. Therefore, revealing the mechanisms that could guide the development of effective strategies to overcome 5-FU resistance is required. Here, we report that the expression of PFKP was higher in HCT116/5-FU CRC. Furthermore, genetic suppression of PFKP suppresses glycolysis, NF-κB activation, and expression of GLUT1 and HK2 in HCT116/5-FU cells. PFKP overexpression promotes glycolysis and expression of GLUT1 and HK2 via the NF-κB signaling pathway in HCT116 cells. Our functional assays demonstrated that PFKP silencing could sensitize HCT116/5-FU cells to 5-FU with an elevated population of apoptotic cells. In contrast, forced expression of PFKP conferred 5-FU resistance in HCT116 cells. Furthermore, PFKP silencing significantly inhibited CRC xenograft tumor growth. Notably, the combination of PFKP silencing and 5-FU inhibited tumor growth. Therefore, our results demonstrated that PFKP enhances 5-FU resistance by promoting glycolysis, indicating that PFKP could be a novel candidate for targeted therapy for 5-FU-resistant CRC.
    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
    有氧糖酵解对癌症进展至关重要,可用于癌症治疗。这里,我们报道,人羧甲基丁烯糖苷酶同源物(类羧甲基丁烯糖苷酶[CMBL])通过重编程糖酵解在结直肠癌(CRC)中发挥肿瘤抑制因子的作用.CMBL的抗癌作用是通过其与E3泛素连接酶TRIM25和糖酵解酶磷酸果糖激酶-1血小板型(PFKP)的相互作用来介导的。异位CMBL增强TRIM25与PFKP的结合,导致PFKP的泛素化和蛋白酶体降解。有趣的是,CMBL被p53转录激活,以响应基因毒性应激,p53激活通过促进PFKP降解抑制糖酵解。值得注意的是,CMBL缺陷,削弱p53抑制糖酵解的能力,使肿瘤对涉及糖酵解抑制剂2-脱氧葡萄糖的联合治疗更敏感。一起来看,我们的研究表明,CMBL通过抑制糖酵解抑制CRC生长,并提出了治疗CMBL缺陷CRC的潜在组合策略.
    Aerobic glycolysis is critical for cancer progression and can be exploited in cancer therapy. Here, we report that the human carboxymethylenebutenolidase homolog (carboxymethylenebutenolidase-like [CMBL]) acts as a tumor suppressor by reprogramming glycolysis in colorectal cancer (CRC). The anti-cancer action of CMBL is mediated through its interactions with the E3 ubiquitin ligase TRIM25 and the glycolytic enzyme phosphofructokinase-1 platelet type (PFKP). Ectopic CMBL enhances TRIM25 binding to PFKP, leading to the ubiquitination and proteasomal degradation of PFKP. Interestingly, CMBL is transcriptionally activated by p53 in response to genotoxic stress, and p53 activation represses glycolysis by promoting PFKP degradation. Remarkably, CMBL deficiency, which impairs p53\'s ability to inhibit glycolysis, makes tumors more sensitive to a combination therapy involving the glycolysis inhibitor 2-deoxyglucose. Taken together, our study demonstrates that CMBL suppresses CRC growth by inhibiting glycolysis and suggests a potential combination strategy for the treatment of CMBL-deficient CRC.
    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
    骨关节炎(OA)是最常见的关节疾病,但尚未批准用于OA治疗的疾病缓解药物.线粒体自噬通过选择性清除功能失调的线粒体参与线粒体稳态调节,这可能有助于OA的软骨退变。这里,我们提供了OA软骨细胞线粒体自噬受损的证据,这加剧了软骨细胞的退化.在几种经典的线粒体自噬调节途径和受体中,我们发现FUNDC1通过诱导线粒体自噬在维持软骨细胞稳态中起关键作用。FUNDC1体外敲除和体内敲除减少线粒体自噬并加剧线粒体功能障碍,加重软骨细胞变性和OA进展。通过关节内注射腺相关病毒的FUNDC1过表达减轻了OA中的软骨退变。机械上,我们的研究表明,PFKP与FUNDC1相互作用并使其去磷酸化,从而诱导软骨细胞中的线粒体自噬。进一步的分析确定KD025是通过增加FUNDC1-PFKP相互作用来恢复软骨细胞线粒体自噬的候选药物,从而减轻DMM诱导的OA小鼠的软骨退化。我们的研究强调了FUNDC1-PFKP相互作用通过线粒体自噬诱导在软骨细胞稳态中的作用,并确定KD025是通过增加软骨细胞线粒体自噬治疗OA的有前途的药物。
    Osteoarthritis (OA) is the most common joint disease, but no disease-modifying drugs have been approved for OA treatment. Mitophagy participates in mitochondrial homeostasis regulation by selectively clearing dysfunctional mitochondria, which might contribute to cartilage degeneration in OA. Here, we provide evidence of impaired mitophagy in OA chondrocytes, which exacerbates chondrocyte degeneration. Among the several classic mitophagy-regulating pathways and receptors, we found that FUNDC1 plays a key role in preserving chondrocyte homeostasis by inducing mitophagy. FUNDC1 knockdown in vitro and knockout in vivo decreased mitophagy and exacerbated mitochondrial dysfunction, exacerbating chondrocyte degeneration and OA progression. FUNDC1 overexpression via intra-articular injection of adeno-associated virus alleviated cartilage degeneration in OA. Mechanistically, our study demonstrated that PFKP interacts with and dephosphorylates FUNDC1 to induce mitophagy in chondrocytes. Further analysis identified KD025 as a candidate drug for restoring chondrocyte mitophagy by increasing the FUNDC1-PFKP interaction and thus alleviating cartilage degeneration in mice with DMM-induced OA. Our study highlights the role of the FUNDC1-PFKP interaction in chondrocyte homeostasis via mitophagy induction and identifies KD025 as a promising agent for treating OA by increasing chondrocyte mitophagy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    磷酸果糖激酶(PFK)是在糖酵解中起作用的关键酶之一。研究表明PFKP调节细胞增殖,凋亡,自噬,细胞迁移/转移,通过糖酵解和独立于糖酵解的功能进行干性。PFKP不仅在细胞质中发挥其功能,而且在细胞膜上,在线粒体上,在溶酶体膜上,在细胞核中。PFKP的功能在癌细胞中被广泛研究。PFKP在某些免疫细胞中也高表达;然而,PFKP在免疫细胞中的作用的研究是有限的。在这次审查中,我们总结了PFKP在癌细胞中的表达和活性调控。PFKP由于其过表达和在癌细胞中的重要功能而可用作预后标志物。因此,特异性靶向/抑制PFKP可能是癌症治疗的关键和有前景的策略.
    Phosphofructokinase (PFK) is one of the key enzymes that functions in glycolysis. Studies show that PFKP regulates cell proliferation, apoptosis, autophagy, cell migration/metastasis, and stemness through glycolysis and glycolysis-independent functions. PFKP performs its function not only in the cytoplasm, but also at the cell membrane, on the mitochondria, at the lysosomal membrane, and in the nucleus. The functions of PFKP are extensively studied in cancer cells. PFKP is also highly expressed in certain immune cells; nevertheless, the study of the PFKP\'s role in immune cells is limited. In this review, we summarize how the expression and activity of PFKP are regulated in cancer cells. PFKP may be applied as a prognostic marker due to its overexpression and significant functions in cancer cells. As such, specifically targeting/inhibiting PFKP may be a critical and promising strategy for cancer 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
    目的:皮肤黑素瘤是一种侵袭性和致命性的癌症,由参与皮肤色素沉着的细胞的恶性转化引起。糖酵解与癌症进展密切相关,但其在黑色素瘤中的确切作用尚未得到广泛研究。这里,我们研究了糖酵解调节因子磷酸果糖激酶1血小板亚型(PFKP)在黑色素瘤进展中的作用.
    方法:用免疫组织化学方法分析人黑色素瘤组织中PFKP的表达。通过siRNA敲低PFKP和PFKP的过表达进行体外评估其功能。CCK-8测定用于评估细胞增殖。通过测量细胞外酸化速率(ECAR)来确定糖酵解活性,乳酸水平,和ATP含量。使用肿瘤异种移植模型来测试PFKP在体内的功能。
    结果:在人类黑素瘤组织中观察到PFKP上调,并与患者生存率低相关。人黑色素瘤细胞中PFKP的敲低抑制了细胞增殖并降低了ECAR,ATP水平,和乳酸水平,而PFKP的过表达表现出相反的效果。在体内,在黑素瘤细胞中敲除PFKP显著减少肿瘤发生。对细胞增殖的抑制作用,糖酵解,用糖酵解抑制剂2-脱氧-D-葡萄糖(2-DG)治疗后,由于PFKP敲低引起的肿瘤发生进一步增强。
    结论:总的来说,这些结果表明,PFKP在黑色素瘤细胞中的表达增加了体外增殖和糖酵解活性,并促进了体内肿瘤发生。提示抑制PKFP和抑制糖酵解可能有效抑制黑色素瘤进展。
    OBJECTIVE: Cutaneous melanoma is an aggressive and deadly cancer resulting from malignant transformation of cells involved in skin pigmentation. Glycolysis is widely implicated in cancer progression, but its precise role in melanoma has not been extensively studied. Here, we investigated the role of the glycolysis regulator phosphofructokinase 1 platelet isoform (PFKP) in melanoma progression.
    METHODS: PFKP expression in human melanoma tissues was analyzed by immunohistochemistry. Knockdown of PFKP by siRNA and overexpression of PFKP were performed to evaluate its functions in vitro. CCK-8 assay was used to assess cell proliferation. Glycolytic activity was determined via measurement of extracellular acidification rate (ECAR), lactic acid level, and ATP content. A tumor xenograft model was used to test the function of PFKP in vivo.
    RESULTS: PFKP upregulation was observed in human melanoma tissues and correlated with poor patient survival. Knockdown of PFKP in human melanoma cells suppressed cell proliferation and reduced ECAR, ATP levels, and lactic acid levels, while overexpression of PFKP displayed the opposite effects. In vivo, knockdown of PFKP in melanoma cells markedly reduced tumorigenesis. Inhibitory effects on cell proliferation, glycolysis, and tumorigenesis due to PFKP knockdown were further augmented upon treatment with the glycolysis inhibitor 2-deoxy-D-glucose (2-DG).
    CONCLUSIONS: Collectively, these results indicate that PFKP expression in melanoma cells increases proliferation and glycolytic activity in vitro and promotes tumorigenesis in vivo, suggesting that suppression of PKFP and inhibition of glycolysis may potently suppress melanoma progression.
    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
    揭示胚胎干细胞(ESC)分化为特定谱系的原理对于理解胚胎发育和干细胞在再生医学中的应用至关重要。这里,我们建立了LIF-Stat3信号之间的交集,这对于维持鼠(m)ESCs多能性至关重要,和糖酵解酶,磷酸果糖激酶(Pfkp)的血小板同工型。在多能状态下,Stat3转录抑制mESC中的Pfkp,同时操纵细胞解除这种抑制导致向外胚层谱系分化。Pfkp表现出作为蛋白激酶的底物特异性变化,催化发育调节因子Lin41的丝氨酸磷酸化。这种磷酸化通过阻止Lin41的自泛素化和蛋白酶体降解来稳定Lin41,允许Lin41介导的编码外胚层特化标志物的mRNA的结合和去稳定化,以有利于内胚层特化基因的表达。这为多能性分化电路的布线提供了新的见解,其中Pfkp在mESC分化过程中在胚层规范中起作用。
    Unveiling the principles governing embryonic stem cell (ESC) differentiation into specific lineages is critical for understanding embryonic development and for stem cell applications in regenerative medicine. Here, we establish an intersection between LIF-Stat3 signaling that is essential for maintaining murine (m) ESCs pluripotency, and the glycolytic enzyme, the platelet isoform of phosphofructokinase (Pfkp). In the pluripotent state, Stat3 transcriptionally suppresses Pfkp in mESCs while manipulating the cells to lift this repression results in differentiation towards the ectodermal lineage. Pfkp exhibits substrate specificity changes to act as a protein kinase, catalyzing serine phosphorylation of the developmental regulator Lin41. Such phosphorylation stabilizes Lin41 by impeding its autoubiquitination and proteasomal degradation, permitting Lin41-mediated binding and destabilization of mRNAs encoding ectodermal specification markers to favor the expression of endodermal specification genes. This provides new insights into the wiring of pluripotency-differentiation circuitry where Pfkp plays a role in germ layer specification during mESC differentiation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号