FGF1

FGF1
  • 文章类型: Journal Article
    富含脯氨酸/精氨酸的末端和富含亮氨酸的蛋白(PRELP)被鉴定为细胞外基质中的小蛋白聚糖,与细胞粘附密切相关。目前,PRELP在结直肠癌(CRC)中的作用尚不清楚.通过qRT-PCR和免疫化学分析人CRC组织样品中的PRELP表达。CCK-8,集落形成,transwell,和试管形成试验用于确定PRELP对CRC细胞恶性表型的影响。构建小鼠异种移植和肿瘤转移模型以进一步验证PRELP的功能。此外,我们研究了PRELP联合贝伐单抗在CRC小鼠异种移植模型中的疗效.此外,进行RNA-seq以分析由PRELP调节的潜在信号传导途径。进行免疫荧光染色和共免疫沉淀以确认PRELP和成纤维细胞生长因子1(FGF1)之间的相互作用。在这项研究中,我们发现PRELP对CRC具有肿瘤抑制作用.PRELP在CRC组织和细胞系中的表达水平显著降低。体内和体外实验均证实PRELP抑制CRC细胞增殖,促进细胞凋亡,并通过减少上皮-间质转化和减弱血管生成来抑制迁移和侵袭,从而抑制肿瘤进展。此外,PRELP显著增强贝伐单抗在小鼠异种移植模型中的功效。机械上,PRELP与FGF1结合并降低FGF1蛋白的稳定性,伴随着它退化的增加,随后灭活PI3K/AKT/mTOR途径,从而导致肿瘤血管生成和转移的减少。我们的研究首次揭示了PRELP在CRC中的肿瘤抑制作用,并为CRC的治疗提供了潜在的有效策略。
    Proline/arginine-rich end and leucine-rich protein (PRELP) is identified as a small proteoglycan in the extracellular matrix that has been tightly associated with cell adhesion. At present, the role of PRELP in colorectal cancer (CRC) remains largely unknown. PRELP expression in human CRC tissue samples was analyzed by qRT-PCR and immunochemistry. CCK-8, colony formation, transwell, and tube formation assays were utilized to determine the influences of PRELP on the malignant phenotypes of CRC cells. Mouse xenograft and tumor metastasis models were constructed to further validate the function of PRELP. Furthermore, we investigated the efficacy of PRELP combined with bevacizumab treatment in a mouse xenograft model of CRC. Additionally, RNA-seq was performed to analyze the potential signaling pathways regulated by PRELP. Immunofluorescence staining and coimmunoprecipitation were conducted to confirm the interaction between PRELP and fibroblast growth factor 1 (FGF1). In this study, we found that PRELP exerted a tumor-suppressive effect on CRC. The expression level of PRELP was significantly reduced in CRC tissues and cell lines. Both in vivo and in vitro experiments confirmed that PRELP inhibited CRC cell proliferation, promoted apoptosis, and suppressed migration and invasion via a reduction in the epithelial-mesenchymal transition and attenuated angiogenesis, thereby dampening tumor progression. In addition, PRELP markedly potentiated the efficacy of bevacizumab in a mouse xenograft model. Mechanistically, PRELP bound to FGF1 and reduced the stability of the FGF1 protein, accompanied by an increase in its degradation, which subsequently inactivated the PI3K/AKT/mTOR pathway, thereby leading to reduction in tumor angiogenesis and metastasis. Our study for the first time unveiled the tumor-suppressive role of PRELP in CRC and provided a potential effective strategy for the treatment of 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
    几种炎性细胞因子结合到变构位点(位点2)并变构激活整联蛋白。位点2也是25-羟基胆固醇的结合位点,炎症脂质介质,并参与炎症信号传导(例如,TNF和IL-6分泌)以及整联蛋白激活。FGF2是促炎和促血栓形成的,与FGF2同源的FGF1具有抗炎和抗血栓形成作用,但是这些作用的机制是未知的。我们假设FGF2和FGF1与整合素的位点2结合并调节炎症信号传导。这里,我们描述了FGF2与位点2和变构激活的β3整合素结合,这表明FGF2的促炎作用是通过与位点2结合来介导的。相比之下,FGF1与位点2结合,但不激活这些整联蛋白,而是抑制了FGF2诱导的整联蛋白激活,表明FGF1充当位点2的拮抗剂,并且FGF1的抗炎作用由阻断位点2介导。非促有丝分裂的FGF1突变体(R50E),与αvβ3的位点1结合有缺陷,与WTFGF1一样有效地抑制了FGF2对β3整联蛋白的激活。
    Several inflammatory cytokines bind to the allosteric site (site 2) and allosterically activate integrins. Site 2 is also a binding site for 25-hydroxycholesterol, an inflammatory lipid mediator, and is involved in inflammatory signaling (e.g., TNF and IL-6 secretion) in addition to integrin activation. FGF2 is pro-inflammatory and pro-thrombotic, and FGF1, homologous to FGF2, has anti-inflammatory and anti-thrombotic actions, but the mechanism of these actions is unknown. We hypothesized that FGF2 and FGF1 bind to site 2 of integrins and regulate inflammatory signaling. Here, we describe that FGF2 is bound to site 2 and allosterically activated β3 integrins, suggesting that the pro-inflammatory action of FGF2 is mediated by binding to site 2. In contrast, FGF1 bound to site 2 but did not activate these integrins and instead suppressed integrin activation induced by FGF2, indicating that FGF1 acts as an antagonist of site 2 and that the anti-inflammatory action of FGF1 is mediated by blocking site 2. A non-mitogenic FGF1 mutant (R50E), which is defective in binding to site 1 of αvβ3, suppressed β3 integrin activation by FGF2 as effectively as WT FGF1.
    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
    血管生成与肿瘤微环境重塑和免疫治疗反应显著相关。我们的研究旨在构建胃癌的预后血管生成相关模型。使用公共数据库,血管生成相关的五基因(FGF1,GRB14,PAK3,PDGFRA,和PRKD1)进行了鉴定。前25%的患者被定义为高风险,其余为低风险。1-的曲线下面积,3-,5年总生存率(OS)分别为0.646,0.711和0.793.生存分析显示,低风险患者在构建(HR=0.57,p=0.002)和验证队列中有更好的10年OS。GO和GSEA揭示了DEGs在细胞外基质受体相互作用中的富集,树突状细胞抗原加工/呈递调节,和血管生成途径。CIBERSORT分析显示丰富的原始B细胞,静息的肥大细胞,静息CD4+记忆T细胞,M2巨噬细胞,和单核细胞在高风险亚组。TIMER数据库显示PAK3,FGF1,PRKD1和PDGFRA表达水平与CD4T细胞和巨噬细胞浸润之间存在强正相关。IOBR分析显示,高危亚组存在免疫抑制环境。低风险患者对抗PD1治疗的反应率较高。TMA显示FGF1过表达与不良预后及CD4+T细胞和巨噬细胞浸润有关。基于615小鼠的体内研究表明,抑制FGF1功能可以抑制肿瘤生长并增强抗PD1治疗功效。总之,我们建立了5项血管生成相关基因模型来预测胃癌患者的生存结局和免疫治疗反应,并将FGF1确定为改善免疫治疗的预后基因和潜在靶点.
    Angiogenesis significantly correlates with tumor microenvironment remodeling and immunotherapy response. Our study aimed to construct a prognostic angiogenesis-related model for gastric cancer. Using public database, a angiogenetic related five-gene (FGF1, GRB14, PAK3, PDGFRA, and PRKD1) model was identified. The top 25 % of patients were defined as high-risk, and the remaining as low-risk. The area under the curve for 1-, 3-, and 5-year overall survival (OS) were 0.646, 0.711, and 0.793, respectively. Survival analysis showed a better 10-year OS in low-risk patients in the construction (HR = 0.57, p = 0.002) and validation cohorts. GO and GSEA revealed that DEGs were enriched in extracellular matrix receptor interactions, dendritic cell antigen processing/presentation regulation, and angiogenesis pathways. CIBERSORT analysis revealed abundant naïve B cells, resting mast cells, resting CD4+ memory T cells, M2 macrophages, and monocytes in high-risk subgroups. The TIMER database showed strong positive correlations between PAK3, FGF1, PRKD1, and PDGFRA expression levels and the infiltration of CD4+ T cells and macrophages. The IOBR analysis revealed an immunosuppressive environment in the high-risk subgroup. Low-risk patients show a higher response rate to anti-PD1 treatment. TMA showed that FGF1 overexpression was associated with poor prognosis and CD4+ T cells and macrophage infiltration. In vivo study based on the 615 mice indicated that inhibiting FGF1 function could suppress tumor growth and enhance anti-PD1 therapeutic efficacy. In summary, we established a five-angiogenesis-related gene model to predict survival outcomes and immunotherapy responses in patients with gastric cancer and identified FGF1 as a prognostic gene and potential target for improving immune 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
    目的:脓毒症,一种全球流行和高度致命的疾病,仍然是一个关键的医学挑战。本研究旨在评估FGF1作为脓毒症潜在治疗靶点的相关性。
    方法:采用LPS诱导C57BL/6小鼠脓毒症,建立体内动物模型。使用人脐静脉内皮细胞进行各种体外测定,以阐明FGF1在与脓毒症相关的凝血系统破坏和肝损伤中的作用。以及探索其潜在的分子机制。
    结果:在体内实验中,FGF1通过降低血液中促炎和凝血相关因子的水平改善脓毒症小鼠的凝血系统破坏。FGF1还增强了败血症小鼠的肝功能,减轻肝脏炎症和细胞凋亡,促进肝脏血管再生,增加肝脏血液灌注,提高老鼠的存活率。体外实验证明FGF1能够抑制LPS诱导的内皮细胞炎症反应和凋亡,强化内皮细胞屏障功能,降低内皮细胞通透性,促进内皮细胞增殖,恢复内皮细胞的成管能力。体内和体外实验均证实FGF1通过抑制IL-6/STAT3信号通路改善脓毒症。
    结论:总之,我们的研究表明,FGF1通过抑制IL-6/STAT3信号通路减轻脓毒症的过度炎症反应,从而改善全身血液循环和改善败血症生物体的肝损伤。因此,本研究将FGF1确定为治疗人类脓毒症的潜在临床靶点.
    OBJECTIVE: Sepsis, a globally prevalent and highly lethal condition, remains a critical medical challenge. This investigation aims to assess the relevance of FGF1 as a potential therapeutic target for sepsis.
    METHODS: Sepsis was induced in C57BL/6 mice through LPS administration to establish an in vivo animal model. Various in vitro assays were conducted using human umbilical vein endothelial cells to elucidate the role of FGF1 in the disruption of the coagulation system and liver injury associated with sepsis, as well as to explore its underlying molecular mechanisms.
    RESULTS: In in vivo experiments, FGF1 ameliorated coagulation system disruption in septic mice by reducing the levels of pro-inflammatory and coagulation-related factors in the bloodstream. FGF1 also enhanced liver function in septic mice, mitigating liver inflammation and cell apoptosis, fostering liver vascular regeneration, increasing liver blood perfusion, and improving mouse survival. In vitro experiments demonstrated that FGF1 could inhibit LPS-induced inflammatory responses and apoptosis in endothelial cells, fortify endothelial cell barrier function, decrease endothelial cell permeability, promote endothelial cell proliferation, and restore endothelial cell tube-forming ability. Both in vivo and in vitro experiments substantiated that FGF1 improved sepsis by inhibiting the IL-6/STAT3 signaling pathway.
    CONCLUSIONS: In summary, our study indicates that FGF1 mitigates excessive inflammatory responses in sepsis by suppressing the IL-6/STAT3 signaling pathway, thereby improving systemic blood circulation and ameliorating liver damage in septic organisms. Consequently, this research identifies FGF1 as a potential clinical target for the treatment of human sepsis.
    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
    Mule鸭倾向于通过喂食在肝脏中积累丰富的脂肪,导致形成比正常肝脏大几倍的脂肪肝。然而,脂肪肝形成的潜在机制尚未阐明。成纤维细胞生长因子1(FGF1),FGF超家族的一员,参与细胞脂质代谢和有丝分裂。本研究旨在探讨FGF1对复合脂肪酸诱导的乳鸭肝细胞脂代谢紊乱的调控作用及其分子机制。通过添加浓度为1,500:750μmol/L的油酸和棕榈酸持续36小时来诱导肝细胞,用浓度为0、10、100和1000ng/mL的FGF1刺激12h。结果表明,FGF1显着降低了复合脂肪酸诱导的肝脂滴沉积和甘油三酯含量;它还降低了氧化应激;降低了活性氧荧光强度和丙二醛含量;上调了抗氧化因子核因子红细胞2相关因子2(Nrf2)的表达,HO-1和NQO-1;显着增强肝细胞活性;促进细胞周期进程;抑制细胞凋亡;上调细胞周期依赖性激酶1(CDK1)和BCL-2mRNA表达;并下调Bax和Caspase-3表达。此外,FGF1促进AMPK磷酸化,激活了AMPK通路,上调AMPK基因表达,下调SREBP1和ACC1基因的表达,从而减轻复杂脂肪酸诱导的肝细胞脂肪过度积累。总之,FGF1可能通过激活AMPK信号通路缓解复合脂肪酸诱导的乳鸭肝细胞脂代谢紊乱。
    Mule ducks tend to accumulate abundant fat in their livers via feeding, which leads to the formation of a fatty liver that is several times larger than a normal liver. However, the mechanism underlying fatty liver formation has not yet been elucidated. Fibroblast growth factor 1 (FGF1), a member of the FGF superfamily, is involved in cellular lipid metabolism and mitosis. This study aims to investigate the regulatory effect of FGF1 on lipid metabolism disorders induced by complex fatty acids in primary mule duck liver cells and elucidate the underlying molecular mechanism. Hepatocytes were induced by adding 1,500:750 µmol/L oleic and palmitic acid concentrations for 36 h, which were stimulated with FGF1 concentrations of 0, 10, 100, and 1000 ng/mL for 12 h. The results showed that FGF1 significantly reduced the hepatic lipid droplet deposition and triglyceride content induced by complex fatty acids; it also reduced oxidative stress; decreased reactive oxygen species fluorescence intensity and malondialdehyde content; upregulated the expression of antioxidant factors nuclear factor erythroid 2 related factor 2 (Nrf2), HO-1, and NQO-1; significantly enhanced liver cell activity; promoted cell cycle progression; inhibited cell apoptosis; upregulated cyclin-dependent kinase 1 (CDK1) and BCL-2 mRNA expression; and downregulated Bax and Caspase-3 expression. In addition, FGF1 promoted AMPK phosphorylation, activated the AMPK pathway, upregulated AMPK gene expression, and downregulated the expression of SREBP1 and ACC1 genes, thereby alleviating excessive fat accumulation in liver cells induced by complex fatty acids. In summary, FGF1 may alleviate lipid metabolism disorders induced by complex fatty acids in primary mule duck liver cells by activating the AMPK signaling pathway.
    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
    血管肉瘤是一种罕见的难治性软组织肿瘤,预后差,可通过放射治疗进行治疗。成纤维细胞生长因子1(FGF1)突变体,由于几个取代的氨基酸,具有增强的热稳定性,抑制血管肉瘤细胞转移,然而,作用机制尚不清楚。本研究旨在阐明使用ISOS-1小鼠血管肉瘤细胞的FGF1突变体的作用机制。将野生型FGF1或FGF1突变体添加到ISOS-1细胞中进行培养,随着时间的推移评估细胞数量。通过transwell分析评估ISOS-1细胞的侵袭和迁移能力。通过X射线照射后的集落形成测定来评估ISOS-1细胞的放射敏感性。为了检查丝裂原活化蛋白激酶(MEK)抑制剂是否抵消FGF1突变体的作用,将MEK抑制剂和FGF1突变体的组合加入ISOS-1细胞并培养.观察到FGF1突变体抑制ISOS-1细胞增殖,通过持续的FGF1信号激活进行入侵和迁移。MEK抑制剂抑制FGF1突变体诱导的增殖抑制,ISOS-1细胞的侵袭和迁移。此外,FGF1突变体增强了ISOS-1细胞的放射敏感性,但是MEK抑制抑制了放射敏感性的增加。此外,我们发现FGF1突变体强烈抑制肌动蛋白聚合,提示肌动蛋白细胞骨架动力学与ISOS-1细胞放射敏感性密切相关。总的来说,这项研究表明,在ISOS-1细胞中,FGF1突变体抑制增殖,侵袭和迁移,同时通过持续激活MEK介导的信号通路增强放射敏感性。
    Angiosarcoma is a rare refractory soft-tissue tumor with a poor prognosis and is treated by radiotherapy. The fibroblast growth factor 1 (FGF1) mutant, with enhanced thermostability due to several substituted amino acids, inhibits angiosarcoma cell metastasis, yet the mechanism of action is unclear. This study aims to clarify the FGF1 mutant mechanism of action using ISOS-1 mouse angiosarcoma cells. The wild-type FGF1 or FGF1 mutant was added to ISOS-1 cells and cultured, evaluating cell numbers over time. The invasive and migratory capacity of ISOS-1 cells was assessed by transwell analysis. ISOS-1 cell radiosensitivity was assessed by colony formation assay after X-ray irradiation. To examine whether mitogen-activated protein kinase (MEK) inhibitor counteracts the FGF1 mutant effects, a combination of MEK inhibitor and FGF1 mutant was added to ISOS-1 cells and cultured. The FGF1 mutant was observed to inhibit ISOS-1 cell proliferation, invasion and migration by sustained FGF1 signaling activation. A MEK inhibitor suppressed the FGF1 mutant-induced inhibition of proliferation, invasion and migration of ISOS-1 cells. Furthermore, the FGF1 mutant enhanced radiosensitivity of ISOS-1 cells, but MEK inhibition suppressed the increased radiosensitivity. In addition, we found that the FGF1 mutant strongly inhibits actin polymerization, suggesting that actin cytoskeletal dynamics are closely related to ISOS-1 cell radiosensitivity. Overall, this study demonstrated that in ISOS-1 cells, the FGF1 mutant inhibits proliferation, invasion and migration while enhancing radiosensitivity through sustained activation of the MEK-mediated signaling pathway.
    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
    几种药物引起的直接肾小管损伤,尤其是化疗药物,是AKI的常见原因。细胞周期蛋白依赖性激酶12(CDK12)的抑制或缺失引发转录延伸缺陷,导致DNA损伤修复缺陷,在各种癌症中产生基因组不稳定性。值得注意的是,10-25%的人在用CDK12抑制剂治疗后发生AKI,潜在的机制还不清楚。这里,我们发现,在AKI患者和小鼠AKI模型中,CDK12在肾小管上皮细胞中下调.此外,小鼠中CDK12的肾小管细胞特异性敲低通过促进基因组不稳定性增强顺铂诱导的AKI,凋亡,和增殖抑制,而CDK12过表达保护抗AKI。在CDK12RTEC+/-小鼠的肾脏上使用单分子实时(SMRT)平台,我们发现CDK12敲低通过转录延伸缺陷靶向Fgf1和Cast,从而增强基因组的不稳定性和细胞凋亡。总的来说,这些数据表明,CDK12敲低可以通过改变Fgf1和Cast基因的转录延伸缺陷来增强AKI的发展,对CDK12抑制剂治疗的患者应给予更多的关注以预防AKI。
    Direct tubular injury caused by several medications, especially chemotherapeutic drugs, is a common cause of AKI. Inhibition or loss of cyclin-dependent kinase 12 (CDK12) triggers a transcriptional elongation defect that results in deficiencies in DNA damage repair, producing genomic instability in a variety of cancers. Notably, 10-25% of individuals developed AKI after treatment with a CDK12 inhibitor, and the potential mechanism is not well understood. Here, we found that CDK12 was downregulated in the renal tubular epithelial cells in both patients with AKI and murine AKI models. Moreover, tubular cell-specific knockdown of CDK12 in mice enhanced cisplatin-induced AKI through promotion of genome instability, apoptosis, and proliferative inhibition, whereas CDK12 overexpression protected against AKI. Using the single molecule real-time (SMRT) platform on the kidneys of CDK12RTEC+/- mice, we found that CDK12 knockdown targeted Fgf1 and Cast through transcriptional elongation defects, thereby enhancing genome instability and apoptosis. Overall, these data demonstrated that CDK12 knockdown could potentiate the development of AKI by altering the transcriptional elongation defect of the Fgf1 and Cast genes, and more attention should be given to patients treated with CDK12 inhibitors to prevent AKI.
    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:糖尿病足溃疡(DFU)对糖尿病患者构成重大健康风险,伤口愈合过程中血运重建不足是主要原因。这项研究旨在使用血管内皮生长因子(VEGF-A)和改良的成纤维细胞生长因子(FGF1)评估微血管发芽和伤口愈合能力。
    方法:在糖尿病小鼠中使用离体主动脉环啮齿动物模型和体内伤口愈合模型来评估VEGF-A和修饰的FGF1作为单一疗法和组合的微血管出芽和伤口愈合能力。
    结果:在离体小鼠主动脉环模型中,VEGF-A和FGF1的组合显示血管出芽增加,在小鼠皮肤伤口中局部施用在脂质纳米颗粒(LNP)中配制的VEGF-A和FGFlmRNAs的组合促进了更快的伤口闭合和在手术伤口产生后7天增加的新血管形成。在伤口形成后第3天的皮肤样本的RNA测序分析揭示了伤口愈合过程的强烈转录反应,联合治疗显示与皮肤生长相关的基因显著富集。
    结论:封装VEGF-A和FGF1mRNA的f-LNP为改善DFU的瘢痕形成过程提供了一种有希望的方法。
    BACKGROUND: Diabetic foot ulcers (DFU) pose a significant health risk in diabetic patients, with insufficient revascularization during wound healing being the primary cause. This study aimed to assess microvessel sprouting and wound healing capabilities using vascular endothelial growth factor (VEGF-A) and a modified fibroblast growth factor (FGF1).
    METHODS: An ex vivo aortic ring rodent model and an in vivo wound healing model in diabetic mice were employed to evaluate the microvessel sprouting and wound healing capabilities of VEGF-A and a modified FGF1 both as monotherapies and in combination.
    RESULTS: The combination of VEGF-A and FGF1 demonstrated increased vascular sprouting in the ex vivo mouse aortic ring model, and topical administration of a combination of VEGF-A and FGF1 mRNAs formulated in lipid nanoparticles (LNPs) in mouse skin wounds promoted faster wound closure and increased neovascularization seven days post-surgical wound creation. RNA-sequencing analysis of skin samples at day three post-wound creation revealed a strong transcriptional response of the wound healing process, with the combined treatment showing significant enrichment of genes linked to skin growth.
    CONCLUSIONS: f-LNPs encapsulating VEGF-A and FGF1 mRNAs present a promising approach to improving the scarring process in DFU.
    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
    成纤维细胞生长因子(FGFs)是具有广泛生物学活性的蛋白质,如细胞发育和修复,葡萄糖和胆汁酸代谢,伤口愈合。由于其关键和多样的生理功能,FGF被认为具有作为许多需要进一步研究的疾病和病症的治疗剂的潜力。因此,一个简单的,需要成本有效的方法来纯化这些生物活性信号蛋白。在这里,我们引入此类技术来纯化具有高肝素结合亲和力或低至无肝素结合亲和力的FGF.该方法利用对来自旁分泌FGF1的硫酸肝素的高亲和力来分离目标蛋白。它也解释了FGF成员具有低肝素亲和力,比如代谢FGFs,通过在重组蛋白中引入多组氨酸标签结合固定化金属亲和层析。随后,通过高速离心将纯化的FGF产物与其它小蛋白分离。然后对产品进行其他生物物理实验,如SDS-PAGE,质谱,圆二色性,固有荧光,等温滴定量热法,差示扫描量热法,和生物细胞活性测定,证实纯化的目的蛋白具有完整的天然构象,内在特征和生物活性没有明显变化。
    Fibroblast growth factors (FGFs) are proteins with a vast array of biological activity, such as cell development and repair, glucose and bile acid metabolisms, and wound healing. Due to their critical and diverse physiological functions, FGFs are believed to possess potential as therapeutic agents for many diseases and conditions that warrant further investigations. Thus, a simple, cost-efficient method to purify these biologically active signaling proteins is desirable. Herein, we introduce such techniques to purify FGFs that possess either high heparin-binding affinity or low to no heparin-binding affinity. This method takes advantage of the high affinity toward heparin sulfate from paracrine FGF1 to isolate the targeted protein. It also accounts for FGF members that have low heparin affinity, such as the metabolic FGFs, by introducing poly-histidine tags in the recombinant protein in combination with the immobilized metal affinity chromatography. Subsequently, the purified FGF products are separated from the other small protein by high-speed centrifugation. Products are then subjected to other biophysical experiments like SDS-PAGE, mass spectrometry, circular dichroism, intrinsic fluorescence, isothermal titration calorimetry, differential scanning calorimetry, and biological cell activity assay to confirm that the target proteins are purified with intact native conformation and no significant change in the intrinsic characteristics and biological activities.
    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
    了解CRLM的分子网络是一个正在进行的研究领域。在这项研究中,对15例非转移性CRC患者的配对CRC组织和邻近非癌组织,15例CRLM患者的配对CRC组织和匹配的肝转移组织及其邻近非癌组织进行了评估.我们评估了Rap1通路相关基因,包括NRAS,FGF-1,NGF,qRT-PCR和KDR表达及其蛋白状态的Westernblot。在CRLM患者中,NRAS,FGF1和KDRmRNA和蛋白在转移性肿瘤中的表达水平高于CRC原发肿瘤和邻近非癌组织(p<0.05)。在非转移性患者中,NRAS,FGF1,KDR,和NGF基因表达在CRC原发肿瘤组织和邻近非癌组织之间没有差异(p>0.05)。ROC曲线分析显示NRAS诊断的准确性,FGF1,KDR,和FGF用于在分析原发性肿瘤时区分转移性患者和非转移性患者。数据表明,需要对Rap1相关基因在CRLM中的作用进行进一步的功能研究。总之,目前的数据拓宽了我们对CRLM特定分子特征的认识。增加对转移的分子特征的了解有可能创造更成功的治疗方法。或预防,转移,尤其是在多模式原发肿瘤治疗中。
    Understanding molecular networks of CRLM is an ongoing area of research. In this study, paired CRC tissue and adjacent noncancerous tissue from 15 non-metastatic CRC patients and paired CRC tissue and matched liver metastatic tissues from 15 CRLM patients along with their adjacent noncancerous tissues were evaluated. We assessed Rap1 pathway-related genes including NRAS, FGF-1, NGF, and KDR expression by qRT-PCR and their protein status by Western blot. In CRLM patients, NRAS, FGF1, and KDR mRNA and protein were expressed at higher levels in metastatic than in CRC primary tumor and adjacent noncancerous tissue (p < 0.05). In non-metastatic patients, NRAS, FGF1, KDR, and NGF gene expression did not differ between CRC primary tumor-and adjacent noncancerous tissue (p > 0.05). ROC curve analysis showed a reasonable diagnostic accuracy of NRAS, FGF1, KDR, and FGF for the discrimination of metastatic patients from non- metastatic ones on analysis of their primary tumors. The data suggest that further functional studies on Rap1-related genes\' role in CRLM are needed. In conclusion, the present data broaden our knowledge about specific molecular characteristics of CRLM. An increased understanding of the molecular features of metastasis has the potential to create more successful treatment, or prevention, of metastasis, especially in multimodal primary tumor treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号