Sp1 Transcription Factor

Sp1 转录因子
  • 文章类型: Journal Article
    食管鳞状细胞癌(ESCC)的预后和治疗效果较差。代谢失调有助于多种癌症的无限制生长。然而,代谢异常,如高度活化的磷酸戊糖途径(PPP)在ESCC进展中的作用仍在很大程度上未知.在这里,我们报道了高移动性组AT-hook1(HMGA1),一种参与染色质重塑的结构转录因子,通过提高PPP水平促进了ESCC的发展。我们发现HMGA1在ESCC中高度表达。升高的HMGA1促进ESCC细胞的恶性表型。HMGA1的条件性敲除显着降低了4-硝基喹啉-1-氧化物(4NQO)诱导的小鼠食管肿瘤发生。通过代谢组学分析和验证试验,我们发现HMGA1上调非氧化性PPP。通过转录组测序,我们确定HMGA1上调转酮醇酶(TKT)的表达,催化非氧化性PPP中的可逆反应,以糖酵解途径交换代谢产物。HMGA1敲低通过下调TKT抑制PPP,导致ESCC细胞中核苷酸的减少。HMGA1的过表达上调PPP并通过激活TKT促进ESCC细胞的存活。我们进一步表征HMGA1通过与转录因子SP1相互作用并增强其与TKT启动子的结合来促进TKT的转录。用抑制剂靶向TKT的治疗学,氧硫胺素,降低HMGA1诱导的ESCC细胞增殖和肿瘤生长。一起,在这项研究中,通过上调TKT介导的PPP激活,我们发现HMGA1在ESCCs中的新作用.我们的研究结果为HMGA1/TKT/PPP在ESCC肿瘤发生和靶向治疗中的作用提供了新的见解。
    Esophageal squamous cell carcinoma (ESCC) possesses a poor prognosis and treatment outcome. Dysregulated metabolism contributes to unrestricted growth of multiple cancers. However, abnormal metabolism, such as highly activated pentose phosphate pathway (PPP) in the progression of ESCC remains largely unknown. Herein, we report that high-mobility group AT-hook 1 (HMGA1), a structural transcriptional factor involved in chromatin remodeling, promoted the development of ESCC by upregulating the PPP. We found that HMGA1 was highly expressed in ESCC. Elevated HMGA1 promoted the malignant phenotype of ESCC cells. Conditional knockout of HMGA1 markedly reduced 4-nitroquinoline-1-oxide (4NQO)-induced esophageal tumorigenesis in mice. Through the metabolomic analysis and the validation assay, we found that HMGA1 upregulated the non-oxidative PPP. With the transcriptome sequencing, we identified that HMGA1 upregulated the expression of transketolase (TKT), which catalyzes the reversible reaction in non-oxidative PPP to exchange metabolites with glycolytic pathway. HMGA1 knockdown suppressed the PPP by downregulating TKT, resulting in the reduction of nucleotides in ESCC cells. Overexpression of HMGA1 upregulated PPP and promoted the survival of ESCC cells by activating TKT. We further characterized that HMGA1 promoted the transcription of TKT by interacting with and enhancing the binding of transcription factor SP1 to the promoter of TKT. Therapeutics targeting TKT with an inhibitor, oxythiamine, reduced HMGA1-induced ESCC cell proliferation and tumor growth. Together, in this study, we identified a new role of HMGA1 in ESCCs by upregulating TKT-mediated activation of PPP. Our results provided a new insight into the role of HMGA1/TKT/PPP in ESCC tumorigenesis and targeted therapy.
    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
    在哺乳动物中,特异性蛋白1(SP1)是特异性蛋白和Krüppel样因子(Sp/KLF)基因家族中第一个分离出的Cys2-His2锌指转录因子。SP1通过与靶基因启动子区上富含鸟嘌呤-胞嘧啶(GC)的序列结合来调节基因表达,影响各种细胞过程。此外,SP1的活性受到翻译后修饰的显著影响,比如磷酸化,乙酰化,糖基化,和蛋白水解。SP1与细胞凋亡的调节有关,细胞肥大,炎症,氧化应激,脂质代谢,斑块稳定,内皮功能障碍,纤维化,钙化,和其他病理过程。这些过程影响许多心血管疾病的发生和进展,包括冠心病,缺血再灌注损伤,心肌病,心律失常,血管疾病。SP1成为心脏病预防和治疗干预的潜在目标。在这次审查中,我们深入研究生物学功能,病理生理机制,以及SP1在心脏病理学中的潜在临床意义,为SP1在心脏病中的调节功能提供有价值的见解,并揭示预防和治疗心血管疾病的新途径。
    In mammals, specificity protein 1 (SP1) was the first Cys2-His2 zinc finger transcription factor to be isolated within the specificity protein and Krüppel-like factor (Sp/KLF) gene family. SP1 regulates gene expression by binding to Guanine-Cytosine (GC)-rich sequences on promoter regions of target genes, affecting various cellular processes. Additionally, the activity of SP1 is markedly influenced by posttranslational modifications, such as phosphorylation, acetylation, glycosylation, and proteolysis. SP1 is implicated in the regulation of apoptosis, cell hypertrophy, inflammation, oxidative stress, lipid metabolism, plaque stabilization, endothelial dysfunction, fibrosis, calcification, and other pathological processes. These processes impact the onset and progression of numerous cardiovascular disorders, including coronary heart disease, ischemia-reperfusion injury, cardiomyopathy, arrhythmia, and vascular disease. SP1 emerges as a potential target for the prevention and therapeutic intervention of cardiac ailments. In this review, we delve into the biological functions, pathophysiological mechanisms, and potential clinical implications of SP1 in cardiac pathology to offer valuable insights into the regulatory functions of SP1 in heart diseases and unveil novel avenues for the prevention and treatment of cardiovascular conditions.
    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
    肿瘤微环境中的外泌体microRNAs(miRNAs)通过参与细胞间的串扰在肿瘤发生和肿瘤进展中发挥关键作用。然而,外泌体miRNAs的功能及其调节食管鳞状细胞癌(ESCC)进展的机制尚不清楚.
    进行RNA测序和GEO分析以鉴定参与ESCC发育的候选外泌体miRNA。采用受试者工作特征曲线分析评估血浆外泌体miR-493-5p的诊断价值。EdU,管形成和Transwell分析用于研究外泌体miR-493-5p对人脐静脉内皮细胞(HUVEC)的影响。使用皮下异种移植模型来评估miR-493-5p和地西他滨(DNA甲基转移酶抑制剂)的抗肿瘤作用。miR-493-5p和SP1/SP3之间的关系通过双荧光素酶报告基因测定来揭示。随后进行一系列拯救测定以研究SP1/SP3是否参与外泌体miR-493-5p介导的ESCC血管生成。
    我们发现miR-493-5p表达在ESCC患者的血浆外泌体中显著降低,在早期ESCC诊断中具有很高的潜在价值。此外,miR-493-5p,作为候选肿瘤抑制剂,抑制增殖,通过抑制VEGFA的表达,并通过外泌体发挥其血管抑制作用。此外,我们发现SP1/SP3是miR-493-5p的直接靶标,并且SP1/SP3的再表达可以逆转miR-493-5p的抑制作用.进一步研究发现miR-493-5p表达受DNA甲基转移酶3A(DNMT3A)和DNMT3B调控,miR-493-5p过表达或地西他滨恢复miR-493-5p表达均可增加贝伐单抗的抗肿瘤作用.
    外泌体miR-493-5p是一种非常有价值的ESCC诊断标志物,抑制ESCC相关血管生成。miR-493-5p可以通过DNA甲基化沉默,和miR-493-5p表达的恢复与地西他滨增加贝伐单抗的抗肿瘤作用,提示其作为ESCC治疗靶点的潜力。
    UNASSIGNED: Exosomal microRNAs (miRNAs) in the tumor microenvironment play crucial roles in tumorigenesis and tumor progression by participating in intercellular cross-talk. However, the functions of exosomal miRNAs and the mechanisms by which they regulate esophageal squamous cell carcinoma (ESCC) progression are unclear.
    UNASSIGNED: RNA sequencing and GEO analysis were conducted to identify candidate exosomal miRNAs involved in ESCC development. Receiver operating characteristic curve analysis was performed to assess the diagnostic value of plasma exosomal miR-493-5p. EdU, tube formation and Transwell assays were used to investigate the effects of exosomal miR-493-5p on human umbilical vein endothelial cells (HUVECs). A subcutaneous xenograft model was used to evaluate the antitumor effects of miR-493-5p and decitabine (a DNA methyltransferase inhibitor). The relationship between miR-493-5p and SP1/SP3 was revealed via a dual-luciferase reporter assay. A series of rescue assays were subsequently performed to investigate whether SP1/SP3 participate in exosomal miR-493-5p-mediated ESCC angiogenesis.
    UNASSIGNED: We found that miR-493-5p expression was notably reduced in the plasma exosomes of ESCC patients, which showed the high potential value in early ESCC diagnosis. Additionally, miR-493-5p, as a candidate tumor suppressor, inhibited the proliferation, migration and tube formation of HUVECs by suppressing the expression of VEGFA and exerted its angiostatic effect via exosomes. Moreover, we found that SP1/SP3 are direct targets of miR-493-5p and that re-expression of SP1/SP3 could reverse the inhibitory effects of miR-493-5p. Further investigation revealed that miR-493-5p expression could be regulated by DNA methyltransferase 3A (DNMT3A) and DNMT3B, and either miR-493-5p overexpression or restoration of miR-493-5p expression with decitabine increased the antitumor effects of bevacizumab.
    UNASSIGNED: Exosomal miR-493-5p is a highly valuable ESCC diagnosis marker and inhibits ESCC-associated angiogenesis. miR-493-5p can be silenced via DNA methylation, and restoration of miR-493-5p expression with decitabine increases the antitumor effects of bevacizumab, suggesting its potential as a therapeutic target for ESCC 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:乳腺癌是女性中最致命的癌症之一。尽管在乳腺癌的诊断和治疗方面取得了重大进展,许多患者仍然屈服于这种疾病,因此,迫切需要新的有效治疗方法。天然产物香豆素已被广泛研究,因为它揭示了医药领域的各种生物学特性。越来越多的证据表明,组蛋白去乙酰化酶抑制剂(HDACIs)是有前途的新型抗乳腺癌药物。然而,目前的大多数HDACIs对实体肿瘤仅表现出中等效果,并伴有严重的副作用。因此,开发更有效的乳腺癌治疗HDACIs,HDACIs的异羟肟酸盐与香豆素核心有关,并设计合成了香豆素-异羟肟酸盐杂种。
    方法:通过药效团融合策略将取代的香豆素部分掺入到经典的异羟肟酸盐HDACIs中。通过使用HDACI筛选试剂盒和细胞活力测定鉴定ZN444B。进行分子对接以探索ZN444B与HDAC1的结合模式。蛋白质印迹,免疫荧光染色,细胞活力,使用集落形成和细胞迁移以及流式细胞术测定来分析ZN444B的体外抗乳腺癌作用。将小鼠模型中的原位研究用于体内功效和毒性的临床前评估。蛋白质组学分析,双荧光素酶报告分析,染色质免疫沉淀,免疫共沉淀,免疫荧光染色分析和免疫组织化学(IHC)分析用于阐明ZN444B作用的分子基础。
    结果:我们合成并鉴定了一种新的香豆素-异羟肟酸盐偶联物,ZN444B在体外和体内均具有有希望的抗乳腺癌活性。分子对接模型显示ZN444B以高亲和力结合HDAC1。进一步的机理研究表明,ZN444B通过抑制HDAC1在K703对Sp1的脱乙酰酶活性而特异性降低FOS样抗原2(FOSL2)mRNA水平,并消除Sp1与FOSL2启动子的结合能力。此外,FOSL2表达与乳腺癌进展和转移呈正相关。沉默FOSL2表达降低了乳腺癌细胞对ZN444B治疗的敏感性。此外,ZN444B在小鼠中没有显示全身毒性。
    结论:我们的发现强调了FOSL2作为乳腺癌新的生物标志物和治疗靶点的潜力,并且用ZN444B靶向HDAC1-Sp1-FOSL2信号轴可能是一种有希望的乳腺癌治疗策略。
    BACKGROUND: Breast cancer is one of the most lethal cancers in women. Despite significant advances in the diagnosis and treatment of breast cancer, many patients still succumb to this disease, and thus, novel effective treatments are urgently needed. Natural product coumarin has been broadly investigated since it reveals various biological properties in the medicinal field. Accumulating evidence indicates that histone deacetylase inhibitors (HDACIs) are promising novel anti-breast cancer agents. However, most current HDACIs exhibit only moderate effects against solid tumors and are associated with severe side effects. Thus, to develop more effective HDACIs for breast cancer therapy, hydroxamate of HDACIs was linked to coumarin core, and coumarin-hydroxamate hybrids were designed and synthesized.
    METHODS: A substituted coumarin moiety was incorporated into the classic hydroxamate HDACIs by the pharmacophore fusion strategy. ZN444B was identified by using the HDACI screening kit and cell viability assay. Molecular docking was performed to explore the binding mode of ZN444B with HDAC1. Western blot, immunofluorescent staining, cell viability, colony formation and cell migration and flow cytometry assays were used to analyze the anti-breast cancer effects of ZN444B in vitro. Orthotopic studies in mouse models were applied for preclinical evaluation of efficacy and toxicity in vivo. Proteomic analysis, dual-luciferase reporter assay, chromatin immunoprecipitation, co-immunoprecipitation, immunofluorescent staining assays along with immunohistochemical (IHC) analysis were used to elucidate the molecular basis of the actions of ZN444B.
    RESULTS: We synthesized and identified a novel coumarin-hydroxamate conjugate, ZN444B which possesses promising anti-breast cancer activity both in vitro and in vivo. A molecular docking model showed that ZN444B binds to HDAC1 with high affinity. Further mechanistic studies revealed that ZN444B specifically decreases FOS-like antigen 2 (FOSL2) mRNA levels by inhibiting the deacetylase activity of HDAC1 on Sp1 at K703 and abrogates the binding ability of Sp1 to the FOSL2 promoter. Furthermore, FOSL2 expression positively correlates with breast cancer progression and metastasis. Silencing FOSL2 expression decreases the sensitivity of breast cancer cells to ZN444B treatment. In addition, ZN444B shows no systemic toxicity in mice.
    CONCLUSIONS: Our findings highlight the potential of FOSL2 as a new biomarker and therapeutic target for breast cancer and that targeting the HDAC1-Sp1-FOSL2 signaling axis with ZN444B may be a promising therapeutic strategy for breast cancer.
    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:骨质疏松症(OP)的进展可以显着增加骨折的风险,这严重扰乱了老年人的生活。特异性蛋白1(SP1)参与OP进程。然而,SP1调节OP进展的机制尚不清楚.
    目的:本研究探讨了SP1在OP中的作用机制。
    方法:用SAMP6小鼠建立年龄依赖性OP的体内模型,和BALB/c小鼠用作对照。从两种亚型小鼠中提取BMSCs。进行苏木精和伊红染色以标记髓内骨小梁结构以评估组织学变化。ChIP测定用于评估SP1和miR-133a-3p之间的靶向调节。MAPK3和miR-133a-3p之间的结合位点使用双荧光素酶报告物测定来验证。使用定量逆转录聚合酶链反应(RT-qPCR)检测miR-133a-3p和MAPK3的mRNA水平。SP1、MAPK3、Colla1、OCN、使用Western印迹检查Runx2。碱性磷酸酶(ALP)试剂盒和茜素红S染色用于研究ALP活性和矿化结节,分别。
    结果:SP1和miR-133a-3p水平上调,而SAMP6小鼠的BMSCs中MAPK3的表达下调,miR-133a-3p抑制剂可加速BMSCs的成骨分化。SP1直接靶向miR-133a-3p,MAPK3是miR-133a-3p的下游mRNA。机械上,SP1通过miR-133a-3p/MAPK3轴的转录介导加速BMSCs成骨分化。
    结论:SP1通过介导miR-133a-3p/MAPK3轴调节成骨分化,这将为治疗老年OP的策略提供新的思路。
    BACKGROUND: The progression of osteoporosis (OP) can dramatically increase the risk of fractures, which seriously disturb the life of elderly individuals. Specific protein 1 (SP1) is involved in OP progression. However, the mechanism by which SP1 regulates OP progression remains unclear.
    OBJECTIVE: This study investigated the mechanism underlying the function of SP1 in OP.
    METHODS: SAMP6 mice were used to establish an in vivo model of age-dependent OP, and BALB/c mice were used as controls. BMSCs were extracted from two subtypes of mice. Hematoxylin and eosin staining were performed to mark the intramedullary trabecular bone structure to evaluate histological changes. ChIP assay was used to assess the targeted regulation between SP1 and miR-133a-3p. The binding sites between MAPK3 and miR-133a-3p were verified using a dual-luciferase reporter assay. The mRNA levels of miR-133a-3p and MAPK3 were detected using quantitative reverse transcription polymerase chain reaction (RT-qPCR). The protein expression of SP1, MAPK3, Colla1, OCN, and Runx2 was examined using Western blotting. Alkaline phosphatase (ALP) kit and Alizarin Red S staining were used to investigate ALP activity and mineralized nodules, respectively.
    RESULTS: The levels of SP1 and miR-133a-3p were upregulated, whereas the expression of MAPK3 was downregulated in BMSCs from SAMP6 mice, and miR-133a-3p inhibitor accelerated osteogenic differentiation in BMSCs. SP1 directly targeted miR-133a-3p, and MAPK3 was the downstream mRNA of miR-133a-3p. Mechanically, SP1 accelerated osteogenic differentiation in BMSCs via transcriptional mediation of the miR-133a-3p/MAPK3 axis.
    CONCLUSIONS: SP1 regulates osteogenic differentiation by mediating the miR-133a-3p/MAPK3 axis, which would shed new light on strategies for treating senile OP.
    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
    肺腺癌(LUAD)是全球癌症相关死亡的主要原因,但潜在的分子机制仍不清楚。转录因子(TF)特异性蛋白1(SP1)在各种癌症的发生发展中起着至关重要的作用,包括LUAD.最近的研究表明,主TF可以形成相分离的大分子缩合物,以促进超增强子(SE)组装和癌基因表达。在这项研究中,我们证明了SP1经历相分离,其DNA结合域中的锌指3对于该过程至关重要。通过使用针对SP1和H3K27ac的抗体,使用核酸酶(CUT和RUN)在靶标下裂解和释放,我们发现SP1富集和SE元素之间存在显著的相关性,确定G蛋白信号传导20(RGS20)基因的调节因子是SP1通过SE机制调节的最可能的靶标,并使用不同的方法验证了这一发现。SP1的致癌活性依赖于其相分离能力和RGS20基因激活,糖原合成酶激酶J4(GSK-J4)可以消除,去甲基酶抑制剂.一起,我们的发现提供了证据,表明SP1通过相分离和SE机制调节其靶癌基因表达,从而促进LUAD细胞进展。这项研究还揭示了通过干预SP1介导的SE形成来进行LUAD治疗的创新靶标。
    Lung adenocarcinoma (LUAD) is the leading cause of cancer-related death worldwide, but the underlying molecular mechanisms remain largely unclear. The transcription factor (TF) specificity protein 1 (SP1) plays a crucial role in the development of various cancers, including LUAD. Recent studies have indicated that master TFs may form phase-separated macromolecular condensates to promote super-enhancer (SE) assembly and oncogene expression. In this study, we demonstrated that SP1 undergoes phase separation and that its zinc finger 3 in the DNA-binding domain is essential for this process. Through Cleavage Under Targets & Release Using Nuclease (CUT&RUN) using antibodies against SP1 and H3K27ac, we found a significant correlation between SP1 enrichment and SE elements, identified the regulator of the G protein signaling 20 (RGS20) gene as the most likely target regulated by SP1 through SE mechanisms, and verified this finding using different approaches. The oncogenic activity of SP1 relies on its phase separation ability and RGS20 gene activation, which can be abolished by glycogen synthase kinase J4 (GSK-J4), a demethylase inhibitor. Together, our findings provide evidence that SP1 regulates its target oncogene expression through phase separation and SE mechanisms, thereby promoting LUAD cell progression. This study also revealed an innovative target for LUAD therapies through intervening in SP1-mediated SE formation.
    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:糖尿病性白内障(DC)是糖尿病的常见并发症,其病因和进展是多因素的。在这项研究中,探讨了特异性蛋白1(SP1)和成纤维细胞生长因子7(FGF7)在DC发育中的作用。
    方法:用高糖处理SRA01/04细胞建立DC细胞模型。进行MTT测定以评估细胞活力。进行Transwell测定和伤口愈合测定以评估细胞迁移和侵袭。Westernblot和qRT-PCR检测N-cadherin的表达,E-cadherin,胶原蛋白I,纤连蛋白,SP1和FGF7表达。进行CHIP测定和双荧光素酶报告基因测定以分析FGF7和SP1之间的组合。
    结果:FGF7在DC患者和HG诱导的SRA01/04细胞中上调。HG处理促进SRA01/04细胞活力,迁移,侵袭和上皮间质转化(EMT),而FGF7敲除减轻了影响。转录因子SP1激活了FGF7的转录水平,SP1过表达加重了HG诱导的SRA01/04细胞损伤。SP1沉默抑制了HG诱导的SRA01/04细胞活力,迁移,入侵和EMT,但这些影响通过上调FGF7得到改善。此外,SP1敲低通过调节FGF7的转录水平抑制PI3K/AKT通路。
    结论:转录因子SP1激活FGF7的转录水平和PI3K/AKT通路,调节HG诱导的SRA01/04细胞活力,迁移,入侵和EMT。
    BACKGROUND: Diabetic cataract (DC) is a common complication of diabetes and its etiology and progression are multi-factorial. In this study, the roles of specific protein 1 (SP1) and fibroblast growth factor 7 (FGF7) in DC development were explored.
    METHODS: DC cell model was established by treating SRA01/04 cells with high glucose (HG). MTT assay was conducted to evaluate cell viability. Transwell assay and wound-healing assay were performed to assess cell migration and invasion. Western blot assay and qRT-PCR assay were conducted to measure the expression of N-cadherin, E-cadherin, Collagen I, Fibronectin, SP1 and FGF7 expression. CHIP assay and dual-luciferase reporter assay were conducted to analyze the combination between FGF7 and SP1.
    RESULTS: FGF7 was upregulated in DC patients and HG-induced SRA01/04 cells. HG treatment promoted SRA01/04 cell viability, migration, invasion and epithelial-mesenchymal transition (EMT), while FGF7 knockdown abated the effects. Transcription factor SP1 activated the transcription level of FGF7 and SP1 overexpression aggravated HG-induced SRA01/04 cell injury. SP1 silencing repressed HG-induced SRA01/04 cell viability, migration, invasion and EMT, but these effects were ameliorated by upregulating FGF7. Additionally, SP1 knockdown inhibited the PI3K/AKT pathway by regulating the transcription level of FGF7.
    CONCLUSIONS: Transcription factor SP1 activated the transcription level of FGF7 and the PI3K/AKT pathway to regulate HG-induced SRA01/04 cell viability, migration, invasion and EMT.
    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

       PDF(Pubmed)

  • 文章类型: Journal Article
    胃癌(GC)是全球第五大最常见的癌症和第四大原发性癌症相关死亡率。作为第一个鉴定的用于去除RNA甲基化修饰的m6A去甲基酶,脂肪量和肥胖相关蛋白(FTO)在癌症发展中起着重要作用。因此,我们研究了FTO在GC肿瘤发生和发展中的生物学功能和致癌机制。在我们的研究中,FTO表达在GC组织和细胞中明显上调。FTO的上调与晚期神经侵犯有关,肿瘤大小,还有LNM,以及GC患者的不良预后,并促进GC细胞活力,菌落形成,移民和入侵。机械上,FTO靶向特异性蛋白1和Aurora激酶B,导致共济失调毛细血管扩张症的磷酸化突变和P38和P53的去磷酸化。总之,m6A去甲基酶FTO通过调节SP1-AURKB-ATM途径促进GC肿瘤发生和进展,这可能凸显了FTO作为GC患者治疗反应和预后的诊断生物标志物的潜力。
    Gastric cancer (GC) is the 5th most prevalent cancer and the 4th primary cancer-associated mortality globally. As the first identified m6A demethylase for removing RNA methylation modification, fat mass and obesity-associated protein (FTO) plays instrumental roles in cancer development. Therefore, we study the biological functions and oncogenic mechanisms of FTO in GC tumorigenesis and progression. In our study, FTO expression is obviously upregulated in GC tissues and cells. The upregulation of FTO is associated with advanced nerve invasion, tumor size, and LNM, as well as the poor prognosis in GC patients, and promoted GC cell viability, colony formation, migration and invasion. Mechanistically, FTO targeted specificity protein 1 and Aurora Kinase B, resulting in the phosphorylation of ataxia telangiectasia mutated and P38 and dephosphorylation of P53. In conclusion, the m6A demethylase FTO promotes GC tumorigenesis and progression by regulating the SP1-AURKB-ATM pathway, which may highlight the potential of FTO as a diagnostic biomarker for GC patients\' therapy response and prognosis.
    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
    复发性高级别胶质瘤(rHGs)预后不佳,其中IVterameprocol的最大耐受剂量(MTD)(5天/月),特异性蛋白1(Sp1)调节蛋白的转录抑制剂,为1,700mg/天,血浆浓度-时间曲线下的中位面积(AUC)为31.3μg*h/mL。鉴于持续的全身暴露和每日静脉治疗的挑战性后勤可能会增加疗效,在这里,我们研究多中心口服terameprocol治疗rHGs,第一阶段试验(GATOR)。使用3+3剂量递增设计,我们招募了20名患者,中位年龄60岁(范围31-80岁),70%男性,和中位数1次复发(范围1-3)。空腹患者耐受1,200毫克/天(n=3),2,400毫克/天(n=6),3,600毫克/天(n=3),和6,000毫克/天(n=2)口服剂量,无主要毒性。然而,增加剂量不会导致全身暴露增加,包括在美联储州(6,000毫克/天,n=4),最大AUC<5μg*h/mL。这些发现保证了研究提供持续全身水平的转录抑制剂以利用其治疗潜力的方法的试验。本研究在ClinicalTrials.gov(NCT02575794)注册。
    Recurrent high-grade gliomas (rHGGs) have a dismal prognosis, where the maximum tolerated dose (MTD) of IV terameprocol (5 days/month), a transcriptional inhibitor of specificity protein 1 (Sp1)-regulated proteins, is 1,700 mg/day with median area under the plasma concentration-time curve (AUC) of 31.3 μg∗h/mL. Given potentially increased efficacy with sustained systemic exposure and challenging logistics of daily IV therapy, here we investigate oral terameprocol for rHGGs in a multicenter, phase 1 trial (GATOR). Using a 3 + 3 dose-escalation design, we enroll 20 patients, with median age 60 years (range 31-80), 70% male, and median one relapse (range 1-3). Fasting patients tolerate 1,200 mg/day (n = 3), 2,400 mg/day (n = 6), 3,600 mg/day (n = 3), and 6,000 mg/day (n = 2) oral doses without major toxicities. However, increased dosage does not lead to increased systemic exposure, including in fed state (6,000 mg/day, n = 4), with maximal AUC <5 μg∗h/mL. These findings warrant trials investigating approaches that provide sustained systemic levels of transcription inhibitors to exploit their therapeutic potential. This study was registered at ClinicalTrials.gov (NCT02575794).
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号