Dual-Specificity Phosphatases

双特异性磷酸酶
  • 文章类型: Journal Article
    肥胖是由脂肪细胞的病理性扩张引起的低度慢性炎症,其允许肥胖相关的代谢疾病如2型糖尿病(T2D)和非酒精性脂肪肝病(NAFLD)的发展。然而,调节脂肪细胞炎症的机制仍然知之甚少。这里,我们观察到TRIM8在脂肪细胞炎症和胰岛素抵抗中上调,而DUSP14下调.TRIM8缺乏和DUSP14过表达降低了炎性细胞因子的水平,葡萄糖摄取含量增加,与单独的LPS治疗相比,改善了胰岛素信号转导。相反,沉默DUSP14可增加炎性细胞因子的表达。它降低了葡萄糖摄取含量和参与胰岛素信号传导的蛋白质的磷酸化水平,进一步损害胰岛素信号和加重胰岛素抵抗。此外,炎症细胞因子水平降低,葡萄糖摄取增加,由TRIM8缺陷引起的胰岛素信号转导改善被下调的DUSP14逆转。总的来说,我们的研究结果表明,TRIM8可以通过调节依赖DUSP14的MAPKs通路来调节脂肪细胞炎症和胰岛素抵抗.
    Obesity is a low-grade chronic inflammation induced by the pathological expansion of adipocytes which allows the development of obesity-associated metabolic diseases like type 2 diabetes mellitus (T2D) and non-alcoholic fatty liver disease (NAFLD). However, mechanisms regulating adipocyte inflammation remain poorly understood. Here, we observed that TRIM8 was upregulated in adipocyte inflammation and insulin resistance while DUSP14 was downregulated. TRIM8 deficiency and DUSP14 over-expression decreased the level of inflammatory cytokines, increased glucose uptake content, and improved insulin signalling transduction compared to LPS treatment alone. Conversely, silencing DUSP14 increased the expression of inflammatory cytokines. It decreased the glucose uptake content and the phosphorylation level of proteins involved in insulin signalling, further impairing insulin signalling and aggravating insulin resistance. Furthermore, The decreased level of inflammatory cytokines, increased glucose uptake, and improved insulin signalling transduction caused by TRIM8 deficiency were reversed by down-regulated DUSP14. Collectively, our findings revealed that TRIM8 can regulate adipocyte inflammation and insulin resistance by regulating the MAPKs pathway which is dependent on DUSP14.
    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
    肿瘤细胞重新编程其代谢以产生专门的代谢产物,这些代谢产物既促进了自身的生长,又允许肿瘤免疫逃避。然而,这些函数之间的关系仍然知之甚少。这里,我们报道了在结肠直肠癌(CRC)小鼠模型中进行的CRISPR筛选,该筛选提示双特异性磷酸酶18(DUSP18)参与了肿瘤定向免疫逃避的建立.Dusp18抑制降低CRC生长速率,这与高水平的CD8+T细胞活化相关。机械上,DUSP18去磷酸化并稳定USF1bHLH-ZIP转录因子。反过来,USF1诱导SREBF2基因,它允许细胞积累胆固醇生物合成中间体羊毛甾醇并将其释放到肿瘤微环境(TME)中。在那里,CD8+T细胞摄取羊毛甾醇抑制甲羟戊酸途径,降低KRAS蛋白戊烯化和功能,这反过来又抑制了它们的激活,并建立了肿瘤细胞免疫逃逸的分子基础。最后,抗PD-1抗体和Lumacaftor的组合,一种FDA批准的DUSP18小分子抑制剂,抑制小鼠CRC生长并协同增强抗肿瘤免疫力。总的来说,我们的研究结果支持免疫检查点和代谢阻断的组合代表了合理设计的,基于机械和潜在的CRC治疗。
    Tumor cells reprogram their metabolism to produce specialized metabolites that both fuel their own growth and license tumor immune evasion. However, the relationships between these functions remain poorly understood. Here, we report CRISPR screens in a mouse model of colo-rectal cancer (CRC) that implicates the dual specificity phosphatase 18 (DUSP18) in the establishment of tumor-directed immune evasion. Dusp18 inhibition reduces CRC growth rates, which correlate with high levels of CD8+ T cell activation. Mechanistically, DUSP18 dephosphorylates and stabilizes the USF1 bHLH-ZIP transcription factor. In turn, USF1 induces the SREBF2 gene, which allows cells to accumulate the cholesterol biosynthesis intermediate lanosterol and release it into the tumor microenvironment (TME). There, lanosterol uptake by CD8+ T cells suppresses the mevalonate pathway and reduces KRAS protein prenylation and function, which in turn inhibits their activation and establishes a molecular basis for tumor cell immune escape. Finally, the combination of an anti-PD-1 antibody and Lumacaftor, an FDA-approved small molecule inhibitor of DUSP18, inhibits CRC growth in mice and synergistically enhances anti-tumor immunity. Collectively, our findings support the idea that a combination of immune checkpoint and metabolic blockade represents a rationally-designed, mechanistically-based and potential therapy for CRC.
    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
    三阴性乳腺癌(TNBC)是乳腺癌的一种亚型,易于发生转移和治疗抵抗。由于其侵略性和有限的靶向治疗,与其他形式的乳腺癌相比,TNBC的死亡率更高。为了开发新的TNBC治疗方案,我们对参与TNBC生长和进展的因素进行了表征。这里,我们证明N-酰基鞘氨醇酰胺水解酶1(ASAH1)在TNBC细胞中过表达,并通过p53和PI3K-AKT信号通路调节。ASAH1的遗传敲除或药理学抑制抑制TNBC的生长和进展。机械上,ASAH1抑制刺激双特异性磷酸酶5(DUSP5)表达,抑制丝裂原活化蛋白激酶(MAPK)途径。此外,ASAH1和MAPK途径的药理学共定位抑制TNBC生长。总的来说,我们揭示了ASAH1在驱动TNBC中的新作用,并确定了ASAH1和MAPK通路的双重靶向是TNBC治疗的潜在新治疗方法.
    Triple-negative breast cancer (TNBC) is a subtype of breast cancer that is prone to metastasis and therapy resistance. Owing to its aggressive nature and limited availability of targeted therapies, TNBC is associated with higher mortality as compared to other forms of breast cancer. In order to develop new therapeutic options for TNBC, we characterized the factors involved in TNBC growth and progression. Here, we demonstrate that N-acylsphingosine amidohydrolase 1 (ASAH1) is overexpressed in TNBC cells and is regulated via p53 and PI3K-AKT signaling pathways. Genetic knockdown or pharmacological inhibition of ASAH1 suppresses TNBC growth and progression. Mechanistically, ASAH1 inhibition stimulates dual-specificity phosphatase 5 (DUSP5) expression, suppressing the mitogen-activated protein kinase (MAPK) pathway. Furthermore, pharmacological cotargeting of the ASAH1 and MAPK pathways inhibits TNBC growth. Collectively, we unmasked a novel role of ASAH1 in driving TNBC and identified dual targeting of the ASAH1 and MAPK pathways as a potential new therapeutic approach for TNBC 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
    据报道,NepetoidinB(NB)具有抗炎作用,抗菌,和抗氧化性能。然而,其对肝脏缺血/再灌注(I/R)损伤的影响尚不清楚。
    在这项研究中,采用小鼠肝脏I/R损伤模型和小鼠AML12细胞缺氧复氧(H/R)损伤模型研究NB的潜在作用。血清转氨酶水平,肝坏死区,细胞活力,氧化应激,炎症反应,并对细胞凋亡进行评估,以评估NB对肝脏I/R和细胞H/R损伤的影响。定量聚合酶链反应(qPCR)和蛋白质印迹用于测量mRNA和蛋白质表达水平,分别。分子对接用于预测NB和丝裂原活化蛋白激酶磷酸酶5(MKP5)的结合能力。
    结果表明,NB显着降低了血清丙氨酸氨基转移酶(ALT)和天冬氨酸氨基转移酶(AST)水平,肝坏死,氧化应激,活性氧(ROS)含量,炎性细胞因子含量和表达,炎性细胞浸润,和肝I/R和AML12细胞H/R损伤后的凋亡。此外,NB抑制JUN蛋白氨基末端激酶(JNK)/P38通路。分子对接结果显示NB与MKP5蛋白结合良好,Westernblotting结果显示NB增加了MKP5的蛋白表达。MKP5敲除(KO)可显著降低NB对肝损伤的保护作用及其对JNK/P38通路的抑制作用。
    NB通过调节MKP5介导的P38/JNK信号通路对肝脏I/R损伤发挥保肝作用。
    UNASSIGNED: Nepetoidin B (NB) has been reported to possess anti-inflammatory, antibacterial, and antioxidant properties. However, its effects on liver ischemia/reperfusion (I/R) injury remain unclear.
    UNASSIGNED: In this study, a mouse liver I/R injury model and a mouse AML12 cell hypoxia reoxygenation (H/R) injury model were used to investigate the potential role of NB. Serum transaminase levels, liver necrotic area, cell viability, oxidative stress, inflammatory response, and apoptosis were evaluated to assess the effects of NB on liver I/R and cell H/R injury. Quantitative polymerase chain reaction (qPCR) and Western blotting were used to measure mRNA and protein expression levels, respectively. Molecular docking was used to predict the binding capacity of NB and mitogen-activated protein kinase phosphatase 5 (MKP5).
    UNASSIGNED: The results showed that NB significantly reduced serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, liver necrosis, oxidative stress, reactive oxygen species (ROS) content, inflammatory cytokine content and expression, inflammatory cell infiltration, and apoptosis after liver I/R and AML12 cells H/R injury. Additionally, NB inhibited the JUN protein amino-terminal kinase (JNK)/P38 pathway. Molecular docking results showed good binding between NB and MKP5 proteins, and Western blotting results showed that NB increased the protein expression of MKP5. MKP5 knockout (KO) significantly diminished the protective effects of NB against liver injury and its inhibitory effects on the JNK/P38 pathway.
    UNASSIGNED: NB exerts hepatoprotective effects against liver I/R injury by regulating the MKP5-mediated P38/JNK 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
    促炎细胞因子的增多和基质中炎性细胞的浸润是IIIA型慢性前列腺炎/慢性盆腔痛综合征(CP/CPPS-A)的重要病理特点,炎症微环境中基质细胞与其他细胞的相互作用与CP/CPPS-A的炎症过程密切相关。然而,基质细胞和上皮细胞之间的相互作用尚不清楚.在这项研究中,炎性前列腺上皮细胞(PECs)释放富含miR-203a-3p的外泌体,并通过上调MCP-1表达促进前列腺基质细胞(PSC)炎症。机械上,DUSP5被鉴定为miR-203a-3p的新靶基因,并通过ERK1/2/MCP-1信号通路调节PSC炎症。同时,CP/CPPS-A患者前列腺液外泌体的作用与炎性PEC外泌体的作用一致.重要的是,我们证明,来自PECs的miR-203a-3pantagomir负载外泌体靶向前列腺,并通过抑制DUSP5-ERK1/2通路缓解前列腺炎.总的来说,我们的发现为CP/CPPS-A中PEC和PSC之间的相互作用提供了新的见解,为CP/CPPS-A提供有希望的治疗策略
    Increased proinflammatory cytokines and infiltration of inflammatory cells in the stroma are important pathological features of type IIIA chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS-A), and the interaction between stromal cells and other cells in the inflammatory microenvironment is closely related to the inflammatory process of CP/CPPS-A. However, the interaction between stromal and epithelial cells remains unclear. In this study, inflammatory prostate epithelial cells (PECs) released miR-203a-3p-rich exosomes and facilitated prostate stromal cells (PSCs) inflammation by upregulating MCP-1 expression. Mechanistically, DUSP5 was identified as a novel target gene of miR-203a-3p and regulated PSCs inflammation through the ERK1/2/MCP-1 signaling pathway. Meanwhile, the effect of exosomes derived from prostatic fluids of CP/CPPS-A patients was consistent with that of exosomes derived from inflammatory PECs. Importantly, we demonstrated that miR-203a-3p antagomirs-loaded exosomes derived from PECs targeted the prostate and alleviated prostatitis by inhibiting the DUSP5-ERK1/2 pathway. Collectively, our findings provide new insights into underlying the interaction between PECs and PSCs in CP/CPPS-A, providing a promising therapeutic strategy for CP/CPPS-A.
    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:细胞周期蛋白依赖性激酶抑制剂蛋白3(CDKN3),作为蛋白激酶家族的一员,已被证明在几种肿瘤中表现出致癌特性。然而,没有对CDKN3的泛致癌分析。
    方法:使用生物信息学工具,如癌症基因组图谱(TCGA)和UCSCXena数据库,对CDKN3进行了全面的泛癌症分析.倒置包括对33种不同类型肿瘤的CDKN3功能的检查,以及基因表达的探索,生存预后状态,临床意义,DNA甲基化,免疫浸润,和相关的信号通路。
    结果:CDKN3在大多数肿瘤中显著上调,并与患者的总生存期(OS)相关。CDKN3的甲基化水平在肿瘤和正常组织之间显著不同。此外,CD4+T细胞浸润,癌症相关成纤维细胞,巨噬细胞,内皮细胞与CDKN3表达相关。机械上,CDKN3与P53、PI3K-AKT、细胞周期检查点,有丝分裂纺锤体检查点,和染色体维持。
    结论:我们在研究中进行的泛癌症分析提供了对CDKN3基因在肿瘤发生中的参与的全面理解。研究结果表明,靶向CDKN3可能会导致治疗肿瘤的新治疗策略。
    BACKGROUND: Cell cycle protein-dependent kinase inhibitor protein 3 (CDKN3), as a member of the protein kinase family, has been demonstrated to exhibit oncogenic properties in several tumors. However, there are no pan-carcinogenic analyses for CDKN3.
    METHODS: Using bioinformatics tools such as The Cancer Genome Atlas (TCGA) and the UCSC Xena database, a comprehensive pan-cancer analysis of CDKN3 was conducted. The inverstigation encompassed the examination of CDKN3 function actoss 33 different kinds of tumors, as well as the exploration of gene expressions, survival prognosis status, clinical significance, DNA methylation, immune infiltration, and associated signal pathways.
    RESULTS: CDKN3 was significantly upregulated in most of tumors and correlated with overall survival (OS) of patients. Methylation levels of CDKN3 differed significantly between tumors and normal tissues. In addition, infiltration of CD4 + T cells, cancer-associated fibroblasts, macrophages, and endothelial cells were associated with CDKN3 expression in various tumors. Mechanistically, CDKN3 was associated with P53, PI3K-AKT, cell cycle checkpoints, mitotic spindle checkpoint, and chromosome maintenance.
    CONCLUSIONS: Our pan-cancer analysis conducted in the study provides a comprehensive understanding of the involvement of CDKN3 gene in tumorigenesis. The findings suggest that targeting CDKN3 may potentially lead to novel therapeutic strategies for the treatment of tumors.
    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
    DUSPs,一组不同的蛋白质磷酸酶,在通过复杂的信号通路协调细胞生长和发育中发挥关键作用。值得注意的是,他们积极参与MAPK通路,控制植物生理学的关键方面,包括增长调节,抗病性,害虫抗性,和应激反应。DUSP是一种关键酶,它是限制细胞代谢速率的酶。目前,对棉花中DUSP基因家族及其在抗黄萎病(VW)中的特定作用的完整理解仍然难以捉摸。为了解决这个知识差距,我们对四个关键棉花品种进行了全面的鉴定和分析:棉花,巴巴多斯棉属,陆地棉,还有灵蒙地羊.结果表明,在四个棉花品种中总共鉴定了120个DUSP基因,分为六个亚组,随机分布在26条染色体的两端,主要位于细胞核内。我们的分析表明,密切相关的DUSP基因在保守的基序组成和基因结构方面表现出相似性。对GhDUSP基因启动子进行的启动子分析显示存在几个顺式作用元件,与非生物和生物应激反应有关,以及激素信号。组织表达模式分析显示在不同胁迫条件下GhDUSP基因表达的显著变化,根表现出最高水平,其次是茎和叶。在组织特异性检测方面,花瓣,叶子,茎,雄蕊,和容器表现出更高的GhDUSP基因表达水平。胁迫下GhDUSP的基因表达分析结果表明,DUSP基因可能在棉花对胁迫的反应中起着至关重要的作用。通过病毒诱导的基因沉默(VIGS)实验,目的基因的沉默显著降低了抗病品种对黄萎病(VW)的抗性效率。因此,我们得出的结论是,GH_A11G3500介导的双特异性磷酸化基因可能是陆地红对黄萎病(VW)抗性的关键调节因子。本研究提出了一个全面的结构,旨在深入了解棉花的潜在生物学功能,为进一步研究分子育种和植物病原体抗性提供了坚实的基础。
    DUSPs, a diverse group of protein phosphatases, play a pivotal role in orchestrating cellular growth and development through intricate signaling pathways. Notably, they actively participate in the MAPK pathway, which governs crucial aspects of plant physiology, including growth regulation, disease resistance, pest resistance, and stress response. DUSP is a key enzyme, and it is the enzyme that limits the rate of cell metabolism. At present, complete understanding of the DUSP gene family in cotton and its specific roles in resistance to Verticillium wilt (VW) remains elusive. To address this knowledge gap, we conducted a comprehensive identification and analysis of four key cotton species: Gossypium arboreum, Gossypium barbadense, Gossypium hirsutum, and Gossypium raimondii. The results revealed the identification of a total of 120 DUSP genes in the four cotton varieties, which were categorized into six subgroups and randomly distributed at both ends of 26 chromosomes, predominantly localized within the nucleus. Our analysis demonstrated that closely related DUSP genes exhibited similarities in terms of the conserved motif composition and gene structure. A promoter analysis performed on the GhDUSP gene promoter revealed the presence of several cis-acting elements, which are associated with abiotic and biotic stress responses, as well as hormone signaling. A tissue expression pattern analysis demonstrated significant variations in GhDUSP gene expression under different stress conditions, with roots exhibiting the highest levels, followed by stems and leaves. In terms of tissue-specific detection, petals, leaves, stems, stamens, and receptacles exhibited higher expression levels of the GhDUSP gene. The gene expression analysis results for GhDUSPs under stress suggest that DUSP genes may have a crucial role in the cotton response to stress in cotton. Through Virus-Induced Gene Silencing (VIGS) experiments, the silencing of the target gene significantly reduced the resistance efficiency of disease-resistant varieties against Verticillium wilt (VW). Consequently, we conclude that GH_A11G3500-mediated bispecific phosphorylated genes may serve as key regulators in the resistance of G. hirsutum to Verticillium wilt (VW). This study presents a comprehensive structure designed to provide an in-depth understanding of the potential biological functions of cotton, providing a strong foundation for further research into molecular breeding and resistance to plant pathogens.
    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
    Lafora病(LD)是一种常染色体隐性遗传性肌阵挛性癫痫,在青少年时期发作,导致发病十年内死亡。LD的特征是过度积累的过度磷酸化,分枝差,不溶性,糖原样聚合物称为Lafora体。这种疾病是由EPM2A的突变引起的,编码拉福林,一种去磷酸化糖原的双特异性磷酸酶,或EMP2B,编码马林,E3-泛素连接酶。虽然糖原是一种被广泛接受的laforin底物,malin的底物难以鉴定,部分原因是缺乏能够在体内检测到malin的malin抗体。在这里,我们描述了一个小鼠模型,其中malin基因在C端被修饰以包含c-myc标签序列,使malin-myc的表达易于检测。使用c-myc标签抗体对免疫沉淀物进行的质谱分析表明,Malin与Laforin和许多糖原代谢酶相互作用。为了研究laforin在这些相互作用中的作用,我们分析了另外两种小鼠模型:malin-myc/laforin敲除和malin-myc/LaforinCS,其中laforin不存在或催化Cys基因组突变为Ser,分别。Malin与伴侣蛋白的相互作用需要Laforin,但不依赖于其催化活性或糖原的存在。总的来说,结果表明,拉福林和马林在体内形成复合物,它能稳定苹果酸并增强与伴侣蛋白的相互作用,以促进正常的糖原代谢。他们还提供了有关LD发展和通过催化失活磷酸酶挽救疾病的见解。
    Lafora disease (LD) is an autosomal recessive myoclonus epilepsy with onset in the teenage years leading to death within a decade of onset. LD is characterized by the overaccumulation of hyperphosphorylated, poorly branched, insoluble, glycogen-like polymers called Lafora bodies. The disease is caused by mutations in either EPM2A, encoding laforin, a dual specificity phosphatase that dephosphorylates glycogen, or EMP2B, encoding malin, an E3-ubiquitin ligase. While glycogen is a widely accepted laforin substrate, substrates for malin have been difficult to identify partly due to the lack of malin antibodies able to detect malin in vivo. Here we describe a mouse model in which the malin gene is modified at the C-terminus to contain the c-myc tag sequence, making an expression of malin-myc readily detectable. Mass spectrometry analyses of immunoprecipitates using c-myc tag antibodies demonstrate that malin interacts with laforin and several glycogen-metabolizing enzymes. To investigate the role of laforin in these interactions we analyzed two additional mouse models: malin-myc/laforin knockout and malin-myc/LaforinCS, where laforin was either absent or the catalytic Cys was genomically mutated to Ser, respectively. The interaction of malin with partner proteins requires laforin but is not dependent on its catalytic activity or the presence of glycogen. Overall, the results demonstrate that laforin and malin form a complex in vivo, which stabilizes malin and enhances interaction with partner proteins to facilitate normal glycogen metabolism. They also provide insights into the development of LD and the rescue of the disease by the catalytically inactive phosphatase.
    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
    癌细胞采用适应性机制来生存各种压力源,包括基因毒性药物.了解促进生存的因素对于开发有效的治疗方法至关重要。在这项研究中,我们揭示了一个以前未被探索的长的非编码RNA,JUNI(JUN-DT,LINC01135),它通过激活应激激活的MAPK被基因毒性药物上调,JNK,和p38,因此对其相邻基因产物c-Jun的表达施加阳性控制,一种众所周知的癌蛋白,它将信号转换为多个转录输出。JUNI调节细胞迁移并在赋予细胞对化疗药物或紫外线辐射的抗性方面具有关键作用。JUNI的消耗显着增加了培养细胞和球体对化学治疗剂的敏感性。我们鉴定了57种与JUNI相互作用的蛋白质。其中一种MAPK磷酸酶和抑制剂的活性,DUSP14,由JUNI抵消,因此,当细胞暴露于紫外线辐射时,促进有效的JNK磷酸化和c-Jun诱导。与DUSP14的拮抗相互作用不仅有助于c-Jun诱导,而且还增加了紫外线照射细胞的存活。总之,我们引入JUNI作为一种新型的c-Jun应激诱导调节剂,将其定位为增强癌细胞对化疗敏感性的潜在靶标。
    Cancer cells employ adaptive mechanisms to survive various stressors, including genotoxic drugs. Understanding the factors promoting survival is crucial for developing effective treatments. In this study, we unveil a previously unexplored long non-coding RNA, JUNI (JUN-DT, LINC01135), which is upregulated by genotoxic drugs through the activation of stress-activated MAPKs, JNK, and p38 and consequently exerts positive control over the expression of its adjacent gene product c-Jun, a well-known oncoprotein, which transduces signals to multiple transcriptional outputs. JUNI regulates cellular migration and has a crucial role in conferring cellular resistance to chemotherapeutic drugs or UV radiation. Depletion of JUNI markedly increases the sensitivity of cultured cells and spheroids to chemotherapeutic agents. We identified 57 proteins interacting with JUNI. The activity of one of them the MAPK phosphatase and inhibitor, DUSP14, is counteracted by JUNI, thereby, facilitating efficient JNK phosphorylation and c-Jun induction when cells are exposed to UV radiation. The antagonistic interplay with DUSP14 contributes not only to c-Jun induction but also augments the survival of UV-exposed cells. In summary, we introduce JUNI as a novel stress-inducible regulator of c-Jun, positioning it as a potential target for enhancing the sensitivity of cancer cells to chemotherapy.
    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
    内皮-间质转化(EndMT)是内皮细胞失去其内皮特性并获得间充质特性的过程。双特异性蛋白磷酸酶22(DUSP22)通过使丝氨酸/苏氨酸残基脱磷酸化而使各种蛋白激酶和转录因子失活:因此,它在许多疾病中起着关键作用。本研究旨在探讨DUSP22在EndMT中的功能作用。在转化生长因子β诱导的人脐静脉内皮细胞(HUVECs)EndMT模型中,我们观察到DUSP22表达下调。这种DUSP22缺陷可能会加重EndMT。相反,DUSP22的过表达可以改善EndMT。我们使用信号通路抑制剂来验证我们的结果,发现DUSP22可以通过smad2/3和丝裂原活化蛋白激酶(MAPK)信号通路调节EndMT。总之,DUSP22通过smad2/3和MAPK信号通路改善体外HUVECs中的EndMT。
    Endothelial-to-mesenchymal transition (EndMT) is the process by which endothelial cells lose their endothelial properties and acquire mesenchymal characteristics. Dual-specific protein phosphatase 22 (DUSP22) inactivates various protein kinases and transcription factors by dephosphorylating serine/threonine residues: hence, it plays a key role in many diseases. The aim of this study was to explore the functional role of DUSP22 in EndMT. In the transforming growth factor-β-induced EndMT model in human umbilical vein endothelial cells (HUVECs), we observed a downregulation of DUSP22 expression. This DUSP22 deficiency could aggravate EndMT. Conversely, the overexpression of DUSP22 could ameliorate EndMT. We used signaling pathway inhibitors to verify our results and found that DUSP22 could regulate EndMT through the smad2/3 and the mitogen-activated protein kinase (MAPK) signaling pathways. In summary, DUSP22 ameliorates EndMT in HUVECs in vitro through the smad2/3 and MAPK signaling pathways.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号