Kruppel-Like Transcription Factors

Kruppel 样转录因子
  • 文章类型: Journal Article
    在这项研究中,我们旨在探讨Krüppel样因子7(KLF7)在结直肠癌(CRC)细胞侵袭和迁移中的分子机制。分析KLF7在CRC组织中的表达模式以及KLF7表达与CRC临床症状的相关性。用si-KLF7转染CRC细胞系,然后qRT-PCR或蛋白质印迹检测KLF7,miR-139-5p,和肿瘤蛋白D52(TPD52)表达,细胞计数试剂盒-8(CCK-8)检测细胞活力,以及Transwell入侵和迁移的检测。染色质免疫沉淀(ChIP)分析了miR-139-5p启动子中的富集KLF7。双荧光素酶报告基因实验验证了KLF7与miR-139-5p的结合关系,以及miR-139-5p和TPD52之间。在皮下肿瘤发生实验中,观察肿瘤生长,检测到ki67阳性表达.KLF7在CRC细胞中大量表达KLF7沉默抑制CRC细胞活力,入侵,和移民。KLF7通过结合miR-139-5p启动子抑制miR-139-5p表达。miR-139-5p靶向TPD52表达。miR-13-5p抑制或TPD52过表达部分抵消了CRC细胞中KLF7沉默的影响。KLF7沉默在体内抑制肿瘤生长。总之,KLF7通过与miR-139-5p启动子结合抑制miR-139-5p表达,从而上调TPD52表达并增强CRC细胞侵袭和迁移。
    In this study, we aimed to investigate the molecular mechanism of Krüppel-like factor 7 (KLF7) in colorectal cancer (CRC) cell invasion and migration. The expression pattern of KLF7 in CRC tissues and the correlation between KLF7 expression and clinical symptoms of CRC were analyzed. CRC cell lines were transfected with si-KLF7, followed by qRT-PCR or western blot detection of KLF7, miR-139-5p, and tumor protein D52 (TPD52) expression, cell counting kit-8 (CCK-8) assay to detect cell viability, and transwell detection of invasion and migration. Chromatin immunoprecipitation (ChIP) analyzed the enrichment KLF7 in the miR-139-5p promoter. The dual-luciferase reporter assay verified the binding relationship between KLF7 and miR-139-5p, and between miR-139-5p and TPD52. In the subcutaneous tumorigenesis experiment, tumor growth was observed and ki67-positive expression was detected. KLF7 is abundantly expressed in CRC cells KLF7 silencing inhibits CRC cell viability, invasion, and migration. KLF7 represses miR-139-5p expression by binding to the miR-139-5p promoter. miR-139-5p targets TPD52 expression. miR-13-5p inhibition or TPD52 overexpression partially counteracted the effect of KLF7 silencing in CRC cells. KLF7 silencing suppresses tumor growth in vivo. In conclusion, KLF7 suppresses miR-139-5p expression by binding to the miR-139-5p promoter, thereby upregulating TPD52 expression and enhancing CRC cell invasion and migration.
    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:TGF-β基因是吉西他滨(GEM)抗性基因;然而,其调节胰腺癌GEM耐药的机制尚不清楚.
    方法:用GEM处理PANC-1细胞系,然后用TGF-β刺激。随后,我们构建了抗GEM的胰腺癌细胞系,在这些细胞系中敲除TGF-β,并检测耐药癌细胞增殖和凋亡的变化。此外,测定KLF-4、GFI-1和ZEB-1的蛋白表达水平。通过将GEM抗性PANC-1细胞皮下注射到小鼠腋下,构建了裸鼠异种移植瘤模型。肿瘤被切除了,解剖,6周后称重。定量肿瘤组织中KLF-4、GFI-1和ZEB-1的蛋白质水平。此外,使用流式细胞术确定肿瘤组织中M2巨噬细胞的百分比。
    结果:GEM处理后,胰腺癌细胞中TGF-β的蛋白水平明显下降。KLF-4蛋白表达下调,而TGF-β刺激后GFI-1和ZEB-1的表达上调。TGF-β敲低GEM耐药胰腺癌细胞凋亡增加,增殖减少,此外,沉默TGF-β可促进Caspase3和cleavedcaspase3的表达。此外,KLF-4蛋白表达上调,而GFI-1和ZEB-1的表达下调。Further,TGF-β敲除后,移植瘤的体积和重量降低。KLF-4蛋白表达上调,而GFI-1和ZEB-1在肿瘤组织中表达下调。此外,TGF-β敲除后,肿瘤组织中M2巨噬细胞的百分比降低。
    结论:TGF-β的敲低抑制上皮-间质转化,抑制耐药癌细胞的增殖并促进其凋亡,并减少巨噬细胞向M2表型的极化,因此改善胰腺癌的GEM耐药性。
    BACKGROUND: The TGF-β gene is a gemcitabine (GEM) resistance gene; however, the mechanism by which it regulates GEM resistance in pancreatic cancer remains unclear.
    METHODS: The PANC-1 cell line was treated with GEM and then stimulated with TGF-β. Subsequently, we constructed GEM-resistant pancreatic cancer cell lines, knocked down TGF-β in these cell lines, and detected changes in the proliferation and apoptosis of drug-resistant cancer cells. In addition, the protein expression levels of KLF-4, GFI-1, and ZEB-1 were determined. The xenograft tumor models of nude mice were constructed by subcutaneously injecting GEM-resistant PANC-1 cells into mouse axilla. The tumors were removed, dissected, and weighed after 6 weeks. The protein levels of KLF-4, GFI-1, and ZEB-1 in tumor tissues were quantified. In addition, the percentage of M2 macrophages in tumor tissues was determined using flow cytometry.
    RESULTS: The protein levels of TGF-β in pancreatic cancer cells were significantly decreased after GEM treatment. The protein expression of KLF-4 was downregulated, whereas the expressions of GFI-1 and ZEB-1 were upregulated after TGF-β stimulation. Apoptosis increased and proliferation decreased after TGF-β knockdown in GEM-resistant pancreatic cancer cells, moreover, silencing TGF-β promoted the expression of Caspase 3 and Cleaved caspase 3. In addition, the protein expression of KLF-4 was upregulated, whereas the expressions of GFI-1 and ZEB-1 were downregulated. Further, the volume and weight of the transplanted tumor decreased after TGF-β knockdown. The protein expression of KLF-4 was upregulated, whereas the expressions of GFI-1 and ZEB-1 were downregulated in tumor tissues. In addition, the percentage of M2 macrophages decreased in tumor tissues after TGF-β knockdown.
    CONCLUSIONS: The knockdown of TGF-β inhibits epithelial-to-mesenchymal transition, suppresses the proliferation and promotes the apoptosis of drug-resistant cancer cells, and decreases the macrophage polarization to the M2 phenotype, consequently ameliorating GEM resistance in pancreatic cancer.
    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
    这项研究的目的是通过研究槲皮素对血管平滑肌细胞(VSMC)向巨噬细胞样细胞的表型转换和潜在的调节途径的影响来阐明槲皮素在动脉粥样硬化中的保护作用。高脂饮食(HFD)喂养的载脂蛋白E缺陷(ApoEKO)小鼠的主动脉组织,用或不用100mg/kg/天槲皮素治疗,分析了组织病理学变化和分子机制。发现槲皮素可减少动脉粥样硬化病变的大小并减轻HFD引起的脂质积累。荧光共定位分析显示与磷酸-Janus激酶2(p-JAK2)共定位的巨噬细胞样血管平滑肌细胞(VSMC)的存在更高,磷酸信号转导和转录激活因子3(p-STAT3),和Krüppel样因子4(KLF4)在主动脉斑块内的泡沫细胞聚集区域。然而,这种共定位在槲皮素治疗后减少.槲皮素治疗可有效抑制KLF4介导的氧化低密度脂蛋白(ox-LDL)负载小鼠主动脉血管平滑肌细胞(MOVAS)的表型转换,如KLF4,LGALS3,CD68和F4/80的表达降低,α平滑肌肌动蛋白(α-SMA)的表达增加,减少细胞内荧光Dil-ox-LDL摄取,和减少脂质积累。相比之下,APTO-253,一种KLF4激活剂,被发现可以逆转槲皮素的作用。此外,AG490,一种JAK2抑制剂,有效抵消ox-LDL诱导的JAK2/STAT3途径依赖性转换为巨噬细胞样表型和MOVAS细胞中的脂质积累。槲皮素可显着减轻这些作用,但JAK2激活剂CoumermycinA1可加剧这些作用。我们的研究表明,槲皮素抑制KLF4介导的VSMC向巨噬细胞样细胞的表型转换,并通过抑制JAK2/STAT3途径减少动脉粥样硬化。
    The objective of this study was to elucidate the protective role of quercetin in atherosclerosis by examining its effect on the phenotypic switch of vascular smooth muscle cells (VSMCs) to macrophage-like cells and the underlying regulatory pathways. Aorta tissues from apolipoprotein E-deficient (ApoE KO) mice fed a high-fat diet (HFD), treated with or without 100 mg/kg/day quercetin, were analyzed for histopathological changes and molecular mechanisms. Quercetin was found to decrease the size of atherosclerotic lesions and mitigate lipid accumulation induced by HFD. Fluorescence co-localization analysis revealed a higher presence of macrophage-like vascular smooth muscle cells (VSMCs) co-localizing with phospho-Janus kinase 2 (p-JAK2), phospho-signal transducer and activator of transcription 3 (p-STAT3), and Krüppel-like factor 4 (KLF4) in regions of foam cell aggregation within aortic plaques. However, this co-localization was reduced following treatment with quercetin. Quercetin treatment effectively inhibited the KLF4-mediated phenotypic switch in oxidized low-density lipoprotein (ox-LDL)-loaded mouse aortic vascular smooth muscle cells (MOVAS), as indicated by decreased expressions of KLF4, LGALS3, CD68, and F4/80, increased expression of alpha smooth muscle actin (α-SMA), reduced intracellular fluorescence Dil-ox-LDL uptake, and decreased lipid accumulation. In contrast, APTO-253, a KLF4 activator, was found to reverse the effects of quercetin. Furthermore, AG490, a JAK2 inhibitor, effectively counteracted the ox-LDL-induced JAK2/STAT3 pathway-dependent switch to a macrophage-like phenotype and lipid accumulation in MOVAS cells. These effects were significantly mitigated by quercetin but exacerbated by coumermycin A1, a JAK2 activator. Our research illustrates that quercetin inhibits the KLF4-mediated phenotypic switch of VSMCs to macrophage-like cells and reduces atherosclerosis by suppressing the JAK2/STAT3 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
    人脐带间充质干细胞来源的小细胞外囊泡(MSC-sEV)为糖尿病肾病(DKD)患者的无细胞治疗提供了实用的解决方案。然而,在DKD中,MSC-sEV的潜在保护机制仍然未知.体内和体外分析表明,MSC-sEV减轻了DKD的肾脏纤维化和炎症。通过高通量测序探索MSC-sEV诱导治疗效果的潜在机制,其鉴定了与人皮肤成纤维细胞-sEV(HSF-sEV)相比MSC-sEV中一组miRNA的独特富集。体外实验表明,保护潜力主要归因于miR-23a-3p,MSC-sEV中最丰富的miRNA之一。Further,过表达或敲低分析显示miR-23a-3p,其靶标Krüppel样因子3(KLF3)抑制了高糖(HG)诱导的HK-2细胞中的STAT3信号通路,这对于MSC-sEV的肾脏保护特性至关重要。此外,我们发现miR-23a-3p通过RNA结合基序蛋白X连接(RBMX)包装到MSC-sEV中,并传递至HG诱导的HK-2细胞。最后,抑制miR-23a-3p可以减轻db/db小鼠MSC-sEV的保护作用。这些发现表明,系统性给予源自MSC的sEV,具有整合到肾脏中的能力,在那里它们可以通过递送miR-23a-3p来发挥针对HG诱导的损伤的肾脏保护潜力。
    Human umbilical cord mesenchymal stem cells-derived small extracellular vesicles (MSC-sEV) provide a pragmatic solution as a cell-free therapy for patients with diabetic kidney disease (DKD). However, the underlying protective mechanisms of MSC-sEV remain largely unknown in DKD. Invivo and in vitro analyses demonstrated that MSC-sEV attenuated renal fibrosis and inflammation of DKD. The underlying mechanism of the MSC-sEV-induced therapeutic effect was explored by high-throughput sequencing, which identified the unique enrichment of a set of miRNAs in MSC-sEV compared with human skin fibroblasts-sEV (HSF-sEV). Vitro experiments demonstrated that the protective potential was primarily attributed to miR-23a-3p, one of the most abundant miRNAs in MSC-sEV. Further, overexpression or knockdown analyses revealed that miR-23a-3p, and its target Krüppel-like factor 3 (KLF3) suppressed the STAT3 signaling pathway in high glucose (HG) induced HK-2 cells were essential for the renal-protective property of MSC-sEV. Moreover, we found that miR-23a-3p was packaged into MSC-sEV by RNA Binding Motif Protein X-Linked (RBMX) and transmitted to HG-induced HK-2 cells. Finally, inhibiting miR-23a-3p could mitigate the protective effects of MSC-sEV in db/db mice. These findings suggest that a systemic administration of sEV derived from MSC, have the capacity to incorporate into kidney where they can exert renal-protective potential against HG-induced injury through delivery of miR-23a-3p.
    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
    铁稳态对活生物体至关重要。果蝇已经成为研究铁稳态的优秀模型,而铁代谢的调节机制仍然知之甚少。在这里,我们偶然发现,特别是脂肪体内的少年激素(JH)酸甲基转移酶(Jhamt)的敲除,JH合成的关键限速酶,导致当地的铁积累,导致身体脂肪严重流失和功能障碍。Jhamt敲除诱导的表型通过铁剥夺得到缓解,抗氧化剂和Ferrostatin-1,一种众所周知的铁凋亡抑制剂,提示铁性凋亡与Jhamt敲除诱导的脂肪体缺陷有关。进一步的研究表明,Tsf1和Malvolio(Mvl,哺乳动物DMT1的同源物),两个铁进口商,解释了Jhamt敲除诱导的铁积累和脂肪体功能障碍。机械上,Kr-h1是JH的关键转录因子,在Jhamt的下游转录抑制Tsf1和Mvl。总之,研究结果表明,源自脂肪体的Jhamt通过维持脂肪体内的铁稳态来促进果蝇的发育,提供对果蝇铁代谢调节机制的独特见解。
    Iron homeostasis is of critical importance to living organisms. Drosophila melanogaster has emerged as an excellent model to study iron homeostasis, while the regulatory mechanism of iron metabolism remains poorly understood. Herein, we accidently found that knockdown of juvenile hormone (JH) acid methyltransferase (Jhamt) specifically in the fat body, a key rate-limiting enzyme for JH synthesis, led to iron accumulation locally, resulting in serious loss and dysfunction of fat body. Jhamt knockdown-induced phenotypes were mitigated by iron deprivation, antioxidant and Ferrostatin-1, a well-known inhibitor of ferroptosis, suggesting ferroptosis was involved in Jhamt knockdown-induced defects in the fat body. Further study demonstrated that upregulation of Tsf1 and Malvolio (Mvl, homolog of mammalian DMT1), two iron importers, accounted for Jhamt knockdown-induced iron accumulation and dysfunction of the fat body. Mechanistically, Kr-h1, a key transcription factor of JH, acts downstream of Jhamt inhibiting Tsf1 and Mvl transcriptionally. In summary, the findings indicated that fat body-derived Jhamt is required for the development of Drosophila by maintaining iron homeostasis in the fat body, providing unique insight into the regulatory mechanisms of iron metabolism in Drosophila.
    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
    为了解决日益增长的能源需求,肿瘤细胞经历代谢重编程,包括氧化磷酸化(OXPHOS)和有氧糖酵解。本研究调查了Kruppel样因子4(KLF4)的作用,转录因子,通过调节ATP合成在肝细胞癌(HCC)中作为肿瘤抑制因子。进行免疫组织化学以评估HCC组织中的KLF4表达。功能测定,如CCK-8,EdU,和殖民地的形成,以及体内测定,包括皮下肿瘤形成和肝脏原位异种移植小鼠模型,进行以确定KLF4对HCC增殖的影响。荧光素酶报告基因测定和染色质免疫沉淀测定用于评估KLF4、miR-206和RICTOR之间的相互作用。研究结果表明,肝癌中KLF4表达较低,这与不良预后有关。体外和体内功能测定均表明KLF4抑制HCC细胞增殖。机械上,研究表明,KLF4通过抑制RICTOR的表达来减少HCC中的ATP合成,mTORC2的核心组件。这种抑制促进谷氨酰胺分解以补充TCA循环并增加ATP水平,促进miR-206转录。总之,这项研究增强了对KLF4在HCCATP合成中作用的理解,并提示靶向KLF4/miR-206/RICTOR轴可能是一种有前景的抗HCC治疗方法.
    To address the increased energy demand, tumor cells undergo metabolic reprogramming, including oxidative phosphorylation (OXPHOS) and aerobic glycolysis. This study investigates the role of Kruppel-like factor 4 (KLF4), a transcription factor, as a tumor suppressor in hepatocellular carcinoma (HCC) by regulating ATP synthesis. Immunohistochemistry was performed to assess KLF4 expression in HCC tissues. Functional assays, such as CCK-8, EdU, and colony formation, as well as in vivo assays, including subcutaneous tumor formation and liver orthotopic xenograft mouse models, were conducted to determine the impact of KLF4 on HCC proliferation. Luciferase reporter assay and chromatin immunoprecipitation assay were utilized to evaluate the interaction between KLF4, miR-206, and RICTOR. The findings reveal low KLF4 expression in HCC, which is associated with poor prognosis. Both in vitro and in vivo functional assays demonstrate that KLF4 inhibits HCC cell proliferation. Mechanistically, it was demonstrated that KLF4 reduces ATP synthesis in HCC by suppressing the expression of RICTOR, a core component of mTORC2. This suppression promotes glutaminolysis to replenish the TCA cycle and increase ATP levels, facilitated by the promotion of miR-206 transcription. In conclusion, this study enhances the understanding of KLF4\'s role in HCC ATP synthesis and suggests that targeting the KLF4/miR-206/RICTOR axis could be a promising therapeutic approach for anti-HCC therapeutics.
    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
    原理:CD8+T细胞在其激活和增殖过程中经历一系列代谢重编程过程,包括糖酵解增加,减少糖的有氧氧化,氨基酸代谢增加,蛋白质合成增加。然而,目前尚不清楚是什么因素在肿瘤免疫微环境中调节CD8+T细胞的这些代谢重编程过程.方法:采用T细胞染色盒蛋白4(CBX4)敲除小鼠模型,确定CD8+T细胞中CBX4对肿瘤免疫微环境和肿瘤进展的作用。流式细胞术,Cut-TagqPCR,Chip-seq,免疫沉淀,代谢物检测,进行慢病毒感染和过继性T细胞转移,以探讨CBX4敲除促进CD8T细胞活化和抑制肿瘤生长的潜在机制。结果:我们发现CBX4在肿瘤浸润的CD8T细胞中被诱导表达,并通过调节肿瘤组织中的糖代谢来抑制CD8T细胞的功能。机械上,CBX4通过反式作用转录因子1(SP1)和Krüppel样因子3(KLF3)的sumoylation增加代谢相关分子醛缩酶B(Aldob)的表达。此外,Aldob通过减少丝氨酸/苏氨酸蛋白激酶(Akt)的磷酸化来抑制T细胞中的糖酵解和ATP合成,并最终抑制CD8T细胞功能。重要的是,敲除CBX4可能通过增强肿瘤微环境中CD8+T细胞的功能来提高抗PD-1治疗的疗效。结论:CBX4参与肿瘤组织中CD8+T细胞代谢重编程和功能持久性,并作为CD8+T细胞糖酵解和效应子功能的抑制剂。
    Rationale: CD8+ T cells undergo a series of metabolic reprogramming processes during their activation and proliferation, including increased glycolysis, decreased aerobic oxidation of sugars, increased amino acid metabolism and increased protein synthesis. However, it is still unclear what factors regulate these metabolic reprogramming processes in CD8+ T cells in the tumor immune microenvironment. Methods: T cell chromobox protein 4 (CBX4) knock-out mice models were used to determine the role of CBX4 in CD8+ T cells on the tumor immune microenvironment and tumor progression. Flow cytometry, Cut-Tag qPCR, Chip-seq, immunoprecipitation, metabolite detection, lentivirus infection and adoptive T cells transfer were performed to explore the underlying mechanisms of CBX4 knock-out in promoting CD8+ T cell activation and inhibiting tumor growth. Results: We found that CBX4 expression was induced in tumor-infiltrating CD8+ T cells and inhibited CD8+ T cell function by regulating glucose metabolism in tumor tissue. Mechanistically, CBX4 increases the expression of the metabolism-associated molecule aldolase B (Aldob) through sumoylation of trans-acting transcription factor 1 (SP1) and Krüppel-like factor 3 (KLF3). In addition, Aldob inhibits glycolysis and ATP synthesis in T cells by reducing the phosphorylation of the serine/threonine protein kinase (Akt) and ultimately suppresses CD8+ T cell function. Significantly, knocking out CBX4 may improve the efficacy of anti-PD-1 therapy by enhancing the function of CD8+ T cells in the tumor microenvironment. Conclusion: CBX4 is involved in CD8+ T cell metabolic reprogramming and functional persistence in tumor tissues, and serves as an inhibitor in CD8+ T cells\' glycolysis and effector function.
    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
    多西他赛(Doc)在临床抗肿瘤实践中起着至关重要的作用。然而,不断有证据表明,肿瘤经常发生化学耐药性和复发,这可能与多倍体巨癌细胞(PGCC)有关。本研究的目的是探讨Doc诱导的PGCCs的形成机制和生物学行为。卵巢癌细胞用Doc治疗,然后通过MTT测定和显微成像分析评估Doc对细胞活力的影响。通过Hoechst33342染色进一步评估PGCC的生物学特性,细胞周期和DNA含量测定,DNA损伤反应(DDR)信号检测,β-半乳糖苷酶染色,线粒体膜电位检测,和逆转录-定量聚合酶链反应。结果表明Doc降低了细胞活力;然而,许多细胞还活着,是巨大的多倍体。Doc增加了停留在G2/M期的细胞比例并减少了细胞数量。此外,γ-H2A的表达。在Doc治疗后X不断增加。PGCC显示衰老相关的β-半乳糖苷酶活性和JC-1单体形式的增加。PGCC中八聚体结合转录因子4(OCT4)和krüppel样因子4(KLF4)的mRNA水平显着增加。一起来看,我们的结果表明,Doc诱导G2/M细胞周期停滞,抑制增殖并激活持续的DDR信号以促进PGCC的形成。重要的是,PGCC表现出衰老表型并表达干细胞标志物。
    Docetaxel (Doc) plays a crucial role in clinical antineoplastic practice. However, it is continuously documented that tumors frequently develop chemoresistance and relapse, which may be related to polyploid giant cancer cells (PGCCs). The aim of this study was investigate the formation mechanism and biological behavior of PGCCs induced by Doc. Ovarian cancer cells were treated with Doc, and then the effect of Doc on cellular viability was evaluated by MTT assay and microscopic imaging analysis. The biological properties of PGCCs were further evaluated by Hoechst 33342 staining, cell cycle and DNA content assay, DNA damage response (DDR) signaling detection, β-galactosidase staining, mitochondrial membrane potential detection, and reverse transcription-quantitative polymerase chain reaction. The results indicated that Doc reduced cellular viability; however, many cells were still alive, and were giant and polyploid. Doc increased the proportion of cells stayed in the G2/M phase and reduced the number of cells. In addition, the expression of γ-H2A.X was constantly increased after Doc treatment. PGCCs showed senescence-associated β-galactosidase activity and an increase in the monomeric form of JC-1. The mRNA level of octamer-binding transcription factor 4 (OCT4) and krüppel-like factor 4 (KLF4) was significantly increased in PGCCs. Taken together, our results suggest that Doc induces G2/M cell cycle arrest, inhibits the proliferation and activates persistent DDR signaling to promote the formation of PGCCs. Importantly, PGCCs exhibit a senescence phenotype and express stem cell markers.
    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
    糖尿病肾病(DKD)是糖尿病的微血管并发症,肾小球内皮细胞(GEC)损伤是DKD发病的关键驱动因素。Krüppel样因子2(KLF2),剪切应力诱导的转录因子,在早期DKD中高度上调的基因中发现。在肾脏,KLF2的表达大多局限于内皮细胞,但其表达也存在于免疫细胞亚群中。通过机械感觉受体的激活,响应于增加的剪切应力,KLF2表达上调,但被炎症细胞因子抑制,两者都是早期糖尿病肾脏环境的特征。在人和小鼠的进行性DKD和高血压肾病中KLF2表达降低,可能是由于高糖和炎症细胞因子如TNF-α。然而,KLF2表达在肾小球高滤过诱导的剪切应力下增加,而没有代谢失调,例如在单侧肾切除术中。较低的KLF2表达与单侧肾切除术患者的CKD进展有关,与其内保护作用一致。KLF2通过抑制炎症赋予内源性保护,血栓活化,和血管生成,因此KLF2被认为是心血管疾病(CVD)的保护因素。基于类似的机制,KLF2还表现出肾脏保护,在糖尿病或单侧肾切除术中,其在内皮细胞中的表达降低会加重肾小球损伤和蛋白尿。因此,KLF2在CVD和DKD中均具有保护作用,其激动剂可能被开发为糖尿病患者心-肾保护的新型药物。
    Diabetic kidney disease (DKD) is a microvascular complication of diabetes, and glomerular endothelial cell (GEC) dysfunction is a key driver of DKD pathogenesis. Krüppel-like factor 2 (KLF2), a shear stress-induced transcription factor, is among the highly regulated genes in early DKD. In the kidney, KLF2 expression is mostly restricted to endothelial cells, but its expression is also found in immune cell subsets. KLF2 expression is upregulated in response to increased shear stress by the activation of mechanosensory receptors but suppressed by inflammatory cytokines, both of which characterize the early diabetic kidney milieu. KLF2 expression is reduced in progressive DKD and hypertensive nephropathy in humans and mice, likely due to high glucose and inflammatory cytokines such as TNF-α. However, KLF2 expression is increased in glomerular hyperfiltration-induced shear stress without metabolic dysregulation, such as in settings of unilateral nephrectomy. Lower KLF2 expression is associated with CKD progression in patients with unilateral nephrectomy, consistent with its endoprotective role. KLF2 confers endoprotection by inhibition of inflammation, thrombotic activation, and angiogenesis, and thus KLF2 is considered a protective factor for cardiovascular disease (CVD). Based on similar mechanisms, KLF2 also exhibits renoprotection, and its reduced expression in endothelial cells worsens glomerular injury and albuminuria in settings of diabetes or unilateral nephrectomy. Thus KLF2 confers endoprotective effects in both CVD and DKD, and its activators could potentially be developed as a novel class of drugs for cardiorenal protection in diabetic patients.
    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
    慢性胰腺炎(CP)的特点是进行性纤维化和胰腺星状细胞(PSC)的激活,伴随着胰腺实质的破坏,导致腺泡细胞(AC)的损失。很少有研究探讨受损AC(DACs)促进PSC活化和胰腺纤维化的机制。目前,目前尚无治疗CP或限制胰腺纤维化进展的有效药物。在这项研究中,与完整腺泡细胞(IACs)共培养抑制PSC活化,而与DAC的共同文化则相反。Krüppel样因子4(KLF4)在DAC中被显着上调,并被确立为将AC从PSC抑制剂转换为PSC激活剂的关键分子。我们揭示了IACs的外泌体有助于IACs-CS对PSC的抗激活功能。MiRNome谱分析显示let-7家族在IACs衍生的外泌体中显著富集(>30%miRNome),部分介导IAC对PSC的抑制性影响。此外,已经观察到,外来体中let-7的富集受KLF4表达水平的影响。机理研究表明,KLF4在AC中上调Lin28A,从而降低AC衍生的外泌体中的let-7s水平,从而促进PSC活化。我们利用特异性靶向AC中的KLF4的腺相关病毒(shKLF4-pAAV)来抑制CP中的PSC活化,导致胰腺纤维化减少。IACs衍生的外泌体具有作为通过let-7s抵抗PSC激活的有效武器的潜力,而在DAC中激活KLF4/Lin28A信号则削弱了此类功能。ShKLF4-pAAV有望成为CP的新型治疗方法。
    Chronic pancreatitis (CP) is marked by progressive fibrosis and the activation of pancreatic stellate cells (PSCs), accompanied by the destruction of pancreatic parenchyma, leading to the loss of acinar cells (ACs). Few research studies have explored the mechanism by which damaged ACs (DACs) contribute to PSCs activation and pancreatic fibrosis. Currently, there are no effective drugs for curing CP or limiting the progression of pancreatic fibrosis. In this research, co-culture with intact acinar cells (IACs) suppressed PSC activation, while co-culture with DACs did the opposite. Krüppel-like factor 4 (KLF4) was significantly upregulated in DACs and was established as the key molecule that switches ACs from PSCs-suppressor to PSCs-activator. We revealed the exosomes of IACs contributed to the anti-activated function of IACs-CS on PSCs. MiRNome profiling showed that let-7 family is significantly enriched in IAC-derived exosomes (>30% miRNome), which partially mediates IACs\' suppressive impacts on PSCs. Furthermore, it has been observed that the enrichment of let-7 in exosomes was influenced by the expression level of KLF4. Mechanistic studies demonstrated that KLF4 in ACs upregulated Lin28A, thereby decreasing let-7 levels in AC-derived exosomes, and thus promoting PSCs activation. We utilized an adeno-associated virus specifically targeting KLF4 in ACs (shKLF4-pAAV) to suppress PSCs activation in CP, resulting in reduced pancreatic fibrosis. IAC-derived exosomes hold potential as potent weapons against PSCs activation via let-7s, while activated KLF4/Lin28A signaling in DACs diminished such functions. ShKLF4-pAAV holds promise as a novel therapeutic approach for CP.
    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