HIF‐1α

HIF - 1 α
  • 文章类型: Journal Article
    已确定C1R在皮肤鳞状细胞癌中具有独特的功能,超出了其在补体系统中的作用。然而,目前尚不清楚C1R是否参与肝细胞癌(HCC)的进展.HCC组织用于检查C1R表达与临床和病理因素的关系。通过体外和体内实验评估HCC细胞的恶性特征。通过RNA-seq探讨了C1R在肝癌中的作用机制,甲基化特异性PCR,免疫沉淀,和双荧光素酶报告基因测定。本研究发现,C1R的表达随着HCC恶性程度的增加而降低,并与预后不良有关。C1R启动子通过DNMT1和DNMT3a高度甲基化,导致C1R表达减少。C1R表达的下调通过HIF-1α介导的糖酵解的激活导致HCC细胞的恶性特征增强。此外,发现降低的C1R表达促进异种移植肿瘤形成。我们发现C反应蛋白(CRP)与C1R结合,游离的CRP激活NF-κB信号通路,进而增强HIF-1α的表达。HIF-1α的增加导致更高的糖酵解水平,最终促进HCC的攻击行为。C1R启动子区的甲基化导致HCC中C1R表达的下调。C1R通过抑制HIF-1α调节的糖酵解在体外和体内抑制HCC中的攻击行为。这些发现表明,C1R在HCC进展过程中充当抑癌基因,为创新的治疗方法开辟了新的可能性。
    C1R has been identified to have a distinct function in cutaneous squamous cell carcinoma that goes beyond its role in the complement system. However, it is currently unknown whether C1R is involved in the progression of hepatocellular carcinoma (HCC). HCC tissues were used to examine C1R expression in relation to clinical and pathological factors. Malignant characteristics of HCC cells were assessed through in vitro and in vivo experiments. The mechanism underlying the role of C1R in HCC was explored through RNA-seq, methylation-specific PCR, immuno-precipitation, and dual-luciferase reporter assays. This study found that the expression of C1R decreased as the malignancy of HCC increased and was associated with poor prognosis. C1R promoter was highly methylated through DNMT1 and DNMT3a, resulting in a decrease in C1R expression. Downregulation of C1R expression resulted in heightened malignant characteristics of HCC cells through the activation of HIF-1α-mediated glycolysis. Additionally, decreased C1R expression was found to promote xenograft tumor formation. We found that C-reactive protein (CRP) binds to C1R, and the free CRP activates the NF-κB signaling pathway, which in turn boosts the expression of HIF-1α. This increase in HIF-1α leads to higher glycolysis levels, ultimately promoting aggressive behavior in HCC. Methylation of the C1R promoter region results in the downregulation of C1R expression in HCC. C1R inhibits aggressive behavior in HCC in vitro and in vivo by inhibiting HIF-1α-regulated glycolysis. These findings indicate that C1R acts as a tumor suppressor gene during HCC progression, opening up new possibilities for innovative therapeutic approaches.
    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:血管生成是成功愈合牙髓损伤的关键事件,缺氧是牙髓血管生成的主要刺激因子。在这项研究中,我们研究了缺氧对人牙髓干细胞(hDPSCs)促血管生成潜能的影响以及miR-143-5p在此过程中的作用.
    方法:分离人牙髓干细胞,体外培养和表征。氯化钴(CoCl2)用于在hDPSC中诱导缺氧。CCK-8和Transwell测定用于确定缺氧对hDPSC增殖和迁移的影响。定量实时聚合酶链反应(qRT-PCR),进行蛋白质印迹(WB)和ELISA以评估hDPSC中HIF-1α和血管生成细胞因子的mRNA和蛋白质水平。使用Matrigel管形成和鸡绒毛尿囊膜(CAM)测定在体外测量缺氧对hDPSC促血管生成潜能的影响。构建重组慢病毒载体以在hDPSC中稳定过表达或抑制miR-143-5p,并使用qRT-PCR评估促血管生成作用,WB,和试管形成测定。使用生物信息学预测工具鉴定和验证miR-143-5p靶基因,双荧光素酶报告基因测定和RNA下拉实验。最后,我们使用裸鼠皮下移植模型来确定缺氧处理和miR-143-5p过表达/抑制hDPSC在牙髓再生中的作用.
    结果:低氧促进hDPSC增殖,迁移和促血管生成潜力。体内实验表明,缺氧处理(50和100μMCoCl2)可促进牙髓血管生成和牙本质形成。与促血管生成因子的水平相反,miR-143-5p水平随着CoCl2浓度的增加而降低。miR-143-5p抑制显著促进hDPSC的促血管生成潜能,而miR-143-5p过表达在体外抑制血管生成。双荧光素酶报告基因测定将视黄酸受体相关的孤儿受体α(RORA)鉴定为hDPSC中的miR-143-5p靶基因。RNA下拉实验表明,HIF-1α和RORA被生物素标记的miR-143-5p拉低,缺氧组HIF-1α和RORA与miR-143-5p结合的水平低于常氧组。hDPSC中miR-143-5p表达的抑制促进异位牙髓组织再生。
    结论:CoCl2诱导的缺氧促进hDPSC驱动的旁分泌血管生成和牙髓再生。miR-143-5p的抑制通过直接靶向HIF-1α和RORA上调缺氧条件下hDPSC的促血管生成潜能。
    OBJECTIVE: Angiogenesis is a key event in the successful healing of pulp injuries, and hypoxia is the main stimulator of pulpal angiogenesis. In this study, we investigated the effect of hypoxia on the proangiogenic potential of human dental pulp stem cells (hDPSCs) and the role of miR-143-5p in the process.
    METHODS: Human dental pulp stem cells were isolated, cultured and characterized in vitro. Cobalt chloride (CoCl2) was used to induce hypoxia in hDPSCs. CCK-8 and Transwell assays were used to determine the effect of hypoxia on hDPSCs proliferation and migration. Quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting (WB) and ELISA were performed to assess the mRNA and protein levels of HIF-1α and angiogenic cytokines in hDPSCs. The effect of hypoxia on hDPSCs proangiogenic potential was measured in vitro using Matrigel tube formation and chick chorioallantoic membrane (CAM) assays. Recombinant lentiviral vectors were constructed to stably overexpress or inhibit miR-143-5p in hDPSCs, and the proangiogenic effects were assessed using qRT-PCR, WB, and tube formation assays. miR-143-5p target genes were identified and verified using bioinformatics prediction tools, dual-luciferase reporter assays and RNA pull-down experiments. Finally, a subcutaneous transplantation model in nude mice was used to determine the effects of hypoxia treatment and miR-143-5p overexpression/inhibition in hDPSCs in dental pulp regeneration.
    RESULTS: Hypoxia promotes hDPSCs proliferation, migration and proangiogenic potential. The in vivo experiments showed that hypoxia treatment (50 and 100 μM CoCl2) promoted pulp angiogenesis and dentine formation. In contrast to the levels of proangiogenic factors, miR-143-5p levels decreased with increasing CoCl2 concentration. miR-143-5p inhibition significantly promoted proangiogenic potential of hDPSCs, whereas miR-143-5p overexpression inhibited angiogenesis in vitro. Dual-luciferase reporter assay identified retinoic acid receptor-related orphan receptor alpha (RORA) as an miR-143-5p target gene in hDPSCs. RNA pull-down experiments demonstrated that HIF-1α and RORA were pulled down by biotin-labelled miR-143-5p, and the levels of HIF-1α and RORA bound to miR-143-5p in the hypoxia group were lower than those in the normoxia group. Inhibition of miR-143-5p expression in hDPSCs promoted ectopic dental pulp tissue regeneration.
    CONCLUSIONS: CoCl2-induced hypoxia promotes hDPSCs-driven paracrine angiogenesis and pulp regeneration. The inhibition of miR-143-5p upregulates the proangiogenic potential of hDPSCs under hypoxic conditions by directly targeting HIF-1α and RORA.
    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:乳腺癌(BC)是全球范围内威胁女性健康的最常见的恶性肿瘤之一。据报道,环状RNA(circularRNAs)在调节肿瘤进展和肿瘤微环境(TME)重塑中起重要作用。
    方法:使用高通量测序和生物信息学分析验证了BC中circRNAs的差异表达特征和免疫相关性。外泌体通过纳米颗粒透射电子显微镜和跟踪分析进行表征。在体外和体内都证明了circ-0100519在BC发育中的生物学功能。西方印迹,RNA下拉,RNA免疫沉淀,流式细胞术,和荧光素酶报告基因进行了研究,以探讨其潜在的机制。
    结果:Circ-0100519在BC肿瘤组织中含量丰富,与预后不良有关。它可以被包裹在分泌的外泌体中,从而通过诱导M2样巨噬细胞极化促进BC细胞侵袭和转移。机械上,circ-0100519充当支架,以增强去泛素化酶泛素特异性蛋白酶7(USP7)与巨噬细胞中核因子样2(NRF2)之间的相互作用,诱导USP7介导的NRF2去泛素化。此外,HIF-1α可以作为上游效应子发挥作用以增强circ-0100519转录。
    结论:我们的研究表明,外泌体circ-0100519是BC诊断和预后的潜在生物标志物,HIF-1α抑制剂PX-478可以为BC提供治疗靶标。
    BACKGROUND: Breast cancer (BC) is one of the most prevalent malignant tumours that threatens women health worldwide. It has been reported that circular RNAs (circRNAs) play an important role in regulating tumour progression and tumour microenvironment (TME) remodelling.
    METHODS: Differentially expression characteristics and immune correlations of circRNAs in BC were verified using high-throughput sequencing and bioinformatic analysis. Exosomes were characterised by nanoparticle transmission electron microscopy and tracking analysis. The biological function of circ-0100519 in BC development was demonstrated both in vitro and in vivo. Western blotting, RNA pull-down, RNA immunoprecipitation, flow cytometry, and luciferase reporter were conducted to investigate the underlying mechanism.
    RESULTS: Circ-0100519 was significant abundant in BC tumour tissues and related to poor prognosis. It can be encapsulated into secreted exosomes, thereby promoting BC cell invasion and metastasis via inducing M2-like macrophages polarisation.Mechanistically, circ-0100519 acted as a scaffold to enhance the interaction between the deubiquitinating enzyme ubiquitin-specific protease 7 (USP7) and nuclear factor-like 2 (NRF2) in macrophages, inducing the USP7-mediated deubiquitination of NRF2. Additionally, HIF-1α could function as an upstream effector to enhance circ-0100519 transcription.
    CONCLUSIONS: Our study revealed that exosomal circ-0100519 is a potential biomarker for BC diagnosis and prognosis, and the HIF-1α inhibitor PX-478 may provide a therapeutic target for BC.
    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:血管生成在皮质梗死后继发性丘脑损伤的神经保护中至关重要。p75神经营养蛋白受体(p75NTR)在激活血管生成中起关键作用。然而,p75NTR对皮质梗死后丘脑血管生成的影响尚不清楚.在此,我们研究了p75NTR是否通过激活VonHippel-Lindau(VHL)介导的缺氧诱导因子1α(HIF-1α)/血管内皮生长因子(VEGF)通路在大脑中动脉远端闭塞(dMCAO)后促进血管生成以减轻继发性丘脑损伤。
    方法:建立雄性dMCAO大鼠模型。使用RNA测序评估p75NTR对血管生成的影响,免疫组织化学,westernblot,定量实时聚合酶链反应,磁共振成像,行为测试,病毒和药物干预。
    结果:我们发现dMCAO后同侧丘脑的p75NTR和血管密度降低。p75NTR-VHL相互作用减少,dMCAO后促进HIF-1α的泛素化降解,降低VEGF的表达。值得注意的是,p75NTR过表达通过抑制VHL-HIF-1α相互作用抑制HIF-1α的泛素化降解,进一步促进血管生成,dMCAO后同侧丘脑脑血流量增加,神经功能改善。
    结论:第一次,我们强调,p75NTR-VHL相互作用的增强促进了dMCAO后继发性丘脑损伤的血管生成。
    BACKGROUND: Angiogenesis is crucial in neuroprotection of secondary thalamic injury after cortical infarction. The p75 neurotrophin receptor (p75NTR) plays a key role in activating angiogenesis. However, the effects of p75NTR on angiogenesis in the thalamus after cortical infarction are largely unknown. Herein we investigate whether p75NTR facilitates angiogenesis to attenuate secondary thalamic damage via activating hypoxia-inducible factor 1α (HIF-1α)/vascular endothelial growth factor (VEGF) pathway mediated by Von Hippel-Lindau (VHL) after distal middle cerebral artery occlusion (dMCAO).
    METHODS: The male rat model of dMCAO was established. The effects of p75NTR on the angiogenesis was evaluated using RNA-sequencing, immunohistochemistry, western blot, quantitative real-time polymerase chain reaction, magnetic resonance imaging, behavior tests, viral and pharmacological interventions.
    RESULTS: We found that the p75NTR and vessel density were decreased in ipsilateral thalamus after dMCAO. The p75NTR-VHL interaction was reduced, which promoted the ubiquitination degradation of HIF-1α and reduced VEGF expression after dMCAO. Notably, p75NTR overexpression restrained the ubiquitination degradation of HIF-1α by inhibiting VHL-HIF-1α interaction, further promoted angiogenesis, increased cerebral blood flow of ipsilateral thalamus and improved neurological function after dMCAO.
    CONCLUSIONS: For the first time, we highlighted that the enhancement of p75NTR-VHL interaction promoted angiogenesis in attenuating secondary thalamic damage after dMCAO.
    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
    癌症相关性疲劳(CRF)显著影响癌症患者的生活质量。本研究调查了参芪扶正注射液(SFI)在治疗CRF中的治疗潜力,专注于其在骨骼肌中的机械作用。我们利用CRF小鼠模型来检查SFI对身体耐力的影响,监测活动水平,游泳时间和休息时间。使用等量异位标签和液相色谱-串联质谱法进行腓肠肌的蛋白质组学分析以绘制SFI处理后的肌肉蛋白质组变化。通过ATP生物发光测定法评估骨骼肌中的线粒体功能。此外,通过蛋白质印迹法探讨了缺氧诱导因子1亚基α(HIF-1α)信号通路在介导SFI效应中的调节作用。在CRF诱导的C2C12成肌细胞中,我们评估了细胞活力(CCK-8测定),细胞凋亡(流式细胞术)和线粒体自噬(电子显微镜)。这项研究还采用了下拉法,荧光素酶和染色质免疫沉淀试验,以阐明SFI作用的分子机制,特别关注PINK1通过HIF-1α结合在PINK1启动子区域的转录调节。我们的研究结果表明,SFI增强了身体活动能力,减轻疲劳症状,并通过减轻线粒体损伤和增强抗氧化反应对骨骼肌发挥保护作用。SFI促进细胞活力并诱导线粒体自噬,同时减少细胞凋亡,主要通过HIF-1α的调制,PINK1和p62蛋白。这些结果强调了SFI在增强线粒体自噬方面的功效,从而为改善CRF提供了一种有希望的方法。该研究不仅深入了解SFI的潜在治疗机制,而且为进一步探索SFI干预措施在CRF管理中的应用奠定了基础。
    Cancer-related fatigue (CRF) significantly impacts the quality of life of cancer patients. This study investigates the therapeutic potential of Shenqi Fuzheng injection (SFI) in managing CRF, focusing on its mechanistic action in skeletal muscle. We utilized a CRF mouse model to examine the effects of SFI on physical endurance, monitoring activity levels, swimming times and rest periods. Proteomic analysis of the gastrocnemius muscle was performed using isobaric tags and liquid chromatography-tandem mass spectrometry to map the muscle proteome changes post-SFI treatment. Mitochondrial function in skeletal muscle was assessed via ATP bioluminescence assay. Furthermore, the regulatory role of the hypoxia inducible factor 1 subunit alpha (HIF-1α) signalling pathway in mediating SFI\'s effects was explored through western blotting. In CRF-induced C2C12 myoblasts, we evaluated cell viability (CCK-8 assay), apoptosis (flow cytometry) and mitophagy (electron microscopy). The study also employed pulldown, luciferase and chromatin immunoprecipitation assays to elucidate the molecular mechanisms underlying SFI\'s action, particularly focusing on the transcriptional regulation of PINK1 through HIF-1α binding at the PINK1 promoter region. Our findings reveal that SFI enhances physical mobility, reduces fatigue symptoms and exerts protective effects on skeletal muscles by mitigating mitochondrial damage and augmenting antioxidative responses. SFI promotes cell viability and induces mitophagy while decreasing apoptosis, primarily through the modulation of HIF-1α, PINK1 and p62 proteins. These results underscore SFI\'s efficacy in enhancing mitochondrial autophagy, thereby offering a promising approach for ameliorating CRF. The study not only provides insight into SFI\'s potential therapeutic mechanisms but also establishes a foundation for further exploration of SFI interventions in CRF management.
    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:葡萄糖依赖性促胰岛素多肽(GIP)是葡萄糖依赖性促胰岛素多肽受体(GIPR)的配体,在消化系统中起重要作用。近年来,GIP已被认为是调节局部代谢环境的激素样肽。在这项研究中,我们研究了GIP对神经元的抗氧化作用,并探讨了可能的机制。
    方法:细胞计数试剂盒-8(CCK-8)用于测量细胞存活。使用TdT介导的dUTP尼克末端标记(TUNEL)在体外和体内检测细胞凋亡。活性氧(ROS)水平用2',7\'-二氯二氢荧光素二乙酸酯(DCFH-DA),用2-NBDG检测葡萄糖摄入量。免疫荧光染色和蛋白质印迹用于评估细胞和组织中的蛋白质水平。苏木精-伊红(HE)染色,免疫荧光染色和示踪观察脊髓损伤的形态。Basso-Beattie-Bresnahan(BBB)测定用于评估脊髓损伤后的功能恢复。
    结果:GIP降低了培养的神经元和损伤的脊髓中的ROS水平并保护细胞免于凋亡。GIP促进受伤脊髓的伤口愈合和功能恢复。GIP显著提高了培养神经元的葡萄糖摄取。同时,葡萄糖摄取的抑制显著减弱了GIP的抗氧化作用。GIP通过上调缺氧诱导因子1α(HIF-1α)的水平以Akt依赖性方式增加葡萄糖转运蛋白3(GLUT3)的表达。
    结论:GIP增加GLUT3表达并促进神经元的葡萄糖摄入,它在体外和体内发挥抗氧化作用并保护神经元细胞免受氧化应激。
    OBJECTIVE: Glucose-dependent insulinotropic polypeptide (GIP) is a ligand of glucose-dependent insulinotropic polypeptide receptor (GIPR) that plays an important role in the digestive system. In recent years, GIP has been regarded as a hormone-like peptide to regulate the local metabolic environment. In this study, we investigated the antioxidant role of GIP on the neuron and explored the possible mechanism.
    METHODS: Cell counting Kit-8 (CCK-8) was used to measure cell survival. TdT-mediated dUTP Nick-End Labeling (TUNEL) was used to detect apoptosis in vitro and in vivo. Reactive oxygen species (ROS) levels were probed with 2\', 7\'-Dichloro dihydrofluorescein diacetate (DCFH-DA), and glucose intake was detected with 2-NBDG. Immunofluorescence staining and western blot were used to evaluate the protein level in cells and tissues. Hematoxylin-eosin (HE) staining, immunofluorescence staining and tract-tracing were used to observe the morphology of the injured spinal cord. Basso-Beattie-Bresnahan (BBB) assay was used to evaluate functional recovery after spinal cord injury.
    RESULTS: GIP reduced the ROS level and protected cells from apoptosis in cultured neurons and injured spinal cord. GIP facilitated wound healing and functional recovery of the injured spinal cord. GIP significantly improved the glucose uptake of cultured neurons. Meanwhile, inhibition of glucose uptake significantly attenuated the antioxidant effect of GIP. GIP increased glucose transporter 3 (GLUT3) expression via up-regulating the level of hypoxia-inducible factor 1α (HIF-1α) in an Akt-dependent manner.
    CONCLUSIONS: GIP increases GLUT3 expression and promotes glucose intake in neurons, which exerts an antioxidant effect and protects neuronal cells from oxidative stress both in vitro and in vivo.
    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
    髓核在人体内处于缺氧环境,当椎间盘退变(IVDD)发生时,缺氧环境被破坏。髓核细胞(NPC)铁性凋亡是IVDD的原因之一。N6-甲基腺苷(m6A)及其阅读器蛋白YTHDF1通过影响RNA代谢来调节细胞活性。然而,在缺氧条件下,m6A修饰的RNA对NPCs中铁凋亡的调节尚未得到很好的研究。在这项研究中,通过体外和体内实验,我们探讨了HIF-1α和YTHDF1调节NPCs铁凋亡的潜在机制。结果表明,HIF-1α或YTHDF1的过表达抑制了NPC的铁性凋亡;相反,HIF-1α或YTHDF1的敲除增加了NPCs中的铁凋亡水平。荧光素酶报告基因测定和染色质免疫沉淀表明HIF-1α通过直接结合其启动子区来调节YTHDF1的转录。多聚体谱分析结果表明,YTHDF1通过与m6A修饰的SLC7A11mRNA结合,促进了SLC7A11的翻译,从而促进了抗铁凋亡蛋白GPX4的表达。总之,HIF-1α诱导的YTHDF1表达通过以m6A依赖性方式促进SLC7A11翻译减少NPC的铁凋亡并延迟IVDD。
    The nucleus pulposus is in a hypoxic environment in the human body, and when intervertebral disc degeneration (IVDD) occurs, the hypoxic environment is disrupted. Nucleus pulposus cell (NPC) ferroptosis is one of the causes of IVDD. N6-methyladenosine (m6A) and its reader protein YTHDF1 regulate cellular activities by affecting RNA metabolism. However, the regulation of ferroptosis in NPCs by m6A-modified RNAs under hypoxic conditions has not been as well studied. In this study, through in vitro and in vivo experiments, we explored the underlying mechanism of HIF-1α and YTHDF1 in regulating ferroptosis in NPCs. The results indicated that the overexpression of HIF-1α or YTHDF1 suppressed NPC ferroptosis; conversely, the knockdown of HIF-1α or YTHDF1 increased ferroptosis levels in NPCs. Luciferase reporter assays and chromatin immunoprecipitation demonstrated that HIF-1α regulated YTHDF1 transcription by directly binding to its promoter region. Polysome profiling results showed that YTHDF1 promoted the translation of SLC7A11 and consequently the expression of the anti-ferroptosis protein GPX4 by binding to m6A-modified SLC7A11 mRNA. In conclusion, HIF-1α-induced YTHDF1 expression reduces NPC ferroptosis and delays IVDD by promoting SLC7A11 translation in a m6A-dependent manner.
    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
    低氧预处理已被认为是加速皮肤伤口愈合的促进因素。我们之前的研究发现,外泌体lncRNAH19,来自脂肪干细胞(ADSCs),在协调皮肤伤口愈合中起着至关重要的作用。在这里,我们旨在探讨缺氧与ADSC来源的外泌体(ADSC-exos)在皮肤伤口愈合中是否存在联系.使用透射电子显微镜(TEM)和粒度分析鉴定在常氧和低氧条件下从ADSC提取的外泌体。ADSCs-exos对细胞增殖的影响,迁移,用CCK-8、EdU、伤口愈合,和试管形成测定。H19、HIF-1α的表达模式,和USP22进行测量。免疫共沉淀,染色质免疫沉淀,泛素化,和荧光素酶报告基因测定进行确认USP22/HIF-1α/H19轴,在小鼠皮肤创伤模型中进一步验证。从缺氧处理的ADSCs中提取的外泌体(称为H-ADSCs-exos)显着增加细胞增殖,迁移,和H2O2暴露的HUVECs中的血管生成,并促进体内皮肤伤口愈合。此外,H-ADSCs和H-ADSCs-exos,H19水平较高,被发现被HIF-1α转录激活。机械上,携带USP22的H-ADSCs可导致去泛素化和稳定HIF-1α。此外,H-ADSCs-exos促进细胞增殖,迁移,通过激活USP22/HIF-1α轴并促进H19表达,在H2O2触发的HUVECs中血管生成,为临床治疗皮肤创面愈合提供新的线索。
    Hypoxic preconditioning has been recognized as a promotive factor for accelerating cutaneous wound healing. Our previous study uncovered that exosomal lncRNA H19, derived from adipose-derived stem cells (ADSCs), plays a crucial role in orchestrating cutaneous wound healing. Herein, we aimed to explore whether there is a connection between hypoxia and ADSC-derived exosomes (ADSCs-exos) in cutaneous wound healing. Exosomes extracted from ADSCs under normoxic and hypoxic conditions were identified using transmission electron microscope (TEM) and particle size analysis. The effects of ADSCs-exos on the proliferation, migration, and angiogenesis of human umbilical vein endothelial cells (HUVECs) were evaluated by CCK-8, EdU, wound healing, and tube formation assays. Expression patterns of H19, HIF-1α, and USP22 were measured. Co-immunoprecipitation, chromatin immunoprecipitation, ubiquitination, and luciferase reporter assays were conducted to confirm the USP22/HIF-1α/H19 axis, which was further validated in a mice model of skin wound. Exosomes extracted from hypoxia-treated ADSCs (termed as H-ADSCs-exos) significantly increased cell proliferation, migration, and angiogenesis in H2O2-exposed HUVECs, and promoted cutaneous wound healing in vivo. Moreover, H-ADSCs and H-ADSCs-exos, which exhibited higher levels of H19, were found to be transcriptionally activated by HIF-1α. Mechanically, H-ADSCs carrying USP22 accounted for deubiquitinating and stabilizing HIF-1α. Additionally, H-ADSCs-exos improved cell proliferation, migration, and angiogenesis in H2O2-triggered HUVECs by activating USP22/HIF-1α axis and promoting H19 expression, which may provide a new clue for the clinical treatment of cutaneous wound healing.
    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:激活肾素-血管紧张素系统,作为高血压和慢性肾脏疾病(CKD)的标志,是导致肾小管间质纤维化进展的关键病理生理因素。LIM和衰老细胞抗原样结构域蛋白1(LIMS1)通过与其他蛋白质形成复合物在控制细胞行为中起重要作用。这里,研究了LIMS1在血管紧张素II(AngII)诱导的高血压和肾小管间质纤维化中的功能和调节。
    方法:用AngII处理C57BL/6小鼠以诱导肾小管间质纤维化。使用缺氧诱导因子-1α(HIF-1α)肾小管特异性敲除小鼠或LIMS1敲除AAV来研究其对AngII诱导的肾间质纤维化的影响。体外,HIF-1α或LIMS1在HK2细胞暴露于AngII后被敲低或过表达。
    结果:在患有高血压肾病的人肾和AngII诱导的高血压模型的鼠肾中观察到肾小管LIMS1的表达增加。LIMS1的肾小管特异性敲除改善了AngII诱导的小鼠肾小管间质纤维化。此外,我们证明LIMS1在肾小管细胞中受HIF-1α转录调节,而肾小管HIF-1α敲除可改善LIMS1介导的肾小管间质纤维化.此外,LIMS1通过与波形蛋白相互作用促进AngII诱导的肾小管间质纤维化。
    结论:我们得出结论,HIF-1α转录调节的LIMS1通过与波形蛋白相互作用在AngII诱导的肾小管间质纤维化中起核心作用。我们的发现代表了对AngII诱导的肾小管间质纤维化机制的新见解,并为CKD的进展提供了新的治疗靶标。
    OBJECTIVE: Activation of the renin-angiotensin system, as a hallmark of hypertension and chronic kidney diseases (CKD) is the key pathophysiological factor contributing to the progression of tubulointerstitial fibrosis. LIM and senescent cell antigen-like domains protein 1 (LIMS1) plays an essential role in controlling of cell behaviour through the formation of complexes with other proteins. Here, the function and regulation of LIMS1 in angiotensin II (Ang II)-induced hypertension and tubulointerstitial fibrosis was investigated.
    METHODS: C57BL/6 mice were treated with Ang II to induce tubulointerstitial fibrosis. Hypoxia-inducible factor-1α (HIF-1α) renal tubular-specific knockout mice or LIMS1 knockdown AAV was used to investigate their effects on Ang II-induced renal interstitial fibrosis. In vitro, HIF-1α or LIMS1 was knocked down or overexpressed in HK2 cells after exposure to Ang II.
    RESULTS: Increased expression of tubular LIMS1 was observed in human kidney with hypertensive nephropathy and in murine kidney from Ang II-induced hypertension model. Tubular-specific knockdown of LIMS1 ameliorated Ang II-induced tubulointerstitial fibrosis in mice. Furthermore, we demonstrated that LIMS1 was transcriptionally regulated by HIF-1α in tubular cells and that tubular HIF-1α knockout ameliorates LIMS1-mediated tubulointerstitial fibrosis. In addition, LIMS1 promotes Ang II-induced tubulointerstitial fibrosis by interacting with vimentin.
    CONCLUSIONS: We conclude that HIF-1α transcriptionally regulated LIMS1 plays a central role in Ang II-induced tubulointerstitial fibrosis through interacting with vimentin. Our finding represents a new insight into the mechanism of Ang II-induced tubulointerstitial fibrosis and provides a novel therapeutic target for progression of CKD.
    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
    缺氧,癌症中普遍存在的标志,强调了靶向HIF-1α的重要性,缺氧反应的主要转录因子,有效的癌症治疗。在这里,我们引入DNA轭,一类具有特定HIF-1α结合序列的新型DNA纳米材料(缺氧反应元件,HREs),作为治疗癌症的纳米药物。包括一个基底四面体DNA纳米结构(TDN)和四个HRE的悬垂链,DNA轭表现出优异的稳定性和延长的细胞内保留。我们的调查揭示了它们结合HIF-1α的能力,从而破坏其与下游基因组DNA的相互作用并阻碍转录活性。此外,DNA轭通过泛素化途径促进HIF-1α降解,从而将其与下游目标隔离并最终促进其降解。此外,DNA轭减少癌细胞增殖,迁移,缺氧条件下的入侵,同时也显示出肿瘤内的优先积累,从而抑制体内肿瘤的生长和转移。这项研究通过开发具有高稳定性和对正常细胞低毒性的基于DNA的药物,开创了一种新的癌症治疗方法。定位DNA轭作为癌症治疗的有希望的候选人。本文受版权保护。保留所有权利。
    Hypoxia, a ubiquitous hallmark in cancer, underscores the significance of targeting HIF-1α, the principal transcriptional factor of hypoxic responses, for effective cancer therapy. Herein, DNA yokes, a novel class of DNA nanomaterials harboring specific HIF-1α binding sequences (hypoxia response elements, HREs), are introduced as nanopharmaceuticals for cancer treatment. Comprising a basal tetrahedral DNA nanostructure and four HRE-bearing overhanging chains, DNA yokes exhibit exceptional stability and prolonged intracellular retention. The investigation reveals their capacity to bind HIF-1α, thereby disrupting its interaction with the downstream genomic DNAs and impeding transcriptional activity. Moreover, DNA yokes facilitate HIF-1α degradation via the ubiquitination pathway, thereby sequestering it from downstream targets and ultimately promoting its degradation. In addition, DNA yokes attenuate cancer cell proliferation, migration, and invasion under hypoxic conditions, while also displaying preferential accumulation within tumors, thereby inhibiting tumor growth and metastasis in vivo. This study pioneers a novel approach to cancer therapy through the development of DNA-based drugs characterized by high stability and low toxicity to normal cells, positioning DNA yokes as promising candidates for cancer treatment.
    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