HIF‐1α

HIF - 1 α
  • 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

  • 文章类型: Journal Article
    运动员越来越多地进行反复的冲刺训练,其中包括反复进行的短暂的全力以赴的努力,以及短暂的恢复。当在缺氧(RSH)中进行时,它可能导致更大的训练效果比在常氧(RSN);然而,潜在的分子机制尚不清楚.本研究旨在阐明与RSN相比RSH对骨骼肌代谢适应的影响。16名健康的年轻人在两种正常的情况下进行了9次重复的冲刺训练(FIO2=0.209,RSN,n=7)或常压缺氧(FIO2=0.136,RSH,n=9)。培训前后,通过重复冲刺能力(RSA)和Wingate测试评估运动表现。使用蛋白质组学结合蛋白质印迹分析进行股外侧肌活检以研究肌肉代谢适应。在RSN和RSH组的RSA和Wingate测试中观察到类似的改善。在肌肉水平,RSN和RSH降低了氧化磷酸化蛋白含量,但触发了线粒体生物发生蛋白的增加。蛋白质组学显示RSH组中几种S100A家族蛋白的增加,其中S100A13最强。我们通过蛋白质印迹证实了RSH中S100A13蛋白的显着增加,这与Akt磷酸化增加及其调节蛋白质合成的下游靶标有关。总之,我们的数据表明,与RSN相比,RSH可以激活S100A/Akt途径以触发特定的适应。
    Athletes increasingly engage in repeated sprint training consisting in repeated short all-out efforts interspersed by short recoveries. When performed in hypoxia (RSH), it may lead to greater training effects than in normoxia (RSN); however, the underlying molecular mechanisms remain unclear. This study aimed at elucidating the effects of RSH on skeletal muscle metabolic adaptations as compared to RSN. Sixteen healthy young men performed nine repeated sprint training sessions in either normoxia (FIO2 = 0.209, RSN, n = 7) or normobaric hypoxia (FIO2 = 0.136, RSH, n = 9). Before and after the training period, exercise performance was assessed by using repeated sprint ability (RSA) and Wingate tests. Vastus lateralis muscle biopsies were performed to investigate muscle metabolic adaptations using proteomics combined with western blot analysis. Similar improvements were observed in RSA and Wingate tests in both RSN and RSH groups. At the muscle level, RSN and RSH reduced oxidative phosphorylation protein content but triggered an increase in mitochondrial biogenesis proteins. Proteomics showed an increase in several S100A family proteins in the RSH group, among which S100A13 most strongly. We confirmed a significant increase in S100A13 protein by western blot in RSH, which was associated with increased Akt phosphorylation and its downstream targets regulating protein synthesis. Altogether our data indicate that RSH may activate an S100A/Akt pathway to trigger specific adaptations as compared to RSN.
    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
    我们先前的发现表明,星形胶质细胞HIF-1α在HIV-1Tat介导的淀粉样变性中起着重要作用,这种淀粉样变性可导致阿尔茨海默病-HIV相关神经认知障碍(HAND)的合并症。这些淀粉样蛋白可以在细胞外囊泡中穿梭,我们试图评估HIV-1Tat刺激的含有毒性淀粉样蛋白的星形胶质细胞衍生的EV(ADEV)是否会在体外和体内导致神经元损伤。因此,我们假设阻断HIF-1α可能减轻HIV-1Tat-ADEV介导的神经元损伤。当暴露于携带有毒淀粉样蛋白的HIV-1Tat-ADEVs时,大鼠海马神经元表现出淀粉样蛋白积累和突触树突状损伤,导致功能丧失,如微型兴奋性突触后电流的改变所证明。星形胶质细胞HIF-1α的沉默不仅降低了ADEVs的生物发生,以及淀粉样蛋白货物,而且还改善了神经元突触变性.接下来,我们确定了携带淀粉样蛋白的HIV-1Tat-ADEV在幼稚小鼠大脑海马中的作用。接受HIV-1Tat-ADEVs的幼稚小鼠,表现出行为变化,和伴有突触变性的阿尔茨海默氏症样病理。在注射HIF-1α沉默ADEV的小鼠中未观察到这种损伤。这是第一份报告,证明了携带淀粉样蛋白的ADEVs在介导突触变性中的作用,导致与HAND相关的行为变化,并强调了HIF-1α的保护作用。
    Our previous findings demonstrated that astrocytic HIF-1α plays a major role in HIV-1 Tat-mediated amyloidosis which can lead to Alzheimer\'s-like pathology-a comorbidity of HIV-Associated Neurocognitive Disorders (HAND). These amyloids can be shuttled in extracellular vesicles, and we sought to assess whether HIV-1 Tat stimulated astrocyte-derived EVs (ADEVs) containing the toxic amyloids could result in neuronal injury in vitro and in vivo. We thus hypothesized that blocking HIF-1α could likely mitigate HIV-1 Tat-ADEV-mediated neuronal injury. Rat hippocampal neurons when exposed to HIV-1 Tat-ADEVs carrying the toxic amyloids exhibited amyloid accumulation and synaptodendritic injury, leading to functional loss as evidenced by alterations in miniature excitatory post synaptic currents. The silencing of astrocytic HIF-1α not only reduced the biogenesis of ADEVs, as well as amyloid cargos, but also ameliorated neuronal synaptodegeneration. Next, we determined the effect of HIV-1 Tat-ADEVs carrying amyloids in the hippocampus of naive mice brains. Naive mice receiving the HIV-1 Tat-ADEVs, exhibited behavioural changes, and Alzheimer\'s \'s-like pathology accompanied by synaptodegeneration. This impairment(s) was not observed in mice injected with HIF-1α silenced ADEVs. This is the first report demonstrating the role of amyloid-carrying ADEVs in mediating synaptodegeneration leading to behavioural changes associated with HAND and highlights the protective role of HIF-1α.
    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
    心脏微血管内皮细胞(CMECs)是心脏心肌细胞周围维持微环境稳态的重要细胞。已经报道了丹参酸钠(SAAS)预防心肌梗塞(MI)损伤。然而,SAAS对CMEC增殖的作用尚不清楚.使用暴露于氧葡萄糖剥夺(OGD)的CEMC来探索SAAS的血管生成能力。在体内,将C57BL/6小鼠分为三组:假手术,MI和SAAS+MI组。与OGD组相比,SAAS导致体外凋亡率降低和增殖增加。此外,SAAS随Bax的降低而升高Bcl2、HIF-1α和血管内皮生长因子(VEGF)的蛋白水平。从具体机制来看,SAAS可能抑制HIF-1α泛素化并增强HIF-1α/VEGF信号通路以增加CMEC增殖。此外,SAAS增加了血管的密度,抑制心肌纤维化,改善体内心功能不全。本研究表明,SAAS可能用作促进MI后CMEC增殖的活性物质。
    Cardiac microvascular endothelial cells (CMECs) are important cells surrounding the cardiomyocytes in the heart that maintain microenvironment homeostasis. Salvianic acid A sodium (SAAS) has been reported to prevent myocardial infarction (MI) injury. However, the role of SAAS on CMEC proliferation remains unclear. CEMCs exposed to oxygen glucose deprivation (OGD) were used to explore the angiogenic abilities of SAAS. In vivo, C57BL/6 mice were divided into three groups: sham, MI and SAAS + MI groups. Compared to OGD group, SAAS led to a reduction in the apoptotic rate and an increase of the proliferation in vitro. Additionally, SAAS increased the protein levels of Bcl2, HIF-1α and vascular endothelial growth factor (VEGF) with the reduction of Bax. In terms of the specific mechanisms, SAAS might inhibit HIF-1α ubiquitination and enhance the HIF-1α/VEGF signalling pathway to increase CMEC proliferation. Furthermore, SAAS increased the density of vessels, inhibited myocardial fibrosis and improved cardiac dysfunction in vivo. The present study has revealed that SAAS could potentially be used as an active substance to facilitate CMEC proliferation post-MI.
    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
    MUC1-C在肿瘤中的表达模式与肿瘤进展密切相关;然而,其具体机制尚不清楚。免疫组化和Westernblot检测MUC1-C在癌组织和癌旁正常组织中的表达。使用CCK8测定法测定细胞对吉西他滨的IC50。研究了缺氧和MUC1-C对膀胱癌细胞行为和代谢特征的影响。通过蛋白质印迹和聚合酶链反应评估基因表达。通过免疫共沉淀分析基因之间的关系,免疫荧光和蛋白质印迹。最后,研究了EGLN2和NF-κB信号通路在MUC1-C与缺氧诱导因子-1α(HIF-1α)相互作用中的作用。MUC1-C在膀胱癌组织中的表达明显高于癌旁正常组织,特别是在大体积肿瘤中,与肿瘤分级等临床特征密切相关。肿瘤体积介导的缺氧导致膀胱癌细胞中MUC1-C和HIF-1α的表达增加。在缺氧的刺激下,EGLN2对NF-κB信号通路的抑制作用减弱,允许NF-κB促进MUC1-C和HIF-1α的正反馈形成。同时,EGLN2介导的HIF-1α降解减少。这最终导致HIF-1α介导的下游基因表达升高,促进葡萄糖摄取和糖酵解增加,并最终导致化疗耐药性和恶性肿瘤。
    The expression pattern of MUC1-C in tumors is closely linked to tumor progression; however, its specific mechanism remains unclear. The expression of MUC1-C in cancer and adjacent normal tissues was detected using immunohistochemistry and Western blot. The IC50 of cells to gemcitabine was determined using the CCK8 assay. The effects of hypoxia and MUC1-C on the behavioral and metabolic characteristics of bladder cancer cells were investigated. Gene expression was assessed through Western blot and polymerase chain reaction. The relationship between the genes was analyzed by co-immunoprecipitation, immunofluorescence and Western blot. Finally, the role of the EGLN2 and NF-κB signaling pathways in the interaction between MUC1-C and hypoxia-inducible factor-1α (HIF-1α) was investigated. MUC1-C expression is significantly higher in bladder cancer tissues than in adjacent normal tissues, particularly in large-volume tumors, and is closely correlated with clinical features such as tumor grade. Tumor volume-mediated hypoxia resulted in increased expression of MUC1-C and HIF-1α in bladder cancer cells. Under stimulation of hypoxia, the inhibitory effect of EGLN2 on the NF-κB signaling pathway was weakened, allowing NF-κB to promote the positive feedback formation of MUC1-C and HIF-1α. Simultaneously, EGLN2-mediated degradation of HIF-1α was reduced. This ultimately led to elevated HIF-1α-mediated downstream gene expression, promoting increased glucose uptake and glycolysis, and ultimately resulting in heightened chemotherapy resistance and malignancy.
    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