HIF-1a

HIF - 1a
  • 文章类型: Journal Article
    免疫检查点阻断疗法在某些类型的癌症中已显示出显著的治疗效果。然而,关于体力活动对其疗效的影响的报道有限。这项研究旨在研究身体活动对抗PDL-1介导的免疫检查点疗法的影响以及其中免疫细胞的相互作用。用抗PDL-1联合身体活动处理HePa1-6荷瘤小鼠以评估肿瘤进展。流式细胞术用于分析肿瘤内的免疫细胞浸润和分化水平。评估由于身体活动引起的HIF-a/CEACAM1在肿瘤内的表达。在小鼠中验证具有高CEACAM1表达的HePa1-6细胞以确定其对免疫细胞增殖和分化的抑制作用。开发了CD3/CEACAM1嵌合抗体用于治疗CEACAM1过表达的肿瘤,流式细胞术用于评估T细胞反应。身体活动通过抑制肿瘤内的HIF-a/CEACAM1轴来增强抗PDL1的功效。体内实验表明,CEACAM1高表达的肿瘤可以减少肿瘤内CD8+T细胞的浸润和活化,抑制T细胞的细胞毒性而不影响Treg浸润。体外,高CEACAM1表达影响共培养系统中CD8+T细胞的增殖和活化。构建的CD3/CEACAM1嵌合抗体显著激活过表达CEACAM1的肿瘤内的TCR并抑制肿瘤进展。研究结果表明,身体活动通过抑制肿瘤内HIF1-α/CEACM1轴来增强免疫检查点阻断的有效性。
    Immune checkpoint blockade therapy has demonstrated significant therapeutic effects in certain types of cancers. However, there is limited reporting on the influence of physical activity on its efficacy. This study aimed to investigate the impact of physical activity on anti-PDL-1-mediated immune checkpoint therapy and the interplay of immune cells therein. HePa1-6 tumor-bearing mice were treated with anti-PDL-1 in conjunction with physical activity to assess tumor progression. Flow cytometry was utilized to analyze immune cell infiltration and differentiation levels within the tumor. The expression of HIF-a/CEACAM1 within the tumor due to physical activity was evaluated. HePa1-6 cells with high CEACAM1 expression were validated in mice to determine their inhibitory effects on immune cell proliferation and differentiation. A CD3/CEACAM1 chimeric antibody was developed for treating CEACAM1-overexpressing tumors, and flow cytometry was employed to assess T-cell response. Physical activity enhanced the efficacy of anti-PDL1 by suppressing the HIF-a/CEACAM1 axis within the tumor. In vivo experiments revealed that tumors with high CEACAM1 expression decreased infiltration and activation of CD8 + T cells within the tumor, suppressing T cell cytotoxicity without affecting Treg infiltration. In vitro, high CEACAM1 expression impacted the proliferation and activation of CD8 + T cells in a co-culture system. The constructed CD3/CEACAM1 chimeric antibody significantly activated the TCR within CEACAM1-overexpressing tumors and inhibited tumor progression. The findings suggest that physical activity augments the effectiveness of immune checkpoint blockade by inhibiting the intratumoral HIF1-α/CEACM1 axis.
    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
    缺氧反应途径能够适应缺氧。它由缺氧诱导因子(HIF)介导,促进代谢重编程,红细胞生成,血管生成和组织重塑。这导致了用于治疗贫血的HIF诱导药物的成功开发,并且这些分子中的一些现在已经在临床上。然而,HIF水平升高通常与肿瘤生长有关,预后不良,以及各种癌症的耐药性,包括肝细胞癌(HCC)。因此,在某些临床情况下,对于HIF诱导药物的推荐存在担忧.这里,我们分析了两种HIF诱导药物的作用,Molidustat和Roxadustat,在特征明确的HCC细胞系Huh7中。这些药物增加了HIF-1α和HIF-2α蛋白水平,两者都参与诱导缺氧反应基因,如BNIP3,SERPINE1,LDHA或EPO。联合转录组学,蛋白质组学和代谢组学研究表明,Molidustat增加了糖酵解酶的表达,而线粒体网络支离破碎,细胞呼吸减少。这种代谢重塑与增殖减少和嘧啶供应需求降低有关。而是细胞将丙酮酸转化为乳酸的能力增强。这伴随着对抗霉素A抑制线粒体呼吸的更高抗性,在Roxadustat治疗的Huh7细胞和Molidustat治疗的肝母细胞瘤细胞(Huh6和HepG2)中证实了一种表型。总的来说,这项研究表明,HIF诱导药物增加了肝癌细胞对代谢应激源的代谢弹性,主张仔细监测使用HIF诱导药物治疗的患者,尤其是当他们有肝癌的风险时。
    The hypoxia response pathway enables adaptation to oxygen deprivation. It is mediated by hypoxia-inducible factors (HIF), which promote metabolic reprogramming, erythropoiesis, angiogenesis and tissue remodeling. This led to the successful development of HIF-inducing drugs for treating anemia and some of these molecules are now in clinic. However, elevated levels of HIFs are frequently associated with tumor growth, poor prognosis, and drug resistance in various cancers, including hepatocellular carcinoma (HCC). Consequently, there are concerns regarding the recommendation of HIF-inducing drugs in certain clinical situations. Here, we analyzed the effects of two HIF-inducing drugs, Molidustat and Roxadustat, in the well-characterized HCC cell line Huh7. These drugs increased HIF-1α and HIF-2α protein levels which both participate in inducing hypoxia response genes such as BNIP3, SERPINE1, LDHA or EPO. Combined transcriptomics, proteomics and metabolomics showed that Molidustat increased the expression of glycolytic enzymes, while the mitochondrial network was fragmented and cellular respiration decreased. This metabolic remodeling was associated with a reduced proliferation and a lower demand for pyrimidine supply, but an increased ability of cells to convert pyruvate to lactate. This was accompanied by a higher resistance to the inhibition of mitochondrial respiration by antimycin A, a phenotype confirmed in Roxadustat-treated Huh7 cells and Molidustat-treated hepatoblastoma cells (Huh6 and HepG2). Overall, this study shows that HIF-inducing drugs increase the metabolic resilience of liver cancer cells to metabolic stressors, arguing for careful monitoring of patients treated with HIF-inducing drugs, especially when they are at risk of liver 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
    赖氨酸特异性去甲基酶1(KDM1A/LSD1)调节线粒体呼吸并稳定HIF-1A(缺氧诱导因子1A)。HIF-1A通过增加细胞葡萄糖摄取来调节活性氧(ROS)水平,糖酵解,和内源性抗氧化剂。KDM1A在细胞ROS应答中的作用以前没有描述过。我们确定了KDM1A在调节ROS反应中的作用以及KDM1A抑制剂与ROS诱导癌症疗法组合的效用。我们的结果表明,KDM1A抑制使细胞对氧化应激敏感,并增加总细胞ROS,通过用抗氧化剂N-乙酰半胱氨酸处理来缓解。KDM1A抑制降低ROS暴露后基础线粒体呼吸和HIF-1A诱导受损。来自KDM1A抑制的HIF-1A补救细胞的过表达增强了对ROS的敏感性。因此,我们发现KDM1A抑制后ROS的敏感性增加是由HIF-1A和内源性谷胱甘肽的消耗介导的。我们还表明,KDM1A特异性抑制剂bizine与抗氧化剂消耗疗法协同作用,丁硫氨酸亚砜胺,横纹肌肉瘤细胞系(Rh28和Rh30)中的乌诺芬。在这项研究中,我们描述了KDM1A在氧化应激下调节HIF-1A功能的新作用,并发现KDM1A和抗氧化剂系统的双重靶向可能是一种有效的联合抗癌策略。
    Lysine Specific Demethylase 1 (KDM1A / LSD1) regulates mitochondrial respiration and stabilizes HIF-1A (hypoxia-inducible factor 1A). HIF-1A modulates reactive oxygen species (ROS) levels by increasing cellular glucose uptake, glycolysis, and endogenous antioxidants. The role of KDM1A in cellular ROS response has not previously been described. We determined the role of KDM1A in regulating the ROS response and the utility of KDM1A inhibitors in combination with ROS-inducing cancer therapies. Our results show that KDM1A inhibition sensitized cells to oxidative stress and increased total cellular ROS, which was mitigated by treatment with the antioxidant N-acetyl cysteine. KDM1A inhibition decreased basal mitochondrial respiration and impaired induction of HIF-1A after ROS exposure. Overexpression of HIF-1A salvaged cells from KDM1A inhibition enhanced sensitivity to ROS. Thus we found that increased sensitivity of ROS after KDM1A inhibition was mediated by HIF-1A and depletion of endogenous glutathione. We also show that KDM1A-specific inhibitor bizine synergized with antioxidant-depleting therapies, buthionine sulfoximine, and auranofin in rhabdomyosarcoma cell lines (Rh28 and Rh30). In this study, we describe a novel role for KDM1A in regulating HIF-1A functions under oxidative stress and found that dual targeting of KDM1A and antioxidant systems may serve as an effective combination anticancer strategy.
    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:防己黄芪汤(FHD)是临床常用的治疗风寒、风湿型浅虚证的处方。对类风湿关节炎(RA)也有显著疗效。
    目的:本研究旨在探讨FHD抗RA的可能机制,为RA的替代治疗提供理论依据。
    方法:我们使用UPLC-Q-TOF-MS分析FHD的成分和吸收的血液成分。同时,建立胶原诱导性关节炎(CIA)大鼠模型,通过考虑体重来评估对FHD的治疗效果,关节炎评分,爪子肿胀,自主运动能力,和滑膜微血管计数。随后,免疫荧光,免疫组织化学,采用westernblot检测FHD在体内的抗血管生成能力,以及滑膜组织中凋亡和自噬的水平。此外,流式细胞术和Westernblot检测FHD对MH7A细胞凋亡和自噬的影响。CCK8法检测FHD对MH7A细胞增殖和迁移的影响,细胞迁移和,入侵实验。最后,在MH7A细胞和HUVEC细胞共培养中,进行了试管形成试验以评估FHD的血管生成能力.
    结果:通过测试FHD的原始配方,总共确定了26种活性成分,其中17个被吸收到血液中。FHD明显改善CIA大鼠的病理症状和滑膜增生。FHD可以抑制HIF-1α的表达,促进CIA大鼠滑膜组织凋亡,并抑制自噬和血管生成。体外实验表明,含FHD的血清抑制细胞增殖,迁移,和MH7A细胞的侵袭,抑制自噬相关蛋白的表达,同时促进细胞凋亡。FHD明显抑制TNF-α刺激的MH7A细胞中HIF-1α蛋白的表达,并抑制MH7A细胞在HUVEC细胞中诱导的管形成能力。
    结论:该研究证明FHD具有出色的抗RA作用,这可能归因于其通过抑制HIF-1α调节RAFLS中自噬与凋亡之间的平衡的潜在机制,从而有助于其抗血管生成活性。
    BACKGROUND: Fangji Huangqi Decoction (FHD) is frequently prescribed for the clinical treatment of wind-cold and wind-dampness pathogenic superficial deficiency syndrome. It also has a notable curative effect on rheumatoid arthritis (RA).
    OBJECTIVE: The study aimed to explore the possible mechanism of FHD against RA and provided a theoretical basis for alternative therapies for RA.
    METHODS: We used UPLC-Q-TOF-MS to analysis the ingredients and absorbed blood components of FHD. At the same time, the collagen-induced arthritis (CIA) rat model was established to estimate the therapeutic effects on FHD by considering body weight, arthritis score, paw swelling, autonomous movement ability, and synovial microvessel counts. Subsequently, immunofluorescence, immunohistochemistry, and Western blot were employed to detect the anti-angiogenic capacity of FHD in vivo, as well as the levels of apoptosis and autophagy in the synovial tissue. In addition, flow cytometry and Western blot were used to assess the effects of FHD on apoptosis and autophagy in MH7A cells. The effects of FHD on the proliferation and migration of MH7A cells were measured by CCK8 assay, cell migration and, invasion experiments. Finally, a tube formation assay was performed to evaluate the angiogenic capacity of FHD in co-cultures of MH7A cells and HUVEC cells.
    RESULTS: Through testing of FHD\'s original formula, a total of 26 active ingredients have been identified, with 17 of them being absorbed into the bloodstream. FHD significantly improved the pathological symptoms and synovial hyperplasia of CIA rats. FHD could suppress the expression of HIF-1α, promote apoptosis in CIA rat synovial tissue, and suppress autophagy and angiogenesis. In vitro experiments showed that serum containing FHD inhibited the proliferation, migration, and invasion of MH7A cells, and also suppressed the expression of autophagy-related proteins while promoting apoptosis. FHD markedly repressed the expression of HIF-1α protein in TNF-α-stimulated MH7A cells and inhibited the tube formation capacity induced by MH7A cells in HUVEC cells.
    CONCLUSIONS: The study had proven that FHD played an excellent anti-RA role, which may be attributed to its potential mechanism of regulating the balance between autophagy and apoptosis in RA FLS by suppressing the HIF-1α, thus contributing to its anti-angiogenic activities.
    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
    缺氧诱导因子-1α(HIF-1α)为外科皮瓣缺血预处理促进其存活提供了新的方向。在之前的研究中,我们证明了HIF-1aDNA质粒在此应用中的有效性。在这项研究中,为了避免与质粒使用相关的并发症,我们试图通过mRNA转染表达HIF-1α,并通过测量下游血管生成基因的上调来确定其生物学活性。我们转染了六种不同的HIF-1amRNA-一种主要的,三个变体,和两个新的突变同工型-使用脂质体进入原代人真皮成纤维细胞,并使用RT-qPCR评估mRNA水平。在转染后检查的所有时间点(3、6和10小时),HIF-1α转录物的水平在所有HIF-1α转染的细胞中相对于对照显著更高(所有p<0.05,未配对的学生T检验)。重要的是,HIF-1α转录反应基因的表达(VEGF,ANG-1,PGF,在转染后6和/或10小时,用所有同种型转染的细胞中的FLT1和EDN1)明显高于对照。所有同种型均成功转染人成纤维细胞,导致所测试的所有五个下游血管生成靶标的快速上调。这些发现支持HIF-1αmRNA用于保护缺血性皮瓣的潜在用途。
    Hypoxia-Inducible Factor-1α (HIF-1α) has presented a new direction for ischemic preconditioning of surgical flaps to promote their survival. In a previous study, we demonstrated the effectiveness of HIF-1a DNA plasmids in this application. In this study, to avoid complications associated with plasmid use, we sought to express HIF-1α through mRNA transfection and determine its biological activity by measuring the upregulation of downstream angiogenic genes. We transfected six different HIF-1a mRNAs-one predominant, three variant, and two novel mutant isoforms-into primary human dermal fibroblasts using Lipofectamine, and assessed mRNA levels using RT-qPCR. At all time points examined after transfection (3, 6, and 10 h), the levels of HIF-1α transcript were significantly higher in all HIF-1α transfected cells relative to the control (all p < 0.05, unpaired Student\'s T-test). Importantly, the expression of HIF-1α transcription response genes (VEGF, ANG-1, PGF, FLT1, and EDN1) was significantly higher in the cells transfected with all isoforms than with the control at six and/or ten hours post-transfection. All isoforms were transfected successfully into human fibroblast cells, resulting in the rapid upregulation of all five downstream angiogenic targets tested. These findings support the potential use of HIF-1α mRNA for protecting ischemic dermal flaps.
    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
    线粒体是细胞质细胞器,在神经发育和维持过程中对神经干细胞(NSC)命运的调节中起着重要作用。在胚胎和成人神经发生期间,NSC经历从糖酵解到氧化磷酸化的代谢转换,线粒体DNA(mtDNA)含量增加,线粒体形状和大小的变化,和线粒体活性氧的生理增强,共同驱动神经干细胞增殖和分化。参与细胞分化的蛋白质的遗传和表观遗传修饰(雷帕霉素的机制靶标),增殖(无翼型),和缺氧(丝裂原激活的蛋白激酶)-和所有由共同的关键调节因子缺氧诱导因子-1A连接-被认为是负责的代谢转变,因此,NSC在生理和病理条件下的命运。由于核DNA或mtDNA突变引起的原发性线粒体功能障碍或氧化磷酸化(OXPHOS)代谢中的继发性线粒体功能障碍,线粒体动力学,和细胞器相互作用通路可以促进神经发育或进行性神经退行性疾病的发展。这篇综述从可用的体外和体内模型开始分析神经发育的生理学和病理学,并强调了有关该过程中涉及的关键线粒体途径的最新知识。
    Mitochondria are cytoplasmic organelles having a fundamental role in the regulation of neural stem cell (NSC) fate during neural development and maintenance.During embryonic and adult neurogenesis, NSCs undergo a metabolic switch from glycolytic to oxidative phosphorylation with a rise in mitochondrial DNA (mtDNA) content, changes in mitochondria shape and size, and a physiological augmentation of mitochondrial reactive oxygen species which together drive NSCs to proliferate and differentiate. Genetic and epigenetic modifications of proteins involved in cellular differentiation (Mechanistic Target of Rapamycin), proliferation (Wingless-type), and hypoxia (Mitogen-activated protein kinase)-and all connected by the common key regulatory factor Hypoxia Inducible Factor-1A-are deemed to be responsible for the metabolic shift and, consequently, NSC fate in physiological and pathological conditions.Both primary mitochondrial dysfunction due to mutations in nuclear DNA or mtDNA or secondary mitochondrial dysfunction in oxidative phosphorylation (OXPHOS) metabolism, mitochondrial dynamics, and organelle interplay pathways can contribute to the development of neurodevelopmental or progressive neurodegenerative disorders.This review analyses the physiology and pathology of neural development starting from the available in vitro and in vivo models and highlights the current knowledge concerning key mitochondrial pathways involved in this process.
    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
    背景缺氧诱导因子-1α(HIF-1α)被认为通过上调缺氧诱导基因来控制血管生成和代谢,如白细胞介素-33(IL-33)基因和血管内皮生长因子(VEGF)基因。本研究旨在研究HIF-1α及其两个缺氧通路基因IL-33和VEGF,以及血管生成,并将它们与一些预后临床病理特征相关联,分别和组合评估它们的依赖性。方法本研究包括87例结直肠癌(CRC)病例,在2019年1月至2022年12月期间诊断。检查了不同的预后临床病理特征,并设计了组织微阵列(TMA)载玻片以进行IHC对肿瘤细胞中的IL-33和VEGF评分,定性解释VEGF在肿瘤血管中的表达。对HIF-1α进行分子分析,所有数据均与临床病理特征相关,单独和集体,评估这些因素的依赖性。结果IL-33、VEGF、和预后临床病理特征。而单独分析HIF-1α在远处转移患者中显示出显着的高平均表达,并且随着淋巴结(LN)受累的增加而增加。然而,当HIF1-α的表达与VEGF和IL-33的临床病理特征相关时,与转移的显着相关性在肿瘤细胞中消失,仅在肿瘤血管生成的内皮中出现。此外,结果与LN的参与相冲突。结论这些发现可能提示HIF1-α在VEGF和IL-33以外的生物标志物的下游调节中的作用,这需要揭示HIF1-α调节的通路和新因子在恶性肿瘤的促炎症和血管生成中的作用。
    Background The hypoxia-inducible factor-1 alpha (HIF-1α) is believed to control angiogenesis and metabolism by upregulating hypoxia-induced genes, such as the interLeukin-33 (IL-33) gene and vascular endothelial growth factor (VEGF) gene. The study aimed to study the HIF-1α and its two hypoxia pathway genes; IL-33 and VEGF, together with the angiogenesis and correlate them with some prognostic clinicopathological features, separately and in combination to assess their dependency. Methodology This study included 87 colorectal cancer (CRC) cases, diagnosed between January 2019 and December 2022. Different prognostic clinicopathological features were examined and tissue microarray (TMA) slides were designed to carry out IHC for IL-33 and VEGF scoring in tumor cells, in addition to qualitative interpretation of VEGF expression in tumor vessels. Molecular analysis was performed for HIF-1α and all data were correlated to the clinicopathological features, separately and collectively, to assess the dependency of these factors. Results No statistical correlation could be seen among the IL-33, VEGF, and prognostic clinicopathological features. Whereas analysis of the HIF-1α alone showed significantly high mean expression in patients with distance metastasis and was increased with the increased involvement of the lymph nodes (LNs). However, when the HIF 1-α expression was correlated with the clinicopathological characteristics on the bases of VEGF and IL-33 expressions the significant association with metastasis disappeared in tumor cells and appeared only with the endothelium of the tumor angiogenesis. Moreover, the results conflicted with the LNs involvement. Conclusions These findings may suggest a role of HIF 1-α in the downstream regulation of biomarkers other than the VEGF and IL-33, which needs to uncover pathways and novel factors regulated by the HIF 1-α for the proinflammation and angiogenesis in malignancy.
    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
    宫颈癌的发病率在女性恶性肿瘤中排名第二,对他们的生活质量和整体福祉产生重大影响。低氧微环境在肿瘤发生的发生和发展中起着关键作用。本研究旨在探讨缺氧诱导因子(HIF-1A)在宫颈癌中的相关基因和通路。旨在确定用于诊断和治疗目的的潜在下游靶标。
    我们从综合基因表达数据库(GEO)获得数据集GSE63514。数据集包括正常组的24名患者和肿瘤组的28名患者。基因集差异分析(GSVA)和基因集富集分析(GSEA)用于鉴定与HIF-1A表达相关的基因和涉及的特定信号通路。在TCGA数据集中检查HIF-1A与肿瘤免疫浸润之间的关联。构建WGCAN网络以识别蓝色模块中的关键基因,以及随后的基因本体论(GO)功能和京都基因和基因组百科全书(KEGG)途径富集分析进行以确定与HIF-1A相关的途径和功能注释。同时从STRING数据库获得HIF-1A基因的蛋白质相互作用网络,并使用Cytoscape进行可视化。在体内验证了HIF-1A的功能及其相关基因的表达。
    HIF-1A是宫颈癌患者单因素和多因素Cox回归分析的危险因素。通过相关性分析,共鉴定出344个与HIF-1A表达显著相关的基因,并获得了表现出最强相关性的基因。参与HIF-1A的主要信号通路包括TNF-α/NF-κB,PI3K/AKT/MTOR,TGF-β,JAK-STAT,和各种其他信号级联。通过qRT-PCR增强,我们鉴定了整合素β-1(ITGB1),C-C基序趋化因子配体2(CCL2),纹状体蛋白3(STRN3),和内皮素-1(EDN1)作为受HIF-1A影响的关键下游基因。HIF-1A与自然杀伤(NK)细胞的免疫浸润有关,肥大细胞,CD4+T细胞,M0巨噬细胞,中性粒细胞,滤泡辅助性T细胞,CD8+T细胞,和调节性T细胞(Treg)。HIF-1A与化疗药物的敏感性相关。HIF-1A途径及其功能的鉴定主要集中在细胞质翻译上,有氧呼吸,细胞呼吸,氧化磷酸化,产热,在其他人中。体内实验结果证实HIF-1A在促进宫颈癌细胞的迁移和侵袭中起着至关重要的作用。此外,HIF-1A的过表达导致ITGB1、CCL2、STRN3和EDN1的表达上调。
    通过生物信息学分析和实验验证相结合,确定了HIF-1A在宫颈癌中的作用。鉴定了可能与HIF-1A的肿瘤发生机制有关的基因。这些发现有可能增强我们对宫颈癌进展的理解,并为其临床管理提供有希望的治疗目标。
    UNASSIGNED: The incidence of cervical cancer ranks second among malignant tumors in women, exerting a significant impact on their quality of life and overall well-being. The hypoxic microenvironment plays a pivotal role in the initiation and progression of tumorigenesis. The present study aims to investigate the fundamental genes and pathways associated with the hypoxia-inducible factor (HIF-1A) in cervical cancer, aiming to identify potential downstream targets for diagnostic and therapeutic purposes.
    UNASSIGNED: We obtained dataset GSE63514 from the Comprehensive Gene Expression Database (GEO). The dataset comprised of 24 patients in the normal group and 28 patients in the tumor group. Gene set difference analysis (GSVA) and gene set enrichment analysis (GSEA) were used to identify the genes related to HIF-1A expression and the specific signaling pathways involved.The association between HIF-1A and tumor immune infiltration was examined in the TCGA dataset. The WGCAN network was constructed to identify key genes within the blue module, and subsequent gene ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were conducted to determine the pathways and functional annotations associated with HIF-1A. The protein interaction network of the HIF-1A gene was obtained from the STRING database and visualized using Cytoscape in the meantime.The function of HIF-1A and its related gene expression were verified in vivo.
    UNASSIGNED: HIF-1A was a risk factor in both univariate and multivariate Cox regression analysis of cervical cancer patients. A total of 344 genes significantly correlated with the expression of HIF-1A were identified through correlation analysis, and the genes exhibiting the strongest correlation were obtained. The major signaling pathways involved in HIF-1A encompass TNF-α/NF-κB, PI3K/AKT/MTOR, TGF-β, JAK-STAT, and various other signaling cascades. Reinforced by qRT-PCR, we identified Integrin beta-1 (ITGB1), C-C motif chemokine ligand 2 (CCL2), striatin 3 (STRN3), and endothelin-1 (EDN1) as pivotal downstream genes influenced by HIF-1A. HIF-1A is associated with immune infiltration of natural killer (NK) cells, mast cells, CD4+T cells, M0 macrophages, neutrophils, follicular helper T cells, CD8+T cells, and regulatory T cells (Treg). HIF-1A is associated with sensitivity to chemotherapy drugs. The identification of the HIF-1A pathway and its function primarily focuses on cytoplasmic translation, aerobic respiration, cellular respiration, oxidative phosphorylation, thermogenesis, among others. The results of in vivo experiments have confirmed that HIF-1A plays a crucial role in promoting the migration and invasion of cervical cancer cells. Moreover, the overexpression of HIF-1A led to an upregulation in the expressions of ITGB1, CCL2, STRN3, and EDN1.
    UNASSIGNED: The role of HIF-1A in cervical cancer was determined through a combination of bioinformatics analysis and experimental validation. The genes potentially implicated in the tumorigenesis mechanism of HIF-1A were identified. These findings has the potential to enhance our comprehension of the progression of cervical cancer and offer promising therapeutic targets for its clinical 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
    肝内胆管癌(ICC)是一种高度恶性的消化系统肿瘤,起病隐匿,预后不良。莲心碱,一种来自植物的小分子,已被证明在其他癌症中具有显著的肿瘤抑制活性。然而,目前尚无关于莲心碱是否能抑制ICC的增殖或转移的报道。本研究旨在探讨莲心碱对ICC的抑瘤作用及其机制。使用细胞功能测试在体外监测ICC细胞的表型变化。Western印迹和免疫荧光分析验证了莲心碱的功效。用荷瘤裸鼠研究莲心碱对肿瘤的影响及其体内毒副作用。莲心碱抑制ICC细胞增殖并将细胞周期阻滞在G1期。ICC细胞的上皮-间质转化(EMT)也受到抑制,从而克制其对肿瘤细胞的侵袭和迁徙。此外,本研究发现莲心碱抑制EMT的潜在机制可能是通过缺氧诱导因子1α(HIF-1a)抑制TGF-β1/P-smad3信号通路。体内实验表明,莲心碱抑制裸鼠Hucc-T1移植瘤的生长。莲心碱通过HIF-1a介导的TGF-β1/P-smad3信号通路抑制ICC细胞增殖并抑制ICC中的EMT。
    Intrahepatic cholangiocarcinoma (ICC) is a high-grade malignant digestive system tumor with an insidious onset and unfavorable prognosis. Liensinine, a small molecule derived from plants, has been proven to have significant tumor suppressor activity in other cancers. However, there are no reports on whether liensinine can inhibit the proliferation or metastasis of ICC. This study aimed to explore the tumor-suppressive activity of liensinine in ICC and its underlying mechanisms. The phenotypic changes in ICC cells were monitored in vitro using cell function tests. Western blot and immunofluorescence analyses verified the efficacy of liensinine. Tumor-bearing nude mice were used to explore the effect of liensinine on tumors and its toxicity and side effects in vivo. Liensinine suppressed ICC cell proliferation and arrested the cell cycle at the G1 phase. The epithelial-mesenchymal transition (EMT) of ICC cells was also inhibited, thereby restraining their invasion and migration of tumor cells. In addition, this study found that the potential mechanism of liensinine inhibiting EMT may be via suppression of the TGF-β1/P-smad3 signaling pathway through hypoxia-inducible factor 1 alpha (HIF-1a). In vivo experiments showed that liensinine inhibited the growth of Hucc-T1 transplanted tumors in nude mice. Liensinine restrained the proliferation of ICC cells and suppressed EMT in ICC via the HIF-1a-mediated TGF-β1/P-smad3 signaling pathway.
    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
    人乳头瘤病毒与宫颈癌的发生有关,影响全球女性的第四大常见癌症,而70%的病例是由高危型HPV16和HPV18感染引起的。这些病毒癌基因E6和E7整合到宿主基因组中影响多种细胞功能并改变分子的表达。这项研究的目的是研究这些癌基因如何促进免疫系统控制分子的表达,使用整合了HPV16基因组的细胞系,在使用CRISPR/Cas9系统敲除E6病毒基因之前和之后,用慢病毒载体递送。研究的分子是T细胞失活蛋白PD-L1,其转录因子HIF-1a和后者的负调节因子,miR-143。根据我们的结果,在E6敲除(E6KO)细胞系中,记录到miR-143的表达增加,而HIF-1a和PD-L1的表达下降。这些发现表明,E6蛋白可能在使宫颈癌细胞逃避免疫系统中起重要作用。虽然我们提出了宫颈癌的分子通路,其中PD-L1的表达由E6蛋白通过miR-143/HIF-1a轴调节。
    Human Papillomaviruses have been associated with the occurrence of cervical cancer, the fourth most common cancer that affects women globally, while 70% of cases are caused by infection with the high-risk types HPV16 and HPV18. The integration of these viruses\' oncogenes E6 and E7 into the host\'s genome affects a multitude of cellular functions and alters the expression of molecules. The aim of this study was to investigate how these oncogenes contribute to the expression of immune system control molecules, using cell lines with integrated HPV16 genome, before and after knocking out E6 viral gene using the CRISPR/Cas9 system, delivered with a lentiviral vector. The molecules studied are the T-cell inactivating protein PD-L1, its transcription factor HIF-1a and the latter\'s negative regulator, miR-143. According to our results, in the E6 knock out (E6KO) cell lines an increased expression of miR-143 was recorded, while a decrease in the expression of HIF-1a and PD-L1 was exhibited. These findings indicate that E6 protein probably plays a significant role in enabling cervical cancer cells to evade the immune system, while we propose a molecular pathway in cervical cancer, where PD-L1\'s expression is regulated by E6 protein through a miR-143/HIF-1a axis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号