p27

P27
  • 文章类型: Journal Article
    结直肠癌是全球癌症相关发病率和死亡率的主要贡献者。癌基因PTOV1与各种人类恶性肿瘤有关,然而其在CRC发病机制中的具体作用需要进一步阐明.
    我们的研究使用了一系列权威的生物信息学工具,比如TIMER,UCSCXena,GEO,人类蛋白质图谱,UALCAN,CIBERSORTx等人用于研究PTOV1对基因表达谱的复杂影响,诊断和预后生物标志物,肿瘤免疫学,信号通路,表观遗传改变,和基因突变。使用Western印迹和qRT-PCR进行基因表达验证。使用CCK-8测定评估CRC细胞的体外增殖和迁移潜力,菌落形成,和transwell迁移分析,分别。MSP用于评估PTOV1启动子区的甲基化状态。
    我们的结果表明,PTOV1表达增加之间存在显着关联,由启动子低甲基化驱动,CRC患者预后不良。升高的PTOV1水平与肿瘤微环境中多种免疫细胞亚群和免疫相关分子的富集呈正相关。体外实验表明,PTOV1敲低显著降低CRC细胞增殖,菌落形成,和移民,而异位PTOV1表达则有相反的作用。重要的是,显示PTOV1调节PI3K-AKT信号通路,显著影响AKT1的磷酸化和细胞周期调控因子P21和P27的表达。使用MK2206的AKT1磷酸化的药理学抑制有效地抵消了由PTOV1过表达诱导的增殖效应。
    PTOV1通过调节AKT1信号通路增强CRC细胞增殖的能力将其确立为潜在的治疗靶标和CRC预后分层的有希望的生物标志物。
    UNASSIGNED: Colorectal cancer is a predominant contributor to global cancer-related morbidity and mortality. The oncogene PTOV1 has been linked to various human malignancies, yet its specific role in CRC pathogenesis requires further elucidation.
    UNASSIGNED: Our study used a comprehensive array of authoritative bioinformatics tools, such as TIMER, UCSC Xena, GEO, Human Protein Atlas, UALCAN, CIBERSORTx and others which used to investigate the complex effects of PTOV1 on gene expression profiles, diagnostic and prognostic biomarkers, tumor immunology, signaling pathways, epigenetic alterations, and genetic mutations. Gene expression validation was conducted using Western blot and qRT-PCR. The in vitro proliferative and migratory potentials of CRC cells were evaluated using CCK-8 assays, colony formation, and transwell migration assays, respectively. MSP was applied to assess the methylation status of the PTOV1 promoter region.
    UNASSIGNED: Our results reveal a significant association between increased PTOV1 expression, driven by promoter hypomethylation, and poor patient prognosis in CRC. Elevated PTOV1 levels were positively correlated with the enrichment of diverse immune cell subsets and immune-related molecules within the tumor microenvironment. In vitro assays demonstrated that PTOV1 knockdown markedly reduced CRC cell proliferation, colony formation, and migration, while ectopic PTOV1 expression had the opposite effect. Importantly, PTOV1 was shown to regulate the PI3K-AKT signaling pathway, significantly influencing the phosphorylation of AKT1 and the expression of cell cycle regulators P21 and P27. The pharmacological inhibition of AKT1 phosphorylation using MK2206 effectively counteracted the proliferative effects induced by PTOV1 overexpression.
    UNASSIGNED: The ability of PTOV1 to enhance CRC cell proliferation via modulation of the AKT1 signaling pathway establishes it as a potential therapeutic target and a promising biomarker for prognostic stratification in CRC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    在许多心血管疾病如心肌梗死(MI)中,增强心肌细胞增殖对于逆转或减缓心力衰竭进展是必需的。已经报道了长链非编码RNA(lncRNA)调节心肌细胞增殖。特别是,lncRNA尿路上皮癌相关1(lncUCA1)在调节细胞周期进程和心血管疾病中起着多重作用,使lncUCA1成为促进心肌细胞增殖的潜在靶标。然而,lncUCA1在心肌细胞增殖中的作用尚不清楚.本研究旨在探讨lncUCA1在心肌细胞增殖中的作用及其分子机制。定量RT-PCR显示lncUCA1在出生后心脏中的表达降低。功能的获得和丧失实验表明,lncUCA1在体外和体内都能正向调节心肌细胞的增殖。生物信息学程序将miR-128鉴定为lncUCA1的潜在靶标,并且据报道miR-128的缺失通过抑制SUZ12/P27途径促进心肌细胞增殖。荧光素酶报告基因测定,qRT-PCR,西方印迹,和免疫染色实验进一步显示,lncUCA1充当miR-128的ceRNA,上调其靶标SUZ12并下调P27,从而增加细胞周期蛋白B1,细胞周期蛋白E,CDK1和CDK2表达增进心肌细胞增殖。总之,lncRNAUCA1的上调通过抑制miR-128/SUZ12/P27途径促进心肌细胞增殖。我们的结果表明,lncUCA1可能是刺激心肌细胞增殖的新治疗靶标。
    Enhancing cardiomyocyte proliferation is essential to reverse or slow down the heart failure progression in many cardiovascular diseases such as myocardial infarction (MI). Long non-coding RNAs (lncRNAs) have been reported to regulate cardiomyocyte proliferation. In particular, lncRNA urothelial carcinoma-associated 1 (lncUCA1) played multiple roles in regulating cell cycle progression and cardiovascular diseases, making lncUCA1 a potential target for promoting cardiomyocyte proliferation. However, the role of lncUCA1 in cardiomyocyte proliferation remains unknown. This study aimed at exploring the function and underlying molecular mechanism of lncUCA1 in cardiomyocyte proliferation. Quantitative RT-PCR showed that lncUCA1 expression decreased in postnatal hearts. Gain-and-loss-of-function experiments showed that lncUCA1 positively regulated cardiomyocyte proliferation in vitro and in vivo. The bioinformatics program identified miR-128 as a potential target of lncUCA1, and loss of miR-128 was reported to promote cardiomyocyte proliferation by inhibiting the SUZ12/P27 pathway. Luciferase reporter assay, qRT-PCR, western blotting, and immunostaining experiments further revealed that lncUCA1 acted as a ceRNA of miR-128 to upregulate its target SUZ12 and downregulate P27, thereby increasing cyclin B1, cyclin E, CDK1 and CDK2 expression to promote cardiomyocyte proliferation. In conclusion, upregulation of lncRNA UCA1 promoted cardiomyocyte proliferation by inhibiting the miR-128/SUZ12/P27 pathway. Our results indicated that lncUCA1 might be a new therapeutic target for stimulating cardiomyocyte proliferation.
    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
    肺癌仍然是癌症相关死亡的主要原因,吸烟是最关键的因素,与近90%的肺癌病例有关。NNK,一种在烟草中发现的高度致癌的亚硝胺,与香烟烟雾的肺癌效应有关。尽管已知NNK会突变或激活某些癌基因,它与p27在调节这些致癌作用方面的潜在相互作用目前尚未被探索。最近的研究已经确定了人类鳞状细胞癌中p27的特定下调,与腺癌相反。此外,暴露于NNK可显着抑制人支气管上皮细胞中p27的表达。随后的研究表明p27的下调在NNK诱导的细胞转化中是关键的。机制研究表明,p27表达减少导致ITCH水平增加,这有利于JunB蛋白的降解。这种退化反过来,增强miR-494表达及其对JAK1mRNA稳定性和蛋白表达的直接调控,最终激活STAT3并驱动细胞转化。总之,我们的研究结果表明:(1)p27的下调通过上调JunBE3连接酶ITCH增加JunB表达,然后促进miR-494转录;(2)升高的miR-494直接结合JAK1mRNA的3'-UTR,(3)JAK1/STAT3途径是p27的下游效应子,介导NNK在肺癌中的致癌作用。这些发现为理解p27抑制NNK诱导的肺鳞状细胞致癌作用的潜在机制提供了重要见解。
    Lung cancer remains the leading cause of cancer-related deaths, with cigarette smoking being the most critical factor, linked to nearly 90% of lung cancer cases. NNK, a highly carcinogenic nitrosamine found in tobacco, is implicated in the lung cancer-causing effects of cigarette smoke. Although NNK is known to mutate or activate certain oncogenes, its potential interaction with p27 in modulating these carcinogenic effects is currently unexplored. Recent studies have identified specific downregulation of p27 in human squamous cell carcinoma, in contrast to adenocarcinoma. Additionally, exposure to NNK significantly suppresses p27 expression in human bronchial epithelial cells. Subsequent studies indicates that the downregulation of p27 is pivotal in NNK-induced cell transformation. Mechanistic investigations have shown that reduced p27 expression leads to increased level of ITCH, which facilitates the degradation of Jun B protein. This degradation in turn, augments miR-494 expression and its direct regulation of JAK1 mRNA stability and protein expression, ultimately activating STAT3 and driving cell transformation. In summary, our findings reveal that: (1) the downregulation of p27 increases Jun B expression by upregulating Jun B E3 ligase ITCH, which then boosts miR-494 transcription; (2) Elevated miR-494 directly binds to 3\'-UTR of JAK1 mRNA, enhancing its stability and protein expression; and (3) The JAK1/STAT3 pathway is a downstream effector of p27, mediating the oncogenic effect of NNK in lung cancer. These findings provide significant insight into understanding the participation of mechanisms underlying p27 inhibition of NNK induced lung squamous cell carcinogenic effect.
    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:PD-L1通过多种机制内在促进肿瘤进展,这可能导致对抗PD-1/PD-L1疗法的耐药性。PD-L1对乳腺癌(BC)细胞增殖的内在作用尚未完全阐明。
    方法:我们使用蛋白质组学,基因表达敲低(KD),定量免疫荧光(qIF),西方印迹,功能测定,包括集落形成测定(CFA)和实时细胞分析仪(RTCA),和体内数据使用免疫组织化学乳腺癌患者。
    结果:PD-L1通过加速G1-S期细胞周期进入促进BC细胞增殖。差异表达的核蛋白的整体蛋白质组学分析表明涉及几种增殖相关分子,包括p21CIP1/WAF1。Western印迹和qIF表明,与PD-L1KD(PD-L1KD)细胞相比,PD-L1表达(PD-L1pos)细胞中SKP2的表达更高,p21CIP1/WAF1和p27Kip1的表达更低。异种移植物来源的细胞和TCGABC数据集在体内证实了这种关系。功能上,CFA和RTCA证明了SKP2在促进PD-L1介导的增殖中的重要作用。最后,74例乳腺癌患者的免疫组织化学证实了PD-L1和SKP-p21/p27轴的关系,因为它显示了SKP2和PD-L1表达之间的高度统计学显着相关性(p<0.001),两者均与增殖标志物Ki-67显著相关(p<0.001)。另一方面,PD-L1和p21CIP1/WAF1表达之间存在统计学上显著的负相关(p=0.005).重要的是,p21CIP1/WAF1和p27Kip1的双阴性与PD-L1显著相关(p<0.001),SKP2(p=0.002),和Ki-67(p=0.002)。
    结论:我们已经证明了SKP2-p27/p21轴在内源性PD-L1增强细胞周期进程中的作用。SKP2表达抑制剂可以减轻对ICPIs的抗性。
    BACKGROUND: PD-L1 intrinsically promotes tumor progression through multiple mechanisms, which potentially leads to resistance to anti-PD-1/PD-L1 therapies. The intrinsic effect of PD-L1 on breast cancer (BC) cell proliferation has not been fully elucidated.
    METHODS: we used proteomics, gene expression knockdown (KD), quantitative immunofluorescence (qIF), western blots, functional assays including colony-forming assay (CFA) and real-time cell analyzer (RTCA), and in vivo data using immunohistochemistry in breast cancer patients.
    RESULTS: PD-L1 promoted BC cell proliferation by accelerating cell cycle entry at the G1-to-S phase transition. Global proteomic analysis of the differentially expressed nuclear proteins indicated the involvement of several proliferation-related molecules, including p21CIP1/WAF1. Western blotting and qIF demonstrated the higher expression of SKP2 and the lower expression of p21CIP1/WAF1 and p27Kip1 in PD-L1 expressing (PD-L1pos) cells as compared to PD-L1 KD (PD-L1KD) cells. Xenograft-derived cells and the TCGA BC dataset confirmed this relationship in vivo. Functionally, CFA and RTCA demonstrated the central role of SKP2 in promoting PD-L1-mediated proliferation. Finally, immunohistochemistry in 74 breast cancer patients confirmed PD-L1 and SKP-p21/p27 axis relationship, as it showed a highly statistically significant correlation between SKP2 and PD-L1 expression (p < 0.001), and both correlated significantly with the proliferation marker Ki-67 (p < 0.001). On the other hand, there was a statistically significant inverse relationship between PD-L1 and p21CIP1/WAF1 expression (p = 0.005). Importantly, double negativity for p21CIP1/WAF1 and p27Kip1 correlated significantly with PD-L1 (p < 0.001), SKP2 (p = 0.002), and Ki-67 (p = 0.002).
    CONCLUSIONS: we have demonstrated the role of the SKP2-p27/p21 axis in intrinsic PD-L1-enhanced cell cycle progression. Inhibitors of SKP2 expression can alleviate resistance to ICPIs.
    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
    泛素化是蛋白质稳定性和功能的关键调节剂。已知多功能蛋白p27在K48连接的泛素化后被蛋白酶体降解。然而,我们最近报道,当泛素结合酶UbcH7(UBE2L3)过表达时,p27稳定,细胞周期在多种不同的细胞类型,包括眼晶状体,视网膜,HEK-293和HELA细胞。但是与p27的这种稳定相关的泛素连接酶仍然是个谜。从体外泛素化筛选开始,我们确定RSP5为酵母E3连接酶伴侣的UbcH7在p27的泛素化。对E3连接酶的同源人NEDD4家族的筛选表明,SMURF1而不是其紧密的同源物SMURF2可以稳定细胞中的p27。我们发现SMURF1在体外用K29O泛素化p27而不是K29R或K63O泛素。表现出对K29链形成的强烈偏好。与SMURF1/UbcH7对p27的稳定一致,我们还发现SMURF1,UbcH7和p27促进细胞迁移,而SMURF1或UbcH7的敲减会降低细胞迁移。我们进一步证明了SMURF1/p27和UbcH7/p27在迁移细胞前缘的共定位。总之,这些结果表明,SMURF1和UbcH7共同在p27上产生K29连接的泛素链,从而稳定p27并促进其细胞周期无关的调节细胞迁移的功能。
    Ubiquitination is a key regulator of protein stability and function. The multifunctional protein p27 is known to be degraded by the proteasome following K48-linked ubiquitination. However, we recently reported that when the ubiquitin-conjugating enzyme UbcH7 (UBE2L3) is overexpressed, p27 is stabilized, and cell cycle is arrested in multiple diverse cell types including eye lens, retina, HEK-293, and HELA cells. However, the ubiquitin ligase associated with this stabilization of p27 remained a mystery. Starting with an in vitro ubiquitination screen, we identified RSP5 as the yeast E3 ligase partner of UbcH7 in the ubiquitination of p27. Screening of the homologous human NEDD4 family of E3 ligases revealed that SMURF1 but not its close homolog SMURF2, stabilizes p27 in cells. We found that SMURF1 ubiquitinates p27 with K29O but not K29R or K63O ubiquitin in vitro, demonstrating a strong preference for K29 chain formation. Consistent with SMURF1/UbcH7 stabilization of p27, we also found that SMURF1, UbcH7, and p27 promote cell migration, whereas knockdown of SMURF1 or UbcH7 reduces cell migration. We further demonstrated the colocalization of SMURF1/p27 and UbcH7/p27 at the leading edge of migrating cells. In sum, these results indicate that SMURF1 and UbcH7 work together to produce K29-linked ubiquitin chains on p27, resulting in the stabilization of p27 and promoting its cell-cycle independent function of regulating cell 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
    原发性甲状旁腺功能亢进伴甲状旁腺肿瘤是多发性内分泌肿瘤1型(MEN1)的典型表现,历史上被称为“原发性增生”。这些肿瘤是否代表多腺克隆疾病或增生至今尚未得到有力证实。Menin蛋白表达的缺失与两个等位基因的失活和MEN1基因突变的良好替代相关。细胞周期蛋白依赖性激酶抑制剂1B(CDKN1B)基因在MEN4中突变,并编码蛋白p27,其表达在综合征MEN1设置中研究甚少。这里,我们分析了25例独立的甲状旁腺腺瘤组织形态学和蛋白表达的Menin和p27,特征良好的MEN1队列。在一个MEN1相关的甲状旁腺腺瘤中,通过荧光原位杂交(FISH)评估了杂合性丢失(LOH)的模式。Further,对4例MEN1患者的11个结节进行了下一代测序(NGS).形态学上,大多数MEN1腺瘤由多个不同的结节组成,其中Menin表达大部分丢失,p27蛋白表达减少。FISH分析显示,大多数结节表现出MEN1丢失,有或没有着丝粒11的损失。NGS证明了亚克隆进化和克隆无关肿瘤的存在。因此,综合征MEN1甲状旁腺腺瘤由多个具有亚克隆的克隆组成,这支持了世卫组织甲状旁腺肿瘤新分类的当前概念(2022年)。p27表达在大部分MEN1甲状旁腺中丢失,因此在提示MEN4时必须谨慎使用。
    Primary hyperparathyroidism with parathyroid tumors is a typical manifestation of Multiple Endocrine Neoplasia Type 1 (MEN1) and is historically termed \"primary hyperplasia\". Whether these tumors represent a multi-glandular clonal disease or hyperplasia has not been robustly proven so far. Loss of Menin protein expression is associated with inactivation of both alleles and a good surrogate for a MEN1 gene mutation. The cyclin-dependent kinase inhibitor 1B (CDKN1B) gene is mutated in MEN4 and encodes for protein p27 whose expression is poorly studied in the syndromic MEN1 setting.Here, we analyzed histomorphology and protein expression of Menin and p27 in parathyroid adenomas of 25 patients of two independent, well-characterized MEN1 cohorts. The pattern of loss of heterozygosity (LOH) was assessed by fluorescence in situ hybridization (FISH) in one MEN1-associated parathyroid adenoma. Further, next-generation sequencing (NGS) was performed on eleven nodules of four MEN1 patients.Morphologically, the majority of MEN1 adenomas consisted of multiple distinct nodules, in which Menin expression was mostly lost and p27 protein expression reduced. FISH analysis revealed that most nodules exhibited MEN1 loss, with or without the loss of centromere 11. NGS demonstrated both subclonal evolution and the existence of clonally unrelated tumors.Syndromic MEN1 parathyroid adenomas therefore consist of multiple clones with subclones, which supports the current concept of the novel WHO classification of parathyroid tumors (2022). p27 expression was lost in a large fraction of MEN1 parathyroids and must therefore be used with caution in suggesting MEN4.
    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
    细胞周期失调是乳腺癌的决定性特征。这里,1-甲基烟酰胺(1-MNA),烟酰胺N-甲基转移酶(NNMT)的代谢物被鉴定,作为乳腺癌细胞周期进展的新驱动因素。NNMT,在乳腺癌组织中高表达,与肿瘤分级呈正相关,TNM阶段,Ki-67指数,和肿瘤大小。NNMT表达的消融显著抑制细胞增殖并导致细胞周期停滞在G0/G1期。这种现象主要源于1-MNA的靶向作用,导致p27蛋白表达的特异性下调。机械上,1-MNA通过增强cullin-1neddylation加速p27蛋白的降解,对于激活Cullin-1-RINGE3泛素连接酶(CRL1)至关重要-一种针对p27蛋白的E3泛素连接酶。NNMT/1-MNA特异性上调UBC12的表达,UBC12是cullin-1neddylation所需的E2NEDD8缀合酶。1-MNA显示出对UBC12的高结合亲和力,通过防止UBC12蛋白定位至溶酶体以降解来延长其半衰期。因此,1-MNA是一种生物活性代谢物,可通过增强neddylation途径介导的p27降解来促进乳腺癌进展。该研究揭示了NNMT酶活性与细胞周期进程之间的联系,提示1-MNA可能参与肿瘤微环境的重塑。
    Cell cycle dysregulation is a defining feature of breast cancer. Here, 1-methyl-nicotinamide (1-MNA), metabolite of nicotinamide N-methyltransferase(NNMT) is identified, as a novel driver of cell-cycle progression in breast cancer. NNMT, highly expressed in breast cancer tissues, positively correlates with tumor grade, TNM stage, Ki-67 index, and tumor size. Ablation of NNMT expression dramatically suppresses cell proliferation and causes cell-cycle arrest in G0/G1 phase. This phenomenon predominantly stems from the targeted action of 1-MNA, resulting in a specific down-regulation of p27 protein expression. Mechanistically, 1-MNA expedites the degradation of p27 proteins by enhancing cullin-1 neddylation, crucial for the activation of Cullin-1-RING E3 ubiquitin ligase(CRL1)-an E3 ubiquitin ligase targeting p27 proteins.  NNMT/1-MNA specifically up-regulates the expression of UBC12, an E2 NEDD8-conjugating enzyme required for cullin-1 neddylation. 1-MNA showes high binding affinity to UBC12, extending the half-life of UBC12 proteins via preventing their localization to lysosome for degradation. Therefore, 1-MNA is a bioactive metabolite that promotes breast cancer progression by reinforcing neddylation pathway-mediated p27 degradation. The study unveils the link between NNMT enzymatic activity with cell-cycle progression, indicating that 1-MNA may be involved in the remodeling of tumor microenvironment.
    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
    氨基酸是细胞生长和增殖所必需的,但目前尚不清楚氨基酸可用性何时以及如何影响增殖-静止决定。这里,我们使用延时显微镜和单细胞追踪细胞周期蛋白依赖性激酶2(CDK2)活性来评估单个细胞对单个氨基酸撤除的反应,并发现不同氨基酸的细胞周期效应显著不同.例如,亮氨酸戒断后,MCF10A细胞完成两个细胞周期,然后进入CDK2低静止期,而赖氨酸戒断会导致细胞周期立即停止。蛋氨酸戒断引发类似于血清饥饿或Mek抑制的限制点表型:在蛋氨酸戒断后,细胞完成当前的细胞周期,并在有丝分裂后进入CDK2低静止期。限制点调节剂p21/p27或细胞周期蛋白D1的调节能够在蛋氨酸和亮氨酸戒断下短期挽救增殖,和较小程度的赖氨酸戒断,揭示了一个连接营养信号和细胞周期进入的检查点。
    Amino acids are required for cell growth and proliferation, but it remains unclear when and how amino acid availability impinges on the proliferation-quiescence decision. Here, we used time-lapse microscopy and single-cell tracking of cyclin-dependent kinase 2 (CDK2) activity to assess the response of individual cells to withdrawal of single amino acids and found strikingly different cell-cycle effects depending on the amino acid. For example, upon leucine withdrawal, MCF10A cells complete two cell cycles and then enter a CDK2-low quiescence, whereas lysine withdrawal causes immediate cell-cycle stalling. Methionine withdrawal triggers a restriction point phenotype similar to serum starvation or Mek inhibition: upon methionine withdrawal, cells complete their current cell cycle and enter a CDK2-low quiescence after mitosis. Modulation of restriction point regulators p21/p27 or cyclin D1 enables short-term rescue of proliferation under methionine and leucine withdrawal, and to a lesser extent lysine withdrawal, revealing a checkpoint connecting nutrient signaling to cell-cycle entry.
    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
    肺动脉高压(PAH)是一种以血管收缩和血管重塑增加为特征的疾病。肺动脉平滑肌细胞(PASMC)高表达转录因子缺氧诱导因子-1α(HIF-1α),然而,PASMCHIF-1α在PAH发展中的作用仍存在争议。研究SMCHIF-1α在急性和慢性低氧肺血管反应中的作用。我们采用了一种获得功能的策略,通过在SM22α表达细胞中产生缺乏脯氨酸羟化酶结构域(PHD)1和2的小鼠来稳定PASMC中的HIF-1α。该策略在常氧和缺氧条件下增加了HIF-1α的表达和转录活性。急性缺氧会增加右心室收缩压(RVSP),但在SM22α-PHD1/2-/-小鼠中没有。与SM22α-PHD1/2-/-小鼠相比,对照SM22α-PHD1/2/-小鼠的慢性缺氧增加了RVSP和血管重塑。体外研究表明,在常氧和低氧条件下,与PHD1/2-/-PASMC相比,分离的PHD1/2/+的收缩性和肌球蛋白轻链磷酸化增加。与SM22α-PHD1/2+/+小鼠相比,SM22α-PHD1/2-/-慢性缺氧后,p27更多,血管重塑更少。从对照组患者中分离出的PASMC中的缺氧增加p27,但不在特发性PAH(IPAH)患者的细胞中。这些发现强调了HIF-1α在抑制肺血管收缩和血管重塑中表达SM22α的细胞特异性作用。调节PASMC中的HIF-1α表达可能代表了PAH患者的有希望的预防和治疗策略。
    Pulmonary arterial hypertension (PAH) is a disease characterized by increased vasoconstriction and vascular remodeling. Pulmonary artery smooth muscle cells (PASMCs) highly express the transcription factor hypoxia-inducible factor-1α (HIF-1α), yet the role of PASMC HIF-1α in the development of PAH remains controversial. To study the role of SMC HIF-1α in the pulmonary vascular response to acute and chronic hypoxia, we used a gain-of-function strategy to stabilize HIF-1α in PASMC by generating mice lacking prolyl hydroxylase domain (PHD) 1 and 2 in SM22α-expressing cells. This strategy increased HIF-1α expression and transcriptional activity under conditions of normoxia and hypoxia. Acute hypoxia increased right ventricular systolic pressure (RVSP) in control, but not in SM22α-PHD1/2-/- mice. Chronic hypoxia increased RVSP and vascular remodeling more in control SM22α-PHD1/2+/+ than in SM22α-PHD1/2-/- mice. In vitro studies demonstrated increased contractility and myosin light chain phosphorylation in isolated PHD1/2+/+ compared with PHD1/2-/- PASMC under both normoxic and hypoxic conditions. After chronic hypoxia, there was more p27 and less vascular remodeling in SM22α-PHD1/2-/- compared with SM22α-PHD1/2+/+ mice. Hypoxia increased p27 in PASMC isolated from control patients, but not in cells from patients with idiopathic pulmonary arterial hypertension (IPAH). These findings highlight an SM22α-expressing cell-specific role for HIF-1α in the inhibition of pulmonary vasoconstriction and vascular remodeling. Modulating HIF-1α expression in PASMC may represent a promising preventative and therapeutic strategy for patients with PAH.NEW & NOTEWORTHY In a mouse model wherein hypoxia-inducible factor 1 alpha (HIF-1α) is stabilized in vascular smooth muscle cells, we found that HIF-1α regulates vasoconstriction by limiting phosphorylation of myosin light chain and regulates vascular remodeling through p27 induction. These findings highlight a cell-specific role for HIF-1α in the inhibition of pulmonary vasoconstriction and vascular remodeling.
    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
    高达50%的肝细胞癌(HCC)是由乙型肝炎病毒(HBV)感染引起的,HBV的表面蛋白对HBV相关HCC的进展至关重要。大HBV表面抗原(LHB)的表达在HBV相关的HCC组织中存在,并且与HCC的发展显着相关。基因集富集分析显示LHB过表达调节细胞周期过程。HCC细胞中过量的LHB诱导慢性内质网(ER)应激,并与体内肿瘤生长显着相关。细胞周期分析表明,从G1期到S期的细胞周期进程在体外得到了极大的增强。我们确定了肝癌中内质网应激和细胞周期进程之间的强烈串扰。作为G1/S检查点的重要调节器,p27被转录因子ATF4和XBP1s转录上调,未折叠蛋白反应途径的下游。此外,LHB诱导的内质网应激促进内部核糖体进入位点介导的p27选择性翻译和E3泛素连接酶HRD1介导的p27泛素化和降解。最终,p27蛋白水平的降低减少了G1/S阻滞,并通过调节细胞周期促进HCC的进展。
    Up to 50% of hepatocellular carcinoma (HCC) is caused by hepatitis B virus (HBV) infection, and the surface protein of HBV is essential for the progression of HBV-related HCC. The expression of large HBV surface antigen (LHB) is presented in HBV-associated HCC tissues and is significantly associated with the development of HCC. Gene set enrichment analysis revealed that LHB overexpression regulates the cell cycle process. Excess LHB in HCC cells induced chronic endoplasmic reticulum (ER) stress and was significantly correlated with tumor growth in vivo. Cell cycle analysis showed that cell cycle progression from G1 to S phase was greatly enhanced in vitro. We identified intensive crosstalk between ER stress and cell cycle progression in HCC. As an important regulator of the G1/S checkpoint, p27 was transcriptionally upregulated by transcription factors ATF4 and XBP1s, downstream of the unfolded protein response pathway. Moreover, LHB-induced ER stress promoted internal ribosome-entry-site-mediated selective translation of p27, and E3 ubiquitin ligase HRD1-mediated p27 ubiquitination and degradation. Ultimately, the decrease in p27 protein levels reduced G1/S arrest and promoted the progress of HCC by regulating the cell cycle.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号