Procollagen-Proline Dioxygenase

前胶原 - 脯氨酸双加氧酶
  • 文章类型: Journal Article
    脯氨酸4-羟化酶2(P4HA2)以其羟化酶活性而闻名,主要参与在生理条件下羟基化胶原前体和促进胶原交联。尽管其过度表达影响多种恶性肿瘤的发生和发展,其在口腔鳞状细胞癌(OSCC)中的具体作用和机制尚不清楚。这项研究的重点是调查表达模式,致癌功能,以及P4HA2在OSCC细胞中的潜在机制。各种数据库,包括TCGA,TIMER,UALCAN,GEPIA,和K-M绘图仪,连同石蜡包埋的样本,用于确定P4HA2在癌症中的表达及其与临床病理特征的相关性。开发了P4HA2敲低和过表达细胞模型以评估其致癌作用和机制。结果表明,P4HA2在OSCC中过表达,与患者生存率呈负相关。敲除P4HA2抑制侵袭,迁移,和OSCC细胞在体外和体内的增殖,而P4HA2的过表达具有相反的作用。机械上,P4HA2沉默后,PI3K/AKT通路的磷酸化水平降低.该研究表明,P4HA2作为预测OSCC预后的有希望的生物标志物,并显著影响转移,入侵,通过PI3K/AKT信号通路调节OSCC细胞的增殖。
    Proline 4-hydroxylase 2 (P4HA2) is known for its hydroxylase activity, primarily involved in hydroxylating collagen precursors and promoting collagen cross-linking under physiological conditions. Although its overexpression influences a wide variety of malignant tumors\' occurrence and development, its specific effects and mechanisms in oral squamous cell carcinoma (OSCC) remain unclear. This study focused on investigating the expression patterns, carcinogenic functions, and underlying mechanisms of P4HA2 in OSCC cells. Various databases, including TCGA, TIMER, UALCAN, GEPIA, and K-M plotter, along with paraffin-embedded samples, were used to ascertain P4HA2 expression in cancer and its correlation with clinicopathological features. P4HA2 knockdown and overexpression cell models were developed to assess its oncogenic roles and mechanisms. The results indicated that P4HA2 was overexpressed in OSCC and inversely correlated with patient survival. Knockdown of P4HA2 suppressed invasion, migration, and proliferation of OSCC cells both in vitro and in vivo, whereas overexpression of P4HA2 had the opposite effects. Mechanistically, the phosphorylation levels of the PI3K/AKT pathway were reduced following P4HA2 silencing. The study reveals that P4HA2 acts as a promising biomarker for predicting prognosis in OSCC and significantly affects metastasis, invasion, and proliferation of OSCC cells through the regulation of the PI3K/AKT signaling pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    热应激(HS)对动物的生长发育构成重大威胁,导致业绩下降和经济损失。肠道系统易受HS的影响,并经历肠道高热和病理性缺氧。缺氧诱导因子-1α(HIF-1α),细胞缺氧适应的关键角色,受脯氨酸-4-羟化酶2(PHD2)和热休克蛋白90(HSP90)的影响。然而,HIF-1α在HS肠道中的综合调控尚不清楚。本研究旨在探讨HS对猪肠道黏膜的影响及HIF-1α的调控机制。将24只从江香猪分为对照组和5只HS处理组(6、12、24、48和72h)。对照组的环境温度和湿度保持在热中性状态(温度-湿度指数(THI)<74),而HS组出现中度HS(78 Heat stress (HS) poses a substantial threat to animal growth and development, resulting in declining performance and economic losses. The intestinal system is susceptible to HS and undergoes intestinal hyperthermia and pathological hypoxia. Hypoxia-inducible factor-1α (HIF-1α), a key player in cellular hypoxic adaptation, is influenced by prolyl-4-hydroxylase 2 (PHD2) and heat shock protein 90 (HSP90). However, the comprehensive regulation of HIF-1α in the HS intestine remains unclear. This study aims to explore the impact of HS on pig intestinal mucosa and the regulatory mechanism of HIF-1α. Twenty-four Congjiang Xiang pigs were divided into the control and five HS-treated groups (6, 12, 24, 48, and 72 h). Ambient temperature and humidity were maintained in a thermally-neutral state (temperature-humidity index (THI) < 74) in the control group, whereas the HS group experienced moderate HS (78 < THI <84). Histological examination revealed villus exfoliation after 12 h of HS in the duodenum, jejunum, and ileum, with increasing damage as HS duration extended. The villus height to crypt depth ratio (V/C) decreased and goblet cell number increased with prolonged HS. Quantitative real-time PCR, Western blot, and immunohistochemistry analysis indicated increased expression of HIF-1α and HSP90 in the small intestine with prolonged HS, whereas PHD2 expression decreased. Further investigation in IPEC-J2 cells subjected to HS revealed that overexpressing PHD2 increased PHD2 mRNA and protein expression, while it decreases HIF-1α. Conversely, interfering with HSP90 expression substantially decreased both HSP90 and HIF-1α mRNA and protein levels. These results suggest that HS induces intestinal hypoxia with concomitant small intestinal mucosal damage. The expression of HIF-1α in HS-treated intestinal epithelial cells may be co-regulated by HSP90 and PHD2 and is possibly linked to intestinal hyperthermia and hypoxia.
    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
    脯氨酸4-羟化酶(P4Hs)是胶原蛋白合成中的关键修饰酶家族。P4Hs已被证实与肿瘤的发生发展密切相关。然而,P4Hs在头颈部恶性肿瘤(HNSC)中的表达及其与预后和肿瘤免疫浸润的关系尚未被分析。我们研究了转录表达,生存数据,来自多个数据库的HNSC患者中P4Hs的免疫浸润。P4H1-3在HNSC肿瘤组织中的表达明显高于正常组织。此外,P4HA1和P4HA2与肿瘤分期相关,患者预后,和免疫细胞浸润。P4HA3与患者预后及免疫细胞浸润有关。相关实验证实,P4HA1可能作为预后标志物,在鼻咽癌的进展中发挥作用。这些结果表明,P4HA1-3可能是HNSC预后和治疗的新生物标志物。有望支持开发头颈部肿瘤患者的新疗法,并改善患者的预后。
    Prolyl 4-hydroxylases (P4Hs) are a family of key modifying enzymes in collagen synthesis. P4Hs have been confirmed to be closely associated with tumor occurrence and development. However, the expression of P4Hs in head and neck cancer (HNSC) as well as its relationship with prognosis and tumor immunity infiltration has not yet been analyzed. We investigated the transcriptional expression, survival data, and immune infiltration of P4Hs in patients with HNSC from multiple databases. P4H1-3 expression was significantly higher in HNSC tumor tissues than in normal tissues. Moreover, P4HA1 and P4HA2 were associated with tumor stage, patient prognosis, and immune cell infiltration. P4HA3 was related to patient prognosis and immune cell infiltration. Correlation experiments confirmed that P4HA1 may serve as a prognosis biomarker and plays a role in the progression of nasopharyngeal carcinoma. These findings suggest that P4HA1-3 may be a novel biomarker for the prognosis and treatment of HNSC, which is expected to support the development of new therapies for patients with head and neck tumors and improve patient outcomes.
    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
    细胞代谢重新编程以维持能量生产,而减少氧气和能量消耗过程对于适应缺氧/缺血至关重要。适应性代谢重新布线受缺氧诱导因子(HIF)控制。越来越多的实验证据表明,及时激活大脑驻留细胞中的HIF可改善急性缺血性中风的预后。然而,潜在的分子机制仍未完全理解。因此,我们调查了HIF依赖性代谢重编程是否影响脑驻留细胞对缺血性应激的脆弱性.方法:我们使用遗传和药理学方法激活体内鼠脑中以及体外原代神经元和星形胶质细胞中的HIF。许多代谢组学方法和分子生物学技术被用于阐明对脑细胞中心碳代谢的潜在HIF依赖性作用。在缺血性中风的动物和细胞模型中,我们分析了HIF依赖性代谢重编程是否影响缺血性损伤的易感性.结果:脯氨酸-4-羟化酶结构域2(PHD2)蛋白的神经元特异性基因消融,以氧依赖的方式负调节HIF-α的蛋白质稳定性,以HIF依赖性方式减少急性中风后小鼠的脑损伤和功能损害。因此,PHD2缺陷型神经元在体外表现出改善的对缺血应激的耐受性,伴随着通过丙酮酸脱氢酶激酶介导的丙酮酸脱氢酶抑制HIF-1介导的糖酵解乳酸产生的增强。用罗沙司他对小鼠进行系统治疗,一种低分子量的泛PHD抑制剂,不仅增加了许多代谢产物的中央碳和氨基酸代谢鼠脑的丰度,而且还可以改善急性缺血性卒中后的脑组织损伤和感觉运动功能障碍。在神经元和星形胶质细胞中,roxadustat引起HIF-1依赖性葡萄糖代谢重编程,包括葡萄糖摄取升高,糖原合成,糖酵解能力,乳酸产生和乳酸释放,增强了星形胶质细胞的缺血耐受性,但不是神经元。我们发现,通过非选择性抑制所有PHD同工酶对神经元中HIF-1的强烈激活导致6-磷酸果糖-2-激酶/果糖-2,6-双磷酸酶-3的HIF-1依赖性上调,将葡萄糖-6-磷酸从戊糖磷酸途径(PPP)重定向到糖酵解途径。这伴随着PPP中NADPH产量的减少,这进一步降低了神经元的内在抗氧化储备,让他们更容易受到缺血性压力的影响.尽管如此,在保留神经元-神经胶质相互作用的器官型海马培养物中,roxadustat降低了神经元对缺血性应激的易感性,通过乳酸转运阻滞限制糖酵解能量的产生在很大程度上阻止了这种情况。结论:集体,我们的结果表明,HIF-1介导的代谢重编程减轻了脑内细胞对缺血性应激的内在脆弱性.
    Cell metabolism reprogramming to sustain energy production, while reducing oxygen and energy consuming processes is crucially important for the adaptation to hypoxia/ischemia. Adaptive metabolic rewiring is controlled by hypoxia-inducible factors (HIFs). Accumulating experimental evidence indicates that timely activation of HIF in brain-resident cells improves the outcome from acute ischemic stroke. However, the underlying molecular mechanisms are still incompletely understood. Thus, we investigated whether HIF-dependent metabolic reprogramming affects the vulnerability of brain-resident cells towards ischemic stress. Methods: We used genetic and pharmacological approaches to activate HIF in the murine brain in vivo and in primary neurons and astrocytes in vitro. Numerous metabolomic approaches and molecular biological techniques were applied to elucidate potential HIF-dependent effects on the central carbon metabolism of brain cells. In animal and cell models of ischemic stroke, we analysed whether HIF-dependent metabolic reprogramming influences the susceptibility to ischemic injury. Results: Neuron-specific gene ablation of prolyl-4-hydroxylase domain 2 (PHD2) protein, negatively regulating the protein stability of HIF-α in an oxygen dependent manner, reduced brain injury and functional impairment of mice after acute stroke in a HIF-dependent manner. Accordingly, PHD2 deficient neurons showed an improved tolerance towards ischemic stress in vitro, which was accompanied by enhanced HIF-1-mediated glycolytic lactate production through pyruvate dehydrogenase kinase-mediated inhibition of the pyruvate dehydrogenase. Systemic treatment of mice with roxadustat, a low-molecular weight pan-PHD inhibitor, not only increased the abundance of numerous metabolites of the central carbon and amino acid metabolism in murine brain, but also ameliorated cerebral tissue damage and sensorimotor dysfunction after acute ischemic stroke. In neurons and astrocytes roxadustat provoked a HIF-1-dependent glucose metabolism reprogramming including elevation of glucose uptake, glycogen synthesis, glycolytic capacity, lactate production and lactate release, which enhanced the ischemic tolerance of astrocytes, but not neurons. We found that strong activation of HIF-1 in neurons by non-selective inhibition of all PHD isoenzymes caused a HIF-1-dependent upregulation of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 redirecting glucose-6-phosphate from pentose phosphate pathway (PPP) to the glycolysis pathway. This was accompanied by a reduction of NADPH production in the PPP, which further decreased the low intrinsic antioxidant reserve of neurons, making them more susceptible to ischemic stress. Nonetheless, in organotypic hippocampal cultures with preserved neuronal-glial interactions roxadustat decreased the neuronal susceptibility to ischemic stress, which was largely prevented by restricting glycolytic energy production through lactate transport blockade. Conclusion: Collectively, our results indicate that HIF-1-mediated metabolic reprogramming alleviates the intrinsic vulnerability of brain-resident cells to ischemic stress.
    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
    哺乳动物雷帕霉素靶蛋白(mTOR)激酶是细胞生长和代谢的中枢调节因子,其复合物mTORC1和mTORC2磷酸化不同的底物。mTOR信号的失调通常与人类疾病有关。包括癌症.尽管对mTOR进行了三十年的积极研究,还有很多事情有待确定。这里,我们证明了脯氨酸4-羟化酶α-2(P4HA2)直接结合mTOR和羟基化一个高度保守的脯氨酸2341(P2341)在mTOR的激酶域内,从而激活mTOR激酶和下游效应蛋白(例如S6K和AKT)。此外,P2341的羟基化增强了mTOR的稳定性,并使mTOR能够准确识别其底物,例如S6K和AKT。与过表达mTORWT的细胞相比,过表达mTORP2341A的肺腺癌细胞的生长显着降低。有趣的是,体内细胞生长测定显示靶向P4HA2-mTOR显著抑制肺腺癌细胞生长。总之,我们的研究揭示了一种未发现的羟基化调节机制,P4HA2通过该机制直接激活mTOR激酶,为治疗靶向mTOR激酶驱动的癌症提供见解。
    Mammalian target of rapamycin (mTOR) kinase functions as a central regulator of cell growth and metabolism, and its complexes mTORC1 and mTORC2 phosphorylate distinct substrates. Dysregulation of mTOR signaling is commonly implicated in human diseases, including cancer. Despite three decades of active research in mTOR, much remains to be determined. Here, we demonstrate that prolyl 4-hydroxylase alpha-2 (P4HA2) binds directly to mTOR and hydroxylates one highly conserved proline 2341 (P2341) within a kinase domain of mTOR, thereby activating mTOR kinase and downstream effector proteins (e.g. S6K and AKT). Moreover, the hydroxylation of P2341 strengthens mTOR stability and allows mTOR to accurately recognize its substrates such as S6K and AKT. The growth of lung adenocarcinoma cells overexpressing mTORP2341A is significantly reduced when compared with that of cells overexpressing mTORWT. Interestingly, in vivo cell growth assays show that targeting P4HA2-mTOR significantly suppresses lung adenocarcinoma cell growth. In summary, our study reveals an undiscovered hydroxylation-regulatory mechanism by which P4HA2 directly activates mTOR kinase, providing insights for therapeutically targeting mTOR kinase-driven cancers.
    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:脯氨酸羟化酶结构域2(PHD2),由Egln1基因编码,作为缺氧诱导因子(HIF)途径的关键调节剂,并充当细胞氧传感器。小鼠体内PHD2的体细胞失活导致红细胞增多症和充血性心力衰竭。然而,由于PHD2缺乏症的胚胎致死性,它在发展中的作用仍然难以捉摸。这里,我们研究了两个egln1同源基因的功能,egln1a和egln1b,斑马鱼。
    方法:使用CRISPR/Cas9系统产生egln1无效斑马鱼。采用实时定量PCR和Westernblot分析检测egln1缺乏对缺氧信号通路的影响。通过分析心率评估egln1突变斑马鱼的缺氧反应,g搅动频率,和血流速度。随后,采用邻茴香胺染色和原位杂交技术研究egln1在斑马鱼造血功能中的作用。
    结果:我们的数据表明,单独损失egln1a或egln1b对生长速率没有明显影响。然而,egln1a;egln1b双突变体在约2.5个月大时表现出明显的生长迟缓和死亡率升高。egln1a-null和egln1b-null斑马鱼胚胎对缺氧的耐受性均增强,全身缺氧反应,包括Hif通路激活,心脏活动增加,和红细胞增多症。
    结论:我们的研究将斑马鱼egln1突变体作为PHD2的第一个先天性胚胎有活力的系统性脊椎动物动物模型,为缺氧信号和PHD2相关疾病的进展提供了新的见解。
    OBJECTIVE: Prolyl hydroxylase domain 2 (PHD2), encoded by the Egln1 gene, serves as a pivotal regulator of the hypoxia-inducible factor (HIF) pathway and acts as a cellular oxygen sensor. Somatic inactivation of Phd2 in mice results in polycythemia and congestive heart failure. However, due to the embryonic lethality of Phd2 deficiency, its role in development remains elusive. Here, we investigated the function of two egln1 paralogous genes, egln1a and egln1b, in zebrafish.
    METHODS: The egln1 null zebrafish were generated using the CRISPR/Cas9 system. Quantitative real-time PCR assays and Western blot analysis were employed to detect the effect of egln1 deficiency on the hypoxia signaling pathway. The hypoxia response of egln1 mutant zebrafish were assessed by analyzing heart rate, gill agitation frequency, and blood flow velocity. Subsequently, o-dianisidine staining and in situ hybridization were used to investigate the role of egln1 in zebrafish hematopoietic function.
    RESULTS: Our data show that the loss of egln1a or egln1b individually has no visible effects on growth rate. However, the egln1a; egln1b double mutant displayed significant growth retardation and elevated mortality at around 2.5 months old. Both egln1a-null and egln1b-null zebrafish embryo exhibited enhanced tolerance to hypoxia, systemic hypoxic response that include hif pathway activation, increased cardiac activity, and polycythemia.
    CONCLUSIONS: Our research introduces zebrafish egln1 mutants as the first congenital embryonic viable systemic vertebrate animal model for PHD2, providing novel insights into hypoxic signaling and the progression of PHD2- associated disease.
    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
    帕卡克-庄综合征是由EPAS1基因突变引起的,它编码三种缺氧诱导因子α(HIFα)旁系同源物HIF2α之一,并与明确但变化的表型表现有关,包括神经内分泌肿瘤和红细胞增多症。然而,复杂的基因型-表型相关性的潜在机制仍未完全了解.这里,我们设计了一种定量方法,使用微尺度热电泳(MST)确定含有疾病相关突变的HIF2α肽的解离常数(Kd),并显示神经内分泌相关的1类HIF2α突变体的Kd明显高于仅与红细胞增多症相关的2类HIF2α突变体。基于PHD2/HIF2α肽复合物在1.8µ分辨率下的共晶结构,我们表明,1类突变残基位于HIF2α和PHD2之间的关键界面,邻近PHD2活性催化位点,而2类突变的残基定位于HIF2α的更柔性的区域,该区域与PHD2的接触较少。和谐地,与2类对应物相比,发现1类突变显着增加了HIF2α介导的纤维素转录激活。这些结果揭示了一种结构机制,其中HIF2α与PHD2之间的相互作用强度是在Pacak-Zhong综合征中观察到的一般基因型-表型相关性的根源。
    Pacak-Zhuang syndrome is caused by mutations in the EPAS1 gene, which encodes for one of the three hypoxia-inducible factor alpha (HIFα) paralogs HIF2α and is associated with defined but varied phenotypic presentations including neuroendocrine tumors and polycythemia. However, the mechanisms underlying the complex genotype-phenotype correlations remain incompletely understood. Here, we devised a quantitative method for determining the dissociation constant (Kd) of the HIF2α peptides containing disease-associated mutations and the catalytic domain of prolyl-hydroxylase (PHD2) using microscale thermophoresis (MST) and showed that neuroendocrine-associated Class 1 HIF2α mutants have distinctly higher Kd than the exclusively polycythemia-associated Class 2 HIF2α mutants. Based on the co-crystal structure of PHD2/HIF2α peptide complex at 1.8 Å resolution, we showed that the Class 1 mutated residues are localized to the critical interface between HIF2α and PHD2, adjacent to the PHD2 active catalytic site, while Class 2 mutated residues are localized to the more flexible region of HIF2α that makes less contact with PHD2. Concordantly, Class 1 mutations were found to significantly increase HIF2α-mediated transcriptional activation in cellulo compared to Class 2 counterparts. These results reveal a structural mechanism in which the strength of the interaction between HIF2α and PHD2 is at the root of the general genotype-phenotype correlations observed in Pacak-Zhuang syndrome.
    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
    金属离子结合与硫醇蛋白活性之间的相互作用,特别是在蛋白质二硫键异构酶家族中,由于这些蛋白质在许多重要过程中发挥关键作用,因此仍然是活跃研究的领域。这项研究调查了重组人PDIA1与锌离子之间的相互作用,重点关注PDIA1的构象稳定性和酶活性的后续含义。采用等温滴定量热法和差示扫描量热法,我们系统地比较了PDIA1的氧化和还原形式的锌结合能力,并评估了这种相互作用的结构后果.我们的结果表明,PDIA1可以在还原和氧化态结合锌,但在还原形式的PDIA1中具有明显不同的化学计量和更明显的构象效应。此外,观察到锌结合抑制还原PDIA1的催化活性,这可能是由于其构象的诱导改变。这些发现揭示了PDIA1中的潜在调节机制,其中在还原条件下的金属离子结合调节其活性。我们的研究强调了锌在通过构象调节PDIA1的催化功能中的潜在作用,表明在细胞氧化还原调节的更广泛背景下,金属结合和蛋白质稳定性之间存在微妙的相互作用。
    The interplay between metal ion binding and the activity of thiol proteins, particularly within the protein disulfide isomerase family, remains an area of active investigation due to the critical role that these proteins play in many vital processes. This research investigates the interaction between recombinant human PDIA1 and zinc ions, focusing on the subsequent implications for PDIA1\'s conformational stability and enzymatic activity. Employing isothermal titration calorimetry and differential scanning calorimetry, we systematically compared the zinc binding capabilities of both oxidized and reduced forms of PDIA1 and assessed the structural consequences of this interaction. Our results demonstrate that PDIA1 can bind zinc both in reduced and oxidized states, but with significantly different stoichiometry and more pronounced conformational effects in the reduced form of PDIA1. Furthermore, zinc binding was observed to inhibit the catalytic activity of reduced-PDIA1, likely due to induced alterations in its conformation. These findings unveil a potential regulatory mechanism in PDIA1, wherein metal ion binding under reductive conditions modulates its activity. Our study highlights the potential role of zinc in regulating the catalytic function of PDIA1 through conformational modulation, suggesting a nuanced interplay between metal binding and protein stability in the broader context of cellular redox regulation.
    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
    VonHippel-Lindau(VHL)蛋白,在透明细胞肾细胞癌(ccRCC)中经常发生突变,是参与氧化应激的缺氧诱导因子(HIF)的主要调节因子。然而,VHL是否具有不依赖HIF的肿瘤抑制活性尚不清楚.这里,我们证明VHL抑制营养应激诱导的自噬,散发性ccRCC标本中其缺乏与自噬水平显著升高有关,并与患者预后较差相关。机械上,VHL在其PHD1介导的Pro54上的羟基化后直接与自噬调节因子Beclin1结合。这种结合抑制了Beclin1-VPS34复合物与ATG14L的缔合,从而抑制对营养缺乏反应的自噬启动。非羟基化Beclin1P54A的表达消除了VHL介导的自噬抑制,并显着降低了VHL的肿瘤抑制作用。此外,在野生型表达VHL的人ccRCC标本中,Beclin1P54-OH水平与自噬水平呈负相关,患者预后差。此外,用自噬抑制剂和HIF2α抑制剂联合治疗VHL缺陷小鼠肿瘤可抑制肿瘤生长。这些发现揭示了VHL抑制肿瘤生长的一个意想不到的机制,并建议通过联合抑制自噬和HIF2α来治疗ccRCC。
    The Von Hippel-Lindau (VHL) protein, which is frequently mutated in clear-cell renal cell carcinoma (ccRCC), is a master regulator of hypoxia-inducible factor (HIF) that is involved in oxidative stresses. However, whether VHL possesses HIF-independent tumor-suppressing activity remains largely unclear. Here, we demonstrate that VHL suppresses nutrient stress-induced autophagy, and its deficiency in sporadic ccRCC specimens is linked to substantially elevated levels of autophagy and correlates with poorer patient prognosis. Mechanistically, VHL directly binds to the autophagy regulator Beclin1, after its PHD1-mediated hydroxylation on Pro54. This binding inhibits the association of Beclin1-VPS34 complexes with ATG14L, thereby inhibiting autophagy initiation in response to nutrient deficiency. Expression of non-hydroxylatable Beclin1 P54A abrogates VHL-mediated autophagy inhibition and significantly reduces the tumor-suppressing effect of VHL. In addition, Beclin1 P54-OH levels are inversely correlated with autophagy levels in wild-type VHL-expressing human ccRCC specimens, and with poor patient prognosis. Furthermore, combined treatment of VHL-deficient mouse tumors with autophagy inhibitors and HIF2α inhibitors suppresses tumor growth. These findings reveal an unexpected mechanism by which VHL suppresses tumor growth, and suggest a potential treatment for ccRCC through combined inhibition of both autophagy and HIF2α.
    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:血管生成对于缺血性疾病的组织修复至关重要,依靠糖酵解作为其主要能源。脯氨酸4-羟化酶亚基α1(P4HA1),胶原蛋白4-羟化酶的催化亚基,是癌症中的糖酵解相关基因。然而,其在糖酵解诱导的血管生成中的作用尚不清楚.
    方法:使用腺病毒调节P4HA1表达。通过5-乙炔基-2'-脱氧尿苷掺入评估内皮血管生成,Transwell迁移,和体外试管形成测定。体内实验测量了后肢缺血(HLI)模型中的血流量和毛细血管密度。糖酵解应激测定,葡萄糖摄取,乳酸生产,和定量逆转录聚合酶链反应(RT-PCR)用于评估糖酵解能力。转录组测序,通过蛋白质印迹和RT-PCR验证,被用来确定潜在的机制。
    结果:在缺氧和HLI模型中,P4HA1在内皮细胞中上调。P4HA1过表达促进体外和体内血管生成,而它的击倒却产生了相反的效果。P4HA1过表达通过在胶原羟基化过程中消耗α-KG来降低细胞α-酮戊二酸(α-KG)水平。α-KG的下调降低了DNA双加氧酶的蛋白质水平,十-十一易位2(TET2),及其对果糖-1,6-双磷酸酶(FBP1)启动子的募集,导致FBP1表达降低。FBP1的减少增强了糖酵解代谢,从而促进内皮血管生成。
    结论:低氧诱导的内皮P4HA1过表达通过P4HA1/α-KG/TET2/FBP1途径促进糖酵解代谢重编程而增强血管生成。这项研究的发现强调了P4HA1在缺血后血管生成中的意义,提示其治疗缺血后组织修复的潜力。
    Angiogenesis is essential for tissue repair in ischemic diseases, relying on glycolysis as its primary energy source. Prolyl 4-hydroxylase subunit alpha 1 (P4HA1), the catalytic subunit of collagen prolyl 4-hydroxylase, is a glycolysis-related gene in cancers. However, its role in glycolysis-induced angiogenesis remains unclear.
    P4HA1 expression was modulated using adenoviruses. Endothelial angiogenesis was evaluated through 5-ethynyl-2\'-deoxyuridine incorporation, transwell migration, and tube formation assays in vitro. In vivo experiments measured blood flow and capillary density in the hindlimb ischemia (HLI) model. Glycolytic stress assays, glucose uptake, lactate production, and quantitative reverse transcription-polymerase chain reaction (RT-PCR) were employed to assess glycolytic capacity. Transcriptome sequencing, validated by western blotting and RT-PCR, was utilized to determine underlying mechanisms.
    P4HA1 was upregulated in endothelial cells under hypoxia and in the HLI model. P4HA1 overexpression promoted angiogenesis in vitro and in vivo, while its knockdown had the opposite effect. P4HA1 overexpression reduced cellular α-ketoglutarate (α-KG) levels by consuming α-KG during collagen hydroxylation. Downregulation of α-KG reduced the protein level of a DNA dioxygenase, ten-eleven translocation 2 (TET2), and its recruitment to the fructose-1,6-biphosphatase (FBP1) promoter, resulting in decreased FBP1 expression. The decrease in FBP1 enhanced glycolytic metabolism, thereby promoting endothelial angiogenesis.
    Hypoxia-induced endothelial P4HA1 overexpression enhanced angiogenesis by promoting glycolytic metabolism reprogramming through the P4HA1/α-KG/TET2/FBP1 pathway. The study\'s findings underscore the significance of P4HA1 in post-ischemic angiogenesis, suggesting its therapeutic potential for post-ischemic tissue repair.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号