Procollagen-Proline Dioxygenase

前胶原 - 脯氨酸双加氧酶
  • 文章类型: Journal Article
    感觉到内质网(ER)钙(Ca2+)的降低,STIM1介导了一种普遍存在的Ca2流入过程,称为存储操作的Ca2进入(SOCE)。STIM1功能失调或SOCE异常与自身免疫性疾病密切相关。动脉粥样硬化,和各种形式的癌症。因此,揭示翻译后修饰的分子复杂性,如氧化,STIM1的功能至关重要。在最近的蛋白质组学筛查中,我们鉴定了三种蛋白质二硫键异构酶(PDIs)-脯氨酸4-羟化酶亚基β(P4HB),蛋白质二硫键异构酶A3(PDIA3),和含硫氧还蛋白结构域的蛋白5(TXNDC5)-作为STIM1的ER腔相互作用物。这里,我们证明了这些PDI与STIM1和STIM2动态关联。STIM1(STIM1-2CA)的两个保守半胱氨酸残基的突变降低了其在细胞和原位的Ca2亲和力。PDIA3或P4HB的敲除增加了野生型STIM1的Ca2亲和力,同时对STIM1-2CA突变体没有影响,表明PDIA3和P4HB通过作用于ER-管腔半胱氨酸残基来调节STIM1的Ca2+亲和力。这种STIM1的Ca2+敏感性的调制通过Ca2+成像实验进一步证实,这表明,这两个PDI的敲低不会影响STIM1介导的SOCE在完全存储耗尽时,但会导致SOCE振幅在部分存储耗尽时增强。因此,P4HB和PDIA3通过微调其Ca2结合亲和力动态调节STIM1激活,调节激活的STIM1水平以响应生理线索。本文报道的STIM1介导的Ca2+信号传导和氧化还原反应之间的协调可能对细胞生理学和病理学有影响。
    Sensing the lowering of endoplasmic reticulum (ER) calcium (Ca2+), STIM1 mediates a ubiquitous Ca2+ influx process called the store-operated Ca2+ entry (SOCE). Dysregulated STIM1 function or abnormal SOCE is strongly associated with autoimmune disorders, atherosclerosis, and various forms of cancers. Therefore, uncovering the molecular intricacies of post-translational modifications, such as oxidation, on STIM1 function is of paramount importance. In a recent proteomic screening, we identified three protein disulfide isomerases (PDIs)-Prolyl 4-hydroxylase subunit beta (P4HB), protein disulfide-isomerase A3 (PDIA3), and thioredoxin domain-containing protein 5 (TXNDC5)-as the ER-luminal interactors of STIM1. Here, we demonstrated that these PDIs dynamically associate with STIM1 and STIM2. The mutation of the two conserved cysteine residues of STIM1 (STIM1-2CA) decreased its Ca2+ affinity both in cellulo and in situ. Knockdown of PDIA3 or P4HB increased the Ca2+ affinity of wild-type STIM1 while showing no impact on the STIM1-2CA mutant, indicating that PDIA3 and P4HB regulate STIM1\'s Ca2+ affinity by acting on ER-luminal cysteine residues. This modulation of STIM1\'s Ca2+ sensitivity was further confirmed by Ca2+ imaging experiments, which showed that knockdown of these two PDIs does not affect STIM1-mediated SOCE upon full store depletion but leads to enhanced SOCE amplitudes upon partial store depletion. Thus, P4HB and PDIA3 dynamically modulate STIM1 activation by fine-tuning its Ca2+ binding affinity, adjusting the level of activated STIM1 in response to physiological cues. The coordination between STIM1-mediated Ca2+ signaling and redox responses reported herein may have implications for cell physiology and pathology.
    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
    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
    Hedgehog(Hh)信号通路中的畸变在各种癌症中明显普遍存在,包括B细胞淋巴瘤.Hh信号转导的一个关键方面涉及动态调节驱动蛋白家族成员7(KIF7)支持的睫状尖隔室中融合同源物(SUFU)-神经胶质瘤相关癌基因同源物(GLI)复合物的抑制剂。然而,在这种情况下,SUFU-GLI复合体的特定翻译后修饰在很大程度上仍未被探索.我们的研究揭示了一种新的调节机制,涉及到4-羟化酶2(P4HA2),它与KIF7形成复合物,对于Hh途径的信号转导至关重要。我们证明,在Hh通路激活后,P4HA2沿着KIF7重新定位到睫状尖端。这里,它羟基化SUFU以抑制其功能,从而放大Hh信号。此外,P4HA2的缺失显著阻碍了B淋巴瘤的进展.这种作用可以归因于基质成纤维细胞中Hh信号的抑制,导致B淋巴瘤细胞恶性增殖所必需的生长因子减少。我们的发现强调了P4HA2介导的羟化在调节Hh信号传导中的作用,并提出了一种新的基质靶向治疗B细胞淋巴瘤的策略。
    Aberrations in the Hedgehog (Hh) signaling pathway are significantly prevailed in various cancers, including B-cell lymphoma. A critical facet of Hh signal transduction involves the dynamic regulation of the suppressor of fused homolog (SUFU)-glioma-associated oncogene homolog (GLI) complex within the kinesin family member 7 (KIF7)-supported ciliary tip compartment. However, the specific post-translational modifications of SUFU-GLI complex within this context have remained largely unexplored. Our study reveals a novel regulatory mechanism involving prolyl 4-hydroxylase 2 (P4HA2), which forms a complex with KIF7 and is essential for signal transduction of Hh pathway. We demonstrate that, upon Hh pathway activation, P4HA2 relocates alongside KIF7 to the ciliary tip. Here, it hydroxylates SUFU to inhibit its function, thus amplifying the Hh signaling. Moreover, the absence of P4HA2 significantly impedes B lymphoma progression. This effect can be attributed to the suppression of Hh signaling in stromal fibroblasts, resulting in decreased growth factors essential for malignant proliferation of B lymphoma cells. Our findings highlight the role of P4HA2-mediated hydroxylation in modulating Hh signaling and propose a novel stromal-targeted therapeutic strategy for B-cell lymphoma.
    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
    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
    氧气传感器使细胞能够适应有限的氧气可用性(缺氧),影响各种细胞和组织反应。脯氨酸基-4-羟化酶结构域1-3(PHD1-3;也称为Egln1-3,HIF-P4H1-3,HIF-PH1-3)蛋白属于Fe2-和2-酮戊二酸依赖性双加氧酶超家族,并利用分子氧(O2)与2-酮戊二酸作为共同底物,以诱导亚单位的两个脯氨酸残基的H-二聚体因子(缺氧)PHD1-3介导的HIF-α的羟基化导致其降解和失活。最近,各种PHD抑制剂(PHI)已进入诊所治疗肾性贫血。临床前分析表明,PHI治疗也可能对许多其他缺氧相关疾病有益。尽管如此,所观察到的PHIs保护作用的潜在分子机制仅被部分理解,目前阻碍他们翻译成诊所。此外,PHI介导的Epo水平增加在所有缺氧相关疾病中并非有益,而PHD选择性抑制可能是有利的。这里,我们总结了目前关于三种PHD同工型在体内的相关性和功能的知识,基于啮齿动物中每个单个相应基因的缺失或RNA干扰介导的敲减。这些信息对于我们理解PHD的生理相关性和功能以及阐明其对缺氧相关疾病的个体影响至关重要。此外,这些知识强调了在获得PHD同工型选择性抑制剂的情况下,哪些疾病可以最好地靶向这些药物.
    Oxygen sensors enable cells to adapt to limited oxygen availability (hypoxia), affecting various cellular and tissue responses. Prolyl-4-hydroxylase domain 1-3 (PHD1-3; also called Egln1-3, HIF-P4H 1-3, HIF-PH 1-3) proteins belong to the Fe2+- and 2-oxoglutarate-dependent dioxygenase superfamily and utilise molecular oxygen (O2) alongside 2-oxoglutarate as co-substrate to hydroxylate two proline residues of α subunits of the dimeric hypoxia inducible factor (HIF) transcription factor. PHD1-3-mediated hydroxylation of HIF-α leads to its degradation and inactivation. Recently, various PHD inhibitors (PHI) have entered the clinics for treatment of renal anaemia. Pre-clinical analyses indicate that PHI treatment may also be beneficial in numerous other hypoxia-associated diseases. Nonetheless, the underlying molecular mechanisms of the observed protective effects of PHIs are only partly understood, currently hindering their translation into the clinics. Moreover, the PHI-mediated increase of Epo levels is not beneficial in all hypoxia-associated diseases and PHD-selective inhibition may be advantageous. Here, we summarise the current knowledge about the relevance and function of each of the three PHD isoforms in vivo, based on the deletion or RNA interference-mediated knockdown of each single corresponding gene in rodents. This information is crucial for our understanding of the physiological relevance and function of the PHDs as well as for elucidating their individual impact on hypoxia-associated diseases. Furthermore, this knowledge highlights which diseases may best be targeted by PHD isoform-selective inhibitors in case such pharmacologic substances become available.
    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:缺氧是大多数癌症的病理标志,包括胶质母细胞瘤(GBM)。癌基因蛋白的缺氧信号激活和翻译后修饰(PTM)在癌症中得到了充分研究。积累的研究表明,糖酵解酶PGK1在肿瘤发生中起着至关重要的作用,然而潜在的机制仍然未知。
    方法:我们首先使用ChIP测定法来揭示HIF1α和ATF3之间的串扰及其在P4HA1调节中的作用。蛋白质降解分析,LC-MS/MS,进行了体外琥珀酸生产测定以检查蛋白质琥珀酰化对GBM病理的影响。海马测定法测量了PGK1琥珀酰化在K191/192或其突变体对葡萄糖代谢的影响。我们利用体内颅内小鼠模型进行生化研究,以阐明ATF3和P4HA1对有氧糖酵解和肿瘤免疫微环境的影响。
    结果:我们证明了HIF1α和ATF3分别正向和负向调节P4HA1的转录,导致琥珀酸产生增加和HIF1α信号激活增加。P4HA1表达升高了琥珀酸浓度,导致PGK1在K191和K192位点的琥珀酰化增强。通过琥珀酰化抑制PGK1的蛋白酶体降解显著增加了产生乳酸的有氧糖酵解。此外,ATF3过表达和P4HA1敲低降低GBM细胞中琥珀酸和乳酸水平,抑制免疫反应和肿瘤生长。
    结论:一起,我们的研究表明HIF1α/ATF3参与了P4HA1/琥珀酸盐信号传导,它是GBM中K191和K192位点琥珀酸酯生物合成和PGK1琥珀酰化的主要调节剂。P4HA1/琥珀酸途径可能是GBM有氧糖酵解的新靶标。
    BACKGROUND: Hypoxia is a pathological hallmark in most cancers, including glioblastoma (GBM). Hypoxic signaling activation and post-translational modification (PTM) of oncogenic proteins are well-studied in cancers. Accumulating studies indicate glycolytic enzyme PGK1 plays a crucial role in tumorigenesis, yet the underlying mechanisms remain unknown.
    METHODS: We first used ChIP assays to uncover the crosstalk between HIF1α and ATF3 and their roles in P4HA1 regulation. Protein degradation analysis, LC-MS/MS, and in vitro succinate production assays were performed to examine the effect of protein succinylation on GBM pathology. Seahorse assay measured the effects of PGK1 succinylation at K191/K192 or its mutants on glucose metabolism. We utilized an in vivo intracranial mouse model for biochemical studies to elucidate the impact of ATF3 and P4HA1 on aerobic glycolysis and the tumor immune microenvironment.
    RESULTS: We demonstrated that HIF1α and ATF3 positively and negatively regulate the transcription of P4HA1, respectively, leading to an increased succinate production and increased activation of HIF1α signaling. P4HA1 expression elevated the succinate concentration, resulting in the enhanced succinylation of PGK1 at the K191 and K192 sites. Inhibition of proteasomal degradation of PGK1 by succinylation significantly increased aerobic glycolysis to generate lactate. Furthermore, ATF3 overexpression and P4HA1 knockdown reduced succinate and lactate levels in GBM cells, inhibiting immune responses and tumor growth.
    CONCLUSIONS: Together, our study demonstrates that HIF1α/ATF3 participated in P4HA1/succinate signaling, which is the major regulator of succinate biosynthesis and PGK1 succinylation at K191 and K192 sites in GBM. The P4HA1/succinate pathway might be a novel and promising target for aerobic glycolysis in GBM.
    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
    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)

公众号