Hif

HIF
  • 文章类型: Journal Article
    在掺杂的情况下,一名顶级运动员挑战违反反兴奋剂规则的行为,牵涉到的是Molidustat.Molidustat是最近开发的缺氧诱导因子(HIF)的稳定剂。目前正在进行与慢性肾脏疾病相关的贫血的临床试验。世界反兴奋剂机构(S2级)始终禁止使用HIF稳定剂。由于它们的药理特性,这些新药可以提高运动成绩。运动员的辩护希望分析多个角化基质,因为它们允许长期调查。有关HIF稳定剂的要求不断增长。因此,我们开发了一种液相色谱与串联质谱法相结合的方法来鉴定和定量这类三种分子:moleidustat,vadadustat,和roxadustat.将30毫克角化基质在1mLpH8.4磷酸氢二铵缓冲液中在40°C下与1ng睾酮-D3孵育16小时,用作内标。用乙酸乙酯/乙醚(80/20)萃取后,蒸发有机相,并且将干燥的残余物在30μL的初始相中重构。对于三种分析物,该方法从5至1000pg/mg是线性的。molidustat的定量限值为2、0.5和5pg/mg,罗沙达,和vadadustat,分别。对运动员头毛的分析(在尿液测试后1个月收集)显示moidustat的浓度为135pg/mg,他的胡须头发和手指甲剪含有55和40皮克/毫克,分别。
    In a doping case, a top athlete challenged an anti-doping rule violation, involving molidustat. Molidustat is a stabilizing agent of the hypoxia-inducible factor (HIF) recently developed. It is currently undergoing clinical trials for anemia associated with chronic kidney disease. HIF stabilizers are banned at all times by the World Anti-Doping Agency (class S2). Because of their pharmacological proprieties, these new drugs can enhance athletic performance. The athlete\'s defense wanted to analyze multiple keratinized matrices as they allow long-term investigations. Requests concerning HIF stabilizers are constantly growing. We have therefore developed a liquid chromatography coupled with tandem mass spectrometry method to identify and quantify three molecules of this class: molidustat, vadadustat, and roxadustat. Thirty milligrams of keratinized matrices were incubated in 1 mL of pH 8.4 diammonium hydrogen phosphate buffer for 16 h at 40°C with 1 ng of testosterone-D3, used as internal standard. After extraction with ethyl acetate/diethyl ether (80/20), the organic phase was evaporated, and the dry residue was reconstituted in 30 μL of initial phase. The method was linear from 5 to 1000 pg/mg for the three analytes. Limits of quantification were 2, 0.5, and 5 pg/mg for molidustat, roxadustat, and vadadustat, respectively. The analysis of the athlete\'s head hair (collected 1 month after the urine test) showed a concentration of molidustat of 135 pg/mg, and his beard hair and his fingernails clippings contained 55 and 40 pg/mg, respectively.
    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
    子宫肌瘤是女性最常见的肿瘤,影响到全世界70%的女性。然而,有针对性的治疗选择是有限的。氧化应激最近已成为肌瘤发病机制的关键驱动因素,并提供了对缺氧诱导的细胞转化的见解。细胞外基质病理生理学,缺氧细胞信号级联,和子宫生物学。缺氧通过(1)促进子宫肌层干细胞增殖驱动肌瘤的发生,(2)引起DNA损伤促进干细胞向肿瘤起始细胞转化,和(3)驱动过量的细胞外基质(ECM)产生。常见的纤维瘤相关DNA突变包括MED12突变,HMGA2过表达,和富马酸水合酶功能丧失。证据表明缺氧信号与这些突变之间存在相互作用。缺氧触发的细胞信号通过多种途径促进纤维的发育和生长,包括HIF-1,TGFβ,和Wnt/β-连环蛋白。由于抗氧化剂失衡,纤维相关的缺氧持续存在,ECM积累,和生长超过足够的血管供应。目前临床上可用的纤维瘤治疗没有利用缺氧靶向治疗。越来越多的临床前和临床研究确定ROS抑制剂,抗HIF-1药物,Wnt/β-连环蛋白抑制,和TGFβ级联抑制剂作为可以通过靶向缺氧来减少肌瘤发育和生长的药物。
    Uterine fibroids are the most common tumors in females affecting up to 70% of women world-wide, yet targeted therapeutic options are limited. Oxidative stress has recently surfaced as a key driver of fibroid pathogenesis and provides insights into hypoxia-induced cell transformation, extracellular matrix pathophysiology, hypoxic cell signaling cascades, and uterine biology. Hypoxia drives fibroid tumorigenesis through (1) promoting myometrial stem cell proliferation, (2) causing DNA damage propelling transformation of stem cells to tumor initiating cells, and (3) driving excess extracellular matrix (ECM) production. Common fibroid-associated DNA mutations include MED12 mutations, HMGA2 overexpression, and Fumarate hydratase loss of function. Evidence suggests an interaction between hypoxia signaling and these mutations. Fibroid development and growth are promoted by hypoxia-triggered cell signaling via various pathways including HIF-1, TGFβ, and Wnt/β-catenin. Fibroid-associated hypoxia persists due to antioxidant imbalance, ECM accumulation, and growth beyond adequate vascular supply. Current clinically available fibroid treatments do not take advantage of hypoxia-targeting therapies. Growing pre-clinical and clinical studies identify ROS inhibitors, anti-HIF-1 agents, Wnt/β-catenin inhibition, and TGFβ cascade inhibitors as agents that may reduce fibroid development and growth through targeting hypoxia.
    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:尽管已经在胶质母细胞瘤中确定了几种生存的预后因素,还有许多其他潜在的标志物(如血红蛋白),其作用尚未得到证实。这项研究的目的是评估广泛的潜在预后因素,包括HIF-1α和血红蛋白水平,在胶质母细胞瘤中存活。次要目的是确定血红蛋白水平是否与HIF-1α表达相关。方法:对我院2012年至2021年收治的136例胶质母细胞瘤患者进行回顾性研究。进行Cox单变量和多变量分析。产生Kaplan-Meier存活曲线。此外,对关键变量进行双变量非参数相关分析.结果:中位生存期为11.9个月(范围:0-119.4)。根据单变量分析,13个变量与生存率显着相关:年龄,性能状态,手术范围,肿瘤深度,肿瘤大小,癫痫,术后放化疗,IDH突变,CD44,HIF-1α,HIF-1β,波形蛋白,和PDFGR。根据多元回归分析,只有四个变量与生存率显着相关:年龄,手术范围,癫痫,和HIF-1α表达。血红蛋白水平之间未观察到显着关联(女性<120g/L或男性<140g/L与高≥120或≥140g/L)和生存率或HIF-1α/HIF-1β表达。结论:在这项对胶质母细胞瘤患者的回顾性研究中,四个变量-年龄,手术范围,HIF-1α表达,和癫痫-是生存的重要预后因素。血红蛋白水平与生存率或HIF-1α表达无显著相关。尽管缺氧是胶质母细胞瘤微环境的公认组成部分,需要更多的研究来了解肿瘤缺氧的发病机制和治疗意义。
    Background: Although several prognostic factors for survival have been identified in glioblastoma, there are numerous other potential markers (such as hemoglobin) whose role has not yet been confirmed. The aim of this study was to evaluate a wide range of potential prognostic factors, including HIF-1α and hemoglobin levels, for survival in glioblastoma. A secondary aim was to determine whether hemoglobin levels were associated with HIF-1α expression. Methods: A retrospective study of 136 patients treated for glioblastoma at our institution between 2012 and 2021 was performed. Cox univariate and multivariate analyses were carried out. Kaplan-Meier survival curves were generated. In addition, bivariate non-parametric correlation analyses were performed for key variables. Results: Median survival was 11.9 months (range: 0-119.4). According to the univariate analysis, 13 variables were significantly associated with survival: age, performance status, extent of surgery, tumor depth, tumor size, epilepsy, postoperative chemoradiotherapy, IDH mutations, CD44, HIF-1α, HIF-1β, vimentin, and PDFGR. According to the multivariate regression analysis, only four variables remained significantly associated with survival: age, extent of surgery, epilepsy, and HIF-1α expression. No significant association was observed between hemoglobin levels (low <120 g/L in females or <140 g/L in males vs. high ≥120 or ≥140 g/L) and survival or HIF-1α/HIF-1β expression. Conclusions: In this retrospective study of patients with glioblastoma, four variables-age, extent of surgery, HIF-1α expression, and epilepsy-were significant prognostic factors for survival. Hemoglobin levels were not significantly associated with survival or HIF-1α expression. Although hypoxia is a well-recognized component of the glioblastoma microenvironment, more research is needed to understand the pathogenesis of onset tumor hypoxia and treatment implication.
    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
    HIF(缺氧诱导因子)调节心脏功能的许多方面。我们和其他人先前表明,在小鼠模型中,心脏中的慢性HIF激活会表现出人类缺血性心肌病的多种特征,包括线粒体丢失,脂质积累,心脏收缩功能障碍.在某些设置中,HIF还导致过氧化物酶体的损失。How,机械上,HIF促进心脏功能障碍是一个悬而未决的问题。
    我们使用缺乏心脏pVHL(vonHippel-Lindau蛋白)的小鼠来研究慢性HIF激活如何导致缺血性心肌病的多种特征,如自噬诱导和脂质积累。我们进行了免疫印迹试验,RNA测序,线粒体和过氧化物酶体自噬通量测量,以及心脏和分离心肌细胞的活细胞成像。我们在小鼠中使用CRISPR-Cas9基因编辑来验证慢性HIF激活背景下心脏功能障碍的新介质。
    我们确定了一种先前未知的途径,通过该途径,心脏HIF激活可促进线粒体和过氧化物酶体的丢失。我们发现DEPP1(孕酮1诱导的蜕膜蛋白)在缺氧下以HIF依赖性方式诱导并定位在线粒体内。DEPP1对于缺氧诱导的自噬和甘油三酸酯在离体心肌细胞中的积累既必要又充分。DEPP1丢失增加了体外慢性HIF激活的心肌细胞存活率,全身Depp1丢失可减少体内VHL丢失引起的慢性HIF激活心脏的心功能不全。
    我们的发现将DEPP1确定为慢性缺血发生的心脏重塑的关键组成部分。
    UNASSIGNED: HIF (hypoxia inducible factor) regulates many aspects of cardiac function. We and others previously showed that chronic HIF activation in the heart in mouse models phenocopies multiple features of ischemic cardiomyopathy in humans, including mitochondrial loss, lipid accumulation, and systolic cardiac dysfunction. In some settings, HIF also causes the loss of peroxisomes. How, mechanistically, HIF promotes cardiac dysfunction is an open question.
    UNASSIGNED: We used mice lacking cardiac pVHL (von Hippel-Lindau protein) to investigate how chronic HIF activation causes multiple features of ischemic cardiomyopathy, such as autophagy induction and lipid accumulation. We performed immunoblot assays, RNA sequencing, mitochondrial and peroxisomal autophagy flux measurements, and live cell imaging on hearts and isolated cardiomyocytes. We used CRISPR-Cas9 gene editing in mice to validate a novel mediator of cardiac dysfunction in the setting of chronic HIF activation.
    UNASSIGNED: We identify a previously unknown pathway by which cardiac HIF activation promotes the loss of mitochondria and peroxisomes. We found that DEPP1 (decidual protein induced by progesterone 1) is induced under hypoxia in a HIF-dependent manner and localizes inside mitochondria. DEPP1 is both necessary and sufficient for hypoxia-induced autophagy and triglyceride accumulation in cardiomyocytes ex vivo. DEPP1 loss increases cardiomyocyte survival in the setting of chronic HIF activation ex vivo, and whole-body Depp1 loss decreases cardiac dysfunction in hearts with chronic HIF activation caused by VHL loss in vivo.
    UNASSIGNED: Our findings identify DEPP1 as a key component in the cardiac remodeling that occurs with chronic ischemia.
    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
    这项研究调查了受刺激的胰腺β细胞和癌细胞之间的代谢相似性,专注于葡萄糖和谷氨酰胺代谢。应对2型糖尿病(T2D)和癌症的重大公共卫生挑战,我们的目标是加深我们对驱动胰岛素分泌和细胞增殖的机制的理解。我们对回补循环和NADPH在生物合成中的作用的分析阐明了它们在这两个过程中的重要功能。此外,我们指出,两种细胞都有一个由Nrf2信号通路介导的抗氧化反应,谷胱甘肽合成,和UCP2上调。值得注意的是,UCP2促进C4代谢物的转移,增强还原性TCA循环代谢。此外,我们观察到,刺激后β细胞的缺氧反应是短暂的,但在癌细胞中持续存在。通过综合这些见解,这项研究可能为T2D提供新的治疗靶点,强调受刺激的β细胞和癌细胞的共同代谢策略。这种比较分析不仅阐明了这些条件的代谢复杂性,而且强调了代谢途径在细胞功能和生存中的关键作用。为应对T2D和癌症挑战提供新的视角。
    This study investigates the metabolic parallels between stimulated pancreatic beta cells and cancer cells, focusing on glucose and glutamine metabolism. Addressing the significant public health challenges of Type 2 Diabetes (T2D) and cancer, we aim to deepen our understanding of the mechanisms driving insulin secretion and cellular proliferation. Our analysis of anaplerotic cycles and the role of NADPH in biosynthesis elucidates their vital functions in both processes. Additionally, we point out that both cell types share an antioxidative response mediated by the Nrf2 signaling pathway, glutathione synthesis, and UCP2 upregulation. Notably, UCP2 facilitates the transfer of C4 metabolites, enhancing reductive TCA cycle metabolism. Furthermore, we observe that hypoxic responses are transient in beta cells post-stimulation but persistent in cancer cells. By synthesizing these insights, the research may suggest novel therapeutic targets for T2D, highlighting the shared metabolic strategies of stimulated beta cells and cancer cells. This comparative analysis not only illuminates the metabolic complexity of these conditions but also emphasizes the crucial role of metabolic pathways in cell function and survival, offering fresh perspectives for tackling T2D and cancer challenges.
    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
    通道cat鱼(Ictaluruspunctatus)的商业培养发生在土塘中,其特征是溶解氧浓度的diel波动,可能会下降到严重的缺氧水平,从而抑制食欲并导致次优生长。鉴于下丘脑在调节其他鱼类这些过程中的重要性,对下丘脑转录组进行了研究,以鉴定响应缺氧的特定基因和表达模式。将常氧水中的Channel鱼与经过12小时缺氧(20%氧饱和度;1.8mgO2/L;27°C),然后在常氧条件下恢复12小时,以模拟cat鱼养殖池塘中的24小时。鱼在0-,6-,12-,18-,和24小时的时间点,在缺氧期间进行6小时和12小时的采样。共有190个基因在实验过程中差异表达,大多数发生在缺氧期间,并在常氧后6小时内恢复到基线值。差异表达的基因通过功能分类为基因本体论生物学过程,并显示大多数被归类为“对缺氧的反应”,“发芽血管生成”,和“细胞对异种生物刺激的反应”。此处报道的基因表达模式表明,随着常氧的开始,对缺氧的转录组反应广泛且迅速可逆。尽管在本实验中没有发现通常报道的调节食欲的基因差异表达,几个候选人被确定为未来的研究调查缺氧和食欲之间的相互作用的通道cat鱼,包括ADM,igfbp1a,igfbp7和stc2b.
    Commercial culture of channel catfish (Ictalurus punctatus) occurs in earthen ponds that are characterized by diel swings in dissolved oxygen concentration that can fall to severe levels of hypoxia which can suppress appetite and lead to suboptimal growth. Given the significance of the hypothalamus in regulating these processes in other fishes, an investigation into the hypothalamus transcriptome was conducted to identify specific genes and expression patterns responding to hypoxia. Channel catfish in normoxic water were compared to catfish subjected to 12 hours of hypoxia (20% oxygen saturation; 1.8 mg O2/L; 27 °C) followed by 12 hours of recovery in normoxia to mimic 24-hours in a catfish aquaculture pond. Fish were sampled at 0-, 6-, 12-, 18-, and 24-hour time points, with the 6- and 12-hour samplings occurring during hypoxia. A total of 190 genes were differentially expressed during the experiment, with most occurring during hypoxia and returning to baseline values within 6 hours of normoxia. Differentially expressed genes were sorted by function into Gene Ontology biological processes and revealed that most were categorized as \"response to hypoxia\", \"sprouting angiogenesis\", and \"cellular response to xenobiotic stimulus\". The patterns of gene expression reported here suggest that transcriptome responses to hypoxia are broad and quickly reversibly with the onset of normoxia. Although no genes commonly reported to modulate appetite were found to be differentially expressed in this experiment, several candidates were identified for future studies investigating the interplay between hypoxia and appetite in channel catfish, including adm, igfbp1a, igfbp7, and stc2b.
    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(HIF)1和2调节相似但不同的靶基因组。尽管HIF以其在介导缺氧反应中的作用而闻名,但积累的证据表明,在某些条件下,HIF,特别是HIF2,也可以在常氧条件下发挥作用。在这里,我们报道了HIF2α在常氧条件下在肾上皮细胞中起作用以调节粘附连接的形成。HIF2α表达是诱导Dock4/Rac1/Pak1信号传导介导的稳定性和E-cadherin在新生粘附连接处的压实所必需的。HIF2α或Dock4缺陷细胞中粘附连接形成受损导致3D肾上皮细胞培养物中囊肿形态发生异常。一起来看,我们表明HIF2α在常氧中起着调节上皮形态发生的作用。
    Hypoxia-inducible factors (HIF) 1 and 2 regulate similar but distinct sets of target genes. Although HIFs are best known for their roles in mediating the hypoxia response accumulating evidence suggests that under certain conditions HIFs, particularly HIF2, may function also under normoxic conditions. Here we report that HIF2α functions under normoxic conditions in kidney epithelial cells to regulate formation of adherens junctions. HIF2α expression was required to induce Dock4/Rac1/Pak1-signaling mediating stability and compaction of E-cadherin at nascent adherens junctions. Impaired adherens junction formation in HIF2α- or Dock4-deficient cells led to aberrant cyst morphogenesis in 3D kidney epithelial cell cultures. Taken together, we show that HIF2α functions in normoxia to regulate epithelial morphogenesis.
    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
    氧化应激在慢性肾脏病(CKD)的发生、发展中起着关键作用,诱导肾细胞损伤,炎症,和纤维化。然而,目前缺乏减缓CKD进展的有效治疗干预措施.分子氢(H2)的多方面药理作用使其成为有希望的治疗途径。H2能够捕获有害的·OH和ONOO-,同时维持参与细胞信号传导的关键活性氧(ROS)。NRF2-KEAP1系统,管理细胞氧化还原平衡,可用于治疗CKD。H2激活了这个途径,强化抗氧化防御和清除ROS以抵消氧化应激。H2可以通过Wnt/β-catenin途径改善NRF2信号传导,并间接激活线粒体中的NRF2-KEAP1。此外,H2通过调节细胞氧化还原状态调节NF-κB活性,抑制MAPK通路,并保持Trx水平。用H2处理还通过中和ROS减弱HIF信号,同时间接增强HIF-1α功能。此外,H2影响FOXO因子并增强抗氧化酶的活性。尽管实验室研究的结果令人鼓舞,临床试验仍然有限,需要进一步研究.这篇综述的重点是氢在治疗肾脏疾病中的作用,特别关注氧化应激和氧化还原信号调节,并讨论了其潜在的临床应用。
    Oxidative stress plays a key role in chronic kidney disease (CKD) development and progression, inducing kidney cell damage, inflammation, and fibrosis. However, effective therapeutic interventions to slow down CKD advancement are currently lacking. The multifaceted pharmacological effects of molecular hydrogen (H2) have made it a promising therapeutic avenue. H2 is capable of capturing harmful •OH and ONOO- while maintaining the crucial reactive oxygen species (ROS) involved in cellular signaling. The NRF2-KEAP1 system, which manages cell redox balance, could be used to treat CKD. H2 activates this pathway, fortifying antioxidant defenses and scavenging ROS to counteract oxidative stress. H2 can improve NRF2 signaling by using the Wnt/β-catenin pathway and indirectly activate NRF2-KEAP1 in mitochondria. Additionally, H2 modulates NF-κB activity by regulating cellular redox status, inhibiting MAPK pathways, and maintaining Trx levels. Treatment with H2 also attenuates HIF signaling by neutralizing ROS while indirectly bolstering HIF-1α function. Furthermore, H2 affects FOXO factors and enhances the activity of antioxidant enzymes. Despite the encouraging results of bench studies, clinical trials are still limited and require further investigation. The focus of this review is on hydrogen\'s role in treating renal diseases, with a specific focus on oxidative stress and redox signaling regulation, and it discusses its potential clinical applications.
    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
    脑转移是肺癌最致命的形式。最近的研究强调了肺癌脑转移(LCBM)和原发性肺癌之间的肿瘤微环境(TME)的显着差异。这对肿瘤的进展和耐药性有很大的贡献。癌相关成纤维细胞(CAF)是具有高可塑性的促肿瘤TME的主要组成部分。然而,LCBM中CAF的谱系组成和功能仍然难以捉摸。通过重新分析肺癌患者的单细胞RNA测序(scRNA-seq)数据(GSE131907),这些患者具有不同的转移阶段,包括原发性病变和脑转移,我们发现,在缺氧情况下,在LCBM期间,CAFs经历了独特的谱系转变,这是由缺氧诱导的HIF-2α激活直接驱动的。转移的CAFs通过VEGF途径增强血管生成,触发代谢重编程,促进肿瘤细胞的生长。使用大量RNA测序数据作为验证队列。对脑转移的四个配对样品及其原发性肺癌对应物进行多重免疫组织化学(mIHC)测定以验证发现。我们的研究揭示了肺癌脑转移的新机制,其特征是HIF-2α诱导的谱系转变和CAFs的功能改变。提供了潜在的治疗靶点。
    Brain metastasis is the most devasting form of lung cancer. Recent studies highlight significant differences in the tumor microenvironment (TME) between lung cancer brain metastasis (LCBM) and primary lung cancer, which contribute significantly to tumor progression and drug resistance. Cancer-associated fibroblasts (CAFs) are the major component of pro-tumor TME with high plasticity. However, the lineage composition and function of CAFs in LCBM remain elusive. By reanalyzing single-cell RNA sequencing (scRNA-seq) data (GSE131907) from lung cancer patients with different stages of metastasis comprising primary lesions and brain metastasis, we found that CAFs undergo distinctive lineage transition during LCBM under a hypoxic situation, which is directly driven by hypoxia-induced HIF-2α activation. Transited CAFs enhance angiogenesis through VEGF pathways, trigger metabolic reprogramming, and promote the growth of tumor cells. Bulk RNA sequencing data was utilized as validation cohorts. Multiplex immunohistochemistry (mIHC) assay was performed on four paired samples of brain metastasis and their primary lung cancer counterparts to validate the findings. Our study revealed a novel mechanism of lung cancer brain metastasis featuring HIF-2α-induced lineage transition and functional alteration of CAFs, which offers potential therapeutic targets.
    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)

公众号