Nuclear Receptor Subfamily 1, Group D, Member 1

核受体亚家族 1, D 组,成员 1
  • 文章类型: Journal Article
    情感障碍通常与昼夜节律紊乱有关。中枢5-羟色胺(5-羟色胺,已经报道了5-HT)模式;但是,5-HTergic情绪调节的昼夜节律控制的功能机制在很大程度上仍然未知。这里,我们研究了昼夜节律核受体REV-ERBα在调节色氨酸羟化酶2(Tph2)中的作用,5-HT合成的限速酶。我们证明了在背侧中缝(DR)5-HTergic神经元中表达的REV-ERBα在功能上与PET-1-一种对5-HTergic神经元发育至关重要的核激活剂竞争。在老鼠身上,DR5-HTergicREV-ERBα的遗传消融增加了Tph2的表达,导致DR5-HT水平升高,黄昏时抑郁样行为减少。Further,小鼠DRREV-ERBα活性的药物操作增加DR5-HT水平并影响绝望相关行为。我们的发现为昼夜节律和情绪控制DR5-HTergic系统之间的分子和细胞联系提供了有价值的见解。
    Affective disorders are frequently associated with disrupted circadian rhythms. The existence of rhythmic secretion of central serotonin (5-hydroxytryptamine, 5-HT) pattern has been reported; however, the functional mechanism underlying the circadian control of 5-HTergic mood regulation remains largely unknown. Here, we investigate the role of the circadian nuclear receptor REV-ERBα in regulating tryptophan hydroxylase 2 (Tph2), the rate-limiting enzyme of 5-HT synthesis. We demonstrate that the REV-ERBα expressed in dorsal raphe (DR) 5-HTergic neurons functionally competes with PET-1-a nuclear activator crucial for 5-HTergic neuron development. In mice, genetic ablation of DR 5-HTergic REV-ERBα increases Tph2 expression, leading to elevated DR 5-HT levels and reduced depression-like behaviors at dusk. Further, pharmacological manipulation of the mice DR REV-ERBα activity increases DR 5-HT levels and affects despair-related behaviors. Our findings provide valuable insights into the molecular and cellular link between the circadian rhythm and the mood-controlling DR 5-HTergic systems.
    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
    昼夜节律失调涉及脊髓损伤(SCI)的病理生理学。昼夜节律的调节为SCI治疗带来了希望。这里,我们旨在研究嗅鞘细胞(OEC)通过调节小胶质细胞时钟基因表达减轻神经炎症的机制。在这项研究中,SCI年夜鼠随机分为OEC组和媒介物组。手术后1天,将OECs静脉移植到OEC组SCI大鼠中,而媒介物组的大鼠接受培养基。OEC移植后7天,从大脑收集组织(前额叶皮质,下丘脑,脊髓)用于PCR,在zeitgeber时间(ZT)6,ZT12,ZT18和ZT24进行蛋白质印迹和免疫组织化学(IHC)测定。通过实验抑制基因表达和共培养实验研究了OEC在调节小胶质细胞REV-ERBα中的作用。在车辆组中,IHC显示与对照组相比,Iba-1在脑白质和脊髓中的表达显着增加(所有比较均P<0.0001)。Iba-1的表达显著降低(所有比较P<0.0001)。在OEC组中,PER1,PER2,CLOCK,REV-ERBα在脊髓和脑区均有节奏。SCI打乱了他们典型的节奏。OECs移植可以通过上调REV-ERBα来调节这些失调。体外研究表明,OECs不能降低REV-ERBα抑制小胶质细胞的活化。OECs的静脉移植可以通过上调SCI后的REV-ERBα来介导脑和脊髓小胶质细胞的活化。
    Circadian dysregulation involved in the pathophysiology of spinal cord injury (SCI). Modulation of circadian rhythms hold promise for the SCI treatment. Here, we aim to investigated the mechanism of olfactory ensheathing cells (OEC) in alleviating neuroinflammation via modulating clock gene expression in microglia. In this study, SCI rats were randomly divided into OEC group and vehicle group. At 1 day after the surgery, OECs were intravenously transplanted into OEC group SCI rat, while the rats in vehicle group received culture medium. After 7 days post of OEC transplantation, tissues were collected from the brain (prefrontal cortex, hypothalamus, spinal cord) for PCR, western blotting and immunohistochemistry (IHC) assay at zeitgeber time (ZT) 6, ZT 12, ZT 18, and ZT 24. The roles of OEC in modulating REV-ERBα in microglia were studied by experimental inhibition of gene expression and the co-culture experiment. In the vehicle group, IHC showed a significant increase of Iba-1 expression in the cerebral white matter and spinal cord compared with control group (P < 0.0001 for all comparisons). The expression of Iba-1 was significantly decreased (P < 0.0001 for all comparisons). In the OEC group, the expression of PER 1, PER 2, CLOCK, and REV-ERBα was in a rhythmical manner in both spinal cord and brain regions. SCI disrupted their typical rhythms. And OECs transplantation could modulate those dysregulations by upregulating REV-ERBα. In vitro study showed that OECs couldn\'t reduce the activation of REV-ERBα inhibited microglia. The intravenous transplantation of OECs can mediate cerebral and spinal microglia activation through upregulation REV-ERBα after SCI.
    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:衰老会增加前列腺癌的患病率。生物钟协调新陈代谢,细胞周期,肿瘤抑制基因p53.尽管体育锻炼对预防前列腺疾病有多种作用,它对前列腺昼夜节律的基因和蛋白质的调节作用需要更好地评估。本研究验证了REV-ERBα(Nr1d1)的表达,Bmal1,凋亡,肿瘤抑制剂,能量代谢标记,接受联合体育锻炼的18个月大小鼠前列腺微环境中的雄激素受体。
    方法:C57BL/6J小鼠分为2组:6月龄(n=10)和18月龄,(n=20)。将18个月大的动物分为2个亚组:久坐(n=10,18mSed)和接受联合体育锻炼(n=10,18mTR)。通过在跑步机上跑步(增量负荷测试的40-60%)和攀岩力量训练(最大重复测试的40-50%)进行组合体能训练方案,由5×/周(3天有氧和2天强度)组成,持续3周。准备前列腺进行Westernblot和RT-qPCR分析,并制备了用于生化分析的浆。
    结果:衰老过程中的联合体育锻炼导致前列腺中Bmal1水平升高和REV-ERBα水平降低。这些结果伴随着AMPK/SIRT1/PGC-1α蛋白的减少和PI3K/AKT和p53/PTEN/caspase3途径的增加,促进凋亡潜能。
    结论:这些研究结果表明,通过使前列腺组织中的Bmal1和REV-ERBα重新同步,力量和有氧体育锻炼可能在肿瘤前期分子改变和年龄相关特征的发展中具有预防性作用。
    BACKGROUND: Aging increases the prevalence of prostate cancer. The circadian clock coordinates metabolism, cell cycle, and tumor suppressor p53. Although physical exercise has several effects on preventing prostate diseases, its effect on regulating genes and proteins of the circadian rhythm of the prostate needs to be better evaluated. The present study verified expression of REV-ERBα (Nr1d1), Bmal1, apoptosis, tumor suppressors, energetic metabolism markers, and androgen receptors in the prostatic microenvironment in 18-month-old mice submitted to combined physical training.
    METHODS: C57BL/6 J mice were divided into 2 groups: 6 months-old (n = 10) and 18 months-old, (n = 20). The 18-month-old animals were divided into 2 subgroups: sedentary (n = 10, 18 m Sed) and submitted to combined physical training (n = 10, 18 m TR). Combined physical training protocol was performed by running on the treadmill (40-60 % of incremental load test) and climbing strength training (40-50 % of maximum repetition test), consisting of 5×/week (3 days aerobic and 2 days strength) for 3 weeks. The prostate was prepared for Western blot and RT-qPCR analysis, and the plasm was prepared for the biochemistry analysis.
    RESULTS: Combined physical exercise during aging led to increased levels of Bmal1 and decreased levels of REV-ERBα in the prostate. These results were accompanied by a reduction in the AMPK/SIRT1/PGC-1α proteins and an increase in the PI3K/AKT and p53/PTEN/caspase 3 pathways, promoting apoptotic potential.
    CONCLUSIONS: These findings suggest that strength and aerobic physical exercise may be preventive in the development of preneoplastic molecular alterations and age-related features by re-synchronizes Bmal1 and REV-ERBα in prostatic tissues.
    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
    背景:阻塞性睡眠呼吸暂停(OSA)是2型糖尿病(DM2)的危险因素之一。由于OSA与昼夜节律的破坏有关,它会影响生物钟蛋白质,包括神经元PAS结构域蛋白2(NPAS2)和核受体亚家族1组D成员1(Rev-Erb-α)。这些蛋白质已被证明与代谢异常有关,i.a.,胰岛素抵抗。
    目的:本试验研究旨在研究重度OSA和重度OSA+DM2患者与健康对照组比较的NPAS2和Rev-Erb-α蛋白血清水平。考虑到与多导睡眠图(PSG)参数的相关性(例如,氧饱和度(SpO2)变量)。
    方法:共有40名参与者被纳入研究。分为3组:OSA组(n=17;呼吸暂停低通气指数(AHI)>30,无DM2);OSADM2组(n=7;AHI>30和DM2);和对照组(n=16;AHI<5,无DM2)。所有参与者都接受了夜间PSG检查,并在第二天早上收集了血液。使用酶联免疫吸附测定(ELISA)评估NPAS2和Rev-Erb-α蛋白的血清水平。
    结果:与健康个体相比,OSA组的平均NPAS2蛋白水平显着降低(p=0.017)。此外,与OSA+DM2组相比,OSA组的NPAS2蛋白水平较低,但只观察到一种趋势(p=0.094)。未观察到Rev-Erb-α蛋白浓度的差异。此外,观察到快速眼动(REM)睡眠期间的AHI与NPAS2蛋白血清水平之间呈负相关(r=-0.478;p=0.002)。
    结论:血清NPAS2蛋白可能参与OSA患者的代谢失调,而机制本身可能与REM睡眠有关。
    BACKGROUND: Obstructive sleep apnea (OSA) is one of the risk factors for diabetes mellitus type 2 (DM2). As OSA is associated with the disruption of the circadian rhythm, it affects circadian clock proteins, including neuronal PAS domain protein 2 (NPAS2) and nuclear receptor subfamily 1 group D member 1 (Rev-Erb-α). These proteins have been shown to be related to metabolic abnormalities, i.a., insulin resistance.
    OBJECTIVE: The present pilot study aimed to investigate the NPAS2 and Rev-Erb-α protein serum levels in the groups of patients with severe OSA and severe OSA+DM2 in comparison with healthy controls, taking into account correlations with polysomnography (PSG) parameters (e.g., oxygen saturation (SpO2) variables).
    METHODS: A total of 40 participants were included in the study. They were split into 3 groups as follows: the OSA group (n = 17; apnea-hypopnea index (AHI) >30, no DM2); the OSA+DM2 group (n = 7; AHI > 30 and DM2); and the control group (n = 16; AHI < 5, no DM2). All participants underwent a nocturnal PSG examination and had their blood collected the following morning. The serum levels of NPAS2 and Rev-Erb-α proteins were assessed using the enzyme-linked immunosorbent assay (ELISA).
    RESULTS: The mean NPAS2 protein level was significantly lower in the OSA group as compared to healthy individuals (p = 0.017). Additionally, the OSA group presented with lower NPAS2 protein levels as compared to the OSA+DM2 group, but only a tendency was observed (p = 0.094). No differences in the Rev-Erb-α protein concentration were noticed. Furthermore, a negative correlation between AHI during rapid eye movement (REM) sleep and the NPAS2 protein serum level was observed (r = -0.478; p = 0.002).
    CONCLUSIONS: Serum NPAS2 protein might be involved in metabolic dysregulation present among OSA patients, while the mechanism itself may be associated with REM sleep.
    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
    目的:探讨妊娠期过度光照对大鼠宫内发育及新生儿早期生长的影响。
    方法:妊娠大鼠随机分为三组:持续光照组,非光照组和对照组。从尾静脉收集血样以分析褪黑激素和皮质醇水平。重量,记录每日食物和水的消耗量.子宫重量,在妊娠第19天测量胎盘重量和胎盘直径。还监测了自然出生和新生儿生长。NR1D1(核受体亚家族1组D成员1)在子代SCN(视交叉上核)中的表达,测量肝脏和脂肪组织。还测量了NR1D1,MT1(褪黑激素1A受体)和11β-HSD2(胎盘11β-羟基类固醇脱氢酶2型)在胎盘中的表达。最后,在NR1D1siRNA转染的JEG-3细胞中评估MT1和11β-HSD2的表达。
    结果:孕妇体重增加无显著差异,妊娠持续时间,子宫重量,胎盘体重,胎盘直径,三组胎儿数。出生时后代的体重或长度没有显着差异。与其他两组相比,持续光照组的后代在出生后第21天的生长速度明显更快。NR1D1在SCN中的表达,后代的肝脏和脂肪组织在三组之间没有显着差异。常光照射组的母体血清褪黑素和皮质醇水平低于和高于其他两组,分别。持续光照组胎盘组织中NR1D1、MT1和11β-HSD2的表达均降低。NR1D1siRNA转染后,JEG-3细胞中MT1和11β-HSD2的表达降低。
    结论:怀孕期间过度的光照会导致皮质醇升高和褪黑激素对子宫胎儿的暴露减少,可能有助于加速大鼠后代的早期生长。
    OBJECTIVE: To investigate the effects of excessive light exposure during gestation on intrauterine development and early growth of neonates in rats.
    METHODS: Pregnant rats were randomly allocated to three groups: the constant light exposure group, non-light exposure group and control group. Blood samples were collected from the tail vein to analyze melatonin and cortisol levels. Weight, daily food and water consumption were recorded. Uterine weight, placental weight and placental diameter were measured on gestational day 19. Natural birth and neonate growth were also monitored. The expression of NR1D1(nuclear receptor subfamily 1 group D member 1) in offspring\'s SCN (suprachiasmatic nuclei), liver and adipose tissue was measured. Expression of NR1D1, MT1(melatonin 1 A receptor) and 11β-HSD2 (placental 11β-hydroxysteroid dehydrogenase type 2) in placenta was also measured. Finally, the expression of MT1 and 11β-HSD2 in NR1D1 siRNA transfected JEG-3 cells was evaluated.
    RESULTS: There were no significant differences in maternal weight gain, pregnancy duration, uterine weight, placental body weight, placental diameter, fetal number among three groups. There were no significant differences in weights or lengths of offspring at birth. Compared to other two groups, constant light exposure group showed significantly more rapid growth of offspring in 21st day post-birth. The expression of NR1D1 in SCN, liver and adipose tissues of offspring was not significantly different among three groups. The maternal serum melatonin and cortisol levels of the constant light exposure group were lower and higher than other two groups, respectively. The expressions of NR1D1, MT1 and 11β-HSD2 were all decreased in placenta of the constant light exposure group. The expression of MT1 and 11β-HSD2 in JEG-3 cells were decreased after NR1D1 siRNA transfection.
    CONCLUSIONS: Excessive light exposure during pregnancy results in elevated cortisol and reduced melatonin exposure to fetuses in uterus, potentially contributing to an accelerated early growth of offspring in rats.
    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
    促红细胞生成素(EPO)对RBC稳态的调节对于O2运输和维持脊椎动物中足够数量的RBC至关重要。因此,在EPO产生过多和贫血的情况下,EPO合成失调会导致红细胞增多症等疾病,这发生在EPO生产不足时。EPO在治疗贫血患者中起着至关重要的作用;然而,它的过量生产会增加血液粘度,可能导致致命的心力衰竭.因此,能够调节EPO的可药用转录因子(TFs)及其相关配体的鉴定为解决EPO相关疾病提供了有希望的治疗方法.这项研究揭示了一种涉及两个关键核受体(NRs)的新调控机制。Rev-erbα和RORα,在EPO基因表达的控制中。Rev-erbα充当细胞固有的负调节剂,在将红细胞生成维持在正确水平中起着至关重要的作用。它通过直接结合到人和小鼠EPO基因启动子内新鉴定的反应元件来实现这一目标,从而抑制了EPO的生产。这些发现进一步得到Rev-erbα激动剂(SR9011)有效抑制小鼠中缺氧诱导的EPO表达的发现的支持。相比之下,RORα作为EPO基因表达的正调节因子,也与启动子中相同的反应元件结合以诱导EPO产生。最后,这项研究的结果表明,这两个NR,Rev-erbα,和RORα,以消极和积极的方式影响EPO合成,这表明这两种NRs的调节活性可以提供一种靶向与EPO失调相关的疾病的方法。
    UNASSIGNED: The regulation of red blood cell (RBC) homeostasis by erythropoietin (EPO) is critical for O2 transport and maintaining the adequate number of RBCs in vertebrates. Therefore, dysregulation in EPO synthesis results in disease conditions such as polycythemia in the case of excessive EPO production and anemia, which occurs when EPO is inadequately produced. EPO plays a crucial role in treating anemic patients; however, its overproduction can increase blood viscosity, potentially leading to fatal heart failure. Consequently, the identification of druggable transcription factors and their associated ligands capable of regulating EPO offers a promising therapeutic approach to address EPO-related disorders. This study unveils a novel regulatory mechanism involving 2 pivotal nuclear receptors (NRs), Rev-ERBA (Rev-erbα, is a truncation of reverse c-erbAa) and RAR-related orphan receptor A (RORα), in the control of EPO gene expression. Rev-erbα acts as a cell-intrinsic negative regulator, playing a vital role in maintaining erythropoiesis at the correct level. It accomplishes this by directly binding to newly identified response elements within the human and mouse EPO gene promoter, thereby repressing EPO production. These findings are further supported by the discovery that a Rev-erbα agonist (SR9011) effectively suppresses hypoxia-induced EPO expression in mice. In contrast, RORα functions as a positive regulator of EPO gene expression, also binding to the same response elements in the promoter to induce EPO production. Finally, the results of this study revealed that the 2 NRs, Rev-erbα and RORα, influence EPO synthesis in a negative and positive manner, respectively, suggesting that the modulating activity of these 2 NRs could provide a method to target disorders linked with EPO dysregulation.
    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
    众所周知,长期太空飞行会导致昼夜节律中断,由位于下丘脑视交叉上核(SCN)的中央起搏器驱动,但潜在的分子机制仍不清楚。这里,我们开发了一种大鼠模型,通过尾部悬吊和隔离(TSI)模拟微重力和隔离环境。我们发现TSI环境会对核心体温造成昼夜节律破坏,心率,和大鼠的运动活动节律,尤其是这些节奏的幅度。在TSI模型大鼠中,核心昼夜节律基因NR1D1显示出更高的蛋白质而不是mRNA水平,随着BMAL1水平的降低,这表明NR1D1可以通过翻译后调节来调节。自噬体标记物LC3可以通过LC3相互作用区(LIR)基序直接与NR1D1结合,并以线粒体自噬依赖性方式诱导NR1D1降解。线粒体自噬的缺陷导致NR1D1降解的逆转,从而抑制BMAL1的表达。在TSI模型的SCN中观察到线粒体自噬缺陷和随后的线粒体功能障碍。尿磷脂A(UA),线粒体自噬激活剂,表现出增强核心体温振幅的能力,心率,通过促进线粒体自噬诱导降解NR1D1来实现运动活动节律。累计,我们的结果表明,线粒体自噬通过调节NR1D1降解来发挥昼夜节律控制,揭示线粒体自噬是长期太空飞行以及SCN昼夜节律紊乱疾病的潜在靶标。
    Long-term spaceflight is known to induce disruptions in circadian rhythms, which are driven by a central pacemaker located in the suprachiasmatic nucleus (SCN) of the hypothalamus, but the underlying molecular mechanisms remain unclear. Here, we developed a rat model that simulated microgravity and isolation environments through tail suspension and isolation (TSI). We found that the TSI environment imposed circadian disruptions to the core body temperature, heart rate, and locomotor-activity rhythms of rats, especially in the amplitude of these rhythms. In TSI model rats\' SCNs, the core circadian gene NR1D1 showed higher protein but not mRNA levels along with decreased BMAL1 levels, which indicated that NR1D1 could be regulated through post-translational regulation. The autophagosome marker LC3 could directly bind to NR1D1 via the LC3-interacting region (LIR) motifs and induce the degradation of NR1D1 in a mitophagy-dependent manner. Defects in mitophagy led to the reversal of NR1D1 degradation, thereby suppressing the expression of BMAL1. Mitophagy deficiency and subsequent mitochondrial dysfunction were observed in the SCN of TSI models. Urolithin A (UA), a mitophagy activator, demonstrated an ability to enhance the amplitude of core body temperature, heart rate, and locomotor-activity rhythms by prompting mitophagy induction to degrade NR1D1. Cumulatively, our results demonstrate that mitophagy exerts circadian control by regulating NR1D1 degradation, revealing mitophagy as a potential target for long-term spaceflight as well as diseases with SCN circadian disruption.
    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
    REV-ERBα,一种有治疗前景的核激素受体,在调节各种生理过程中起着至关重要的作用,如生物钟,炎症,和新陈代谢。然而,研究这种受体药理学的化学探针的可用性是有限的,SR8278是唯一确定的合成拮抗剂。此外,目前没有X-射线晶体结构证明REV-ERBα与拮抗剂配体的结合。这种结构信息的缺乏阻碍了靶向疗法的发展。为了解决这个问题,我们采用高斯加速分子动力学(GaMD)模拟来研究SR8278与REV-ERBα的结合途径。为了比较,我们还使用GaMD观察了STL1267的配体结合过程,对此有X射线结构。GaMD模拟成功捕获了两种配体与受体正构位点的结合,并预测了配体结合途径和参与拮抗剂SR8278结合的重要氨基酸残基。这项研究强调了GaMD在研究蛋白质-配体相互作用中的有效性,特别是在核激素受体的药物识别方面。
    REV-ERBα, a therapeutically promising nuclear hormone receptor, plays a crucial role in regulating various physiological processes such as the circadian clock, inflammation, and metabolism. However, the availability of chemical probes to investigate the pharmacology of this receptor is limited, with SR8278 being the only identified synthetic antagonist. Moreover, no X-ray crystal structures are currently available that demonstrate the binding of REV-ERBα to antagonist ligands. This lack of structural information impedes the development of targeted therapeutics. To address this issue, we employed Gaussian accelerated molecular dynamics (GaMD) simulations to investigate the binding pathway of SR8278 to REV-ERBα. For comparison, we also used GaMD to observe the ligand binding process of STL1267, for which an X-ray structure is available. GaMD simulations successfully captured the binding of both ligands to the receptor\'s orthosteric site and predicted the ligand binding pathway and important amino acid residues involved in the antagonist SR8278 binding. This study highlights the effectiveness of GaMD in investigating protein-ligand interactions, particularly in the context of drug recognition for nuclear hormone receptors.
    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
    糖尿病肾病(DKD)是糖尿病最常见的并发症之一。临床上没有特定的药物。我们先前已经证明,抑制微粒体前列腺素E合酶2(mPGES-2)通过增强β细胞功能和促进胰岛素产生来减轻2型糖尿病。然而,mPGES-2是否参与DKD尚不清楚.这里,我们旨在分析mPGES-2表达增强与肾脂代谢稳态受损以及随后的肾损害之间的关联.值得注意的是,mPGES-2的整体敲除或药物阻断减轻了糖尿病足细胞损伤和肾小管间质纤维化,从而改善脂质积累和脂毒性。这些发现在足细胞或小管特异性mPGES-2缺陷小鼠中得到进一步证实。机械上,mPGES-2和Rev-Erbα竞争血红素结合以调节糖尿病肾脏中脂肪酸结合蛋白5的表达和脂质代谢。我们的发现提示了通过mPGES-2抑制治疗DKD的潜在策略。
    Diabetic kidney disease (DKD) is one of the most common complications of diabetes, and no specific drugs are clinically available. We have previously demonstrated that inhibiting microsomal prostaglandin E synthase-2 (mPGES-2) alleviated type 2 diabetes by enhancing β cell function and promoting insulin production. However, the involvement of mPGES-2 in DKD remains unclear. Here, we aimed to analyze the association of enhanced mPGES-2 expression with impaired metabolic homeostasis of renal lipids and subsequent renal damage. Notably, global knockout or pharmacological blockage of mPGES-2 attenuated diabetic podocyte injury and tubulointerstitial fibrosis, thereby ameliorating lipid accumulation and lipotoxicity. These findings were further confirmed in podocyte- or tubule-specific mPGES-2-deficient mice. Mechanistically, mPGES-2 and Rev-Erbα competed for heme binding to regulate fatty acid binding protein 5 expression and lipid metabolism in the diabetic kidney. Our findings suggest a potential strategy for treating DKD via mPGES-2 inhibition.
    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
    背景:ApoA5主要由肝脏合成和分泌,是脂蛋白脂肪酶(LPL)活性和富含甘油三酯的脂蛋白(TRLs)的关键调节剂。尽管ApoA5在循环中的肝外甘油三酯(TG)代谢中的作用已得到充分证明,ApoA5与非酒精性脂肪性肝病(NAFLD)之间的关系尚不完全清楚,潜在的分子机制仍有待阐明.方法:我们使用CRISPR/Cas9基因编辑从叙利亚金仓鼠中删除Apoa5基因,复制人类代谢特征的小型啮齿动物模型。然后,ApoA5缺陷型(ApoA5-/-)仓鼠用于研究有或没有挑战高脂肪饮食(HFD)的NAFLD。结果:ApoA5-/-仓鼠表现出高甘油三酯血症(HTG),在2300mg/dL时TG水平显着升高,在常规饮食下肝脏脂肪变性,伴随着脂肪组织中调节脂肪分解和小脂肪细胞的基因表达水平的增加。HFD攻击使ApoA5-/-仓鼠易患重度HTG(sHTG)和非酒精性脂肪性肝炎(NASH)。体外和体内机制研究表明,靶向ApoA5会破坏HepG2细胞和肝脏中NR1D1mRNA的稳定性,从而分别降低NR1D1的mRNA和蛋白质水平。腺相关病毒8(AAV8)在ApoA5-/-仓鼠肝脏中过表达人NR1D1可显着改善脂肪肝,而不影响血浆脂质水平。此外,通过冷暴露或CL316243给药恢复肝脏ApoA5或激活棕色脂肪组织(BAT)中的UCP1可以显着纠正ApoA5-/-仓鼠的sHTG和肝脏脂肪变性。结论:我们的数据表明,仓鼠ApoA5缺乏引起的HTG足以引起肝脏脂肪变性,HFD通过降低肝脏NR1D1mRNA和蛋白水平加重NAFLD,这提供了ApoA5和NAFLD之间的机械联系,并提出了在未来的临床试验中治疗HTG和由于ApoA5缺乏引起的相关疾病的潜在治疗方法的新见解。
    Background: ApoA5 mainly synthesized and secreted by liver is a key modulator of lipoprotein lipase (LPL) activity and triglyceride-rich lipoproteins (TRLs). Although the role of ApoA5 in extrahepatic triglyceride (TG) metabolism in circulation has been well documented, the relationship between ApoA5 and nonalcoholic fatty liver disease (NAFLD) remains incompletely understood and the underlying molecular mechanism still needs to be elucidated. Methods: We used CRISPR/Cas9 gene editing to delete Apoa5 gene from Syrian golden hamster, a small rodent model replicating human metabolic features. Then, the ApoA5-deficient (ApoA5-/-) hamsters were used to investigate NAFLD with or without challenging a high fat diet (HFD). Results: ApoA5-/- hamsters exhibited hypertriglyceridemia (HTG) with markedly elevated TG levels at 2300 mg/dL and hepatic steatosis on a regular chow diet, accompanied with an increase in the expression levels of genes regulating lipolysis and small adipocytes in the adipose tissue. An HFD challenge predisposed ApoA5-/- hamsters to severe HTG (sHTG) and nonalcoholic steatohepatitis (NASH). Mechanistic studies in vitro and in vivo revealed that targeting ApoA5 disrupted NR1D1 mRNA stability in the HepG2 cells and the liver to reduce both mRNA and protein levels of NR1D1, respectively. Overexpression of human NR1D1 by adeno-associated virus 8 (AAV8) in the livers of ApoA5-/- hamsters significantly ameliorated fatty liver without affecting plasma lipid levels. Moreover, restoration of hepatic ApoA5 or activation of UCP1 in brown adipose tissue (BAT) by cold exposure or CL316243 administration could significantly correct sHTG and hepatic steatosis in ApoA5-/- hamsters. Conclusions: Our data demonstrate that HTG caused by ApoA5 deficiency in hamsters is sufficient to elicit hepatic steatosis and HFD aggravates NAFLD by reducing hepatic NR1D1 mRNA and protein levels, which provides a mechanistic link between ApoA5 and NAFLD and suggests the new insights into the potential therapeutic approaches for the treatment of HTG and the related disorders due to ApoA5 deficiency in the clinical trials in future.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号