SREBP

SREBP
  • 文章类型: Journal Article
    内质网(ER)应激被认为是创伤性脑损伤(TBI)进展的关键因素,并且是治疗干预的潜在目标。这项研究旨在评估J147的潜力,一种新型的神经营养化合物,通过调节相关的信号通路缓解内质网应激,从而促进TBI中的功能恢复。为此,成年小鼠接受控制性皮质撞击(CCI)损伤以诱导TBI,受伤后一小时口服J147,每日给药3至7天。在35天内进行了多项行为评估,揭示了一个重要的,J147治疗对神经功能恢复的剂量依赖性改善。神经病理学分析表明急性神经变性减少(通过FJC染色在三天观察到),增强长期神经元存活(H&E和Nissl染色),并且在TBI后35天改善了神经可塑性(高尔基染色)。在分子水平上,TBI诱导的AMP激活蛋白激酶(AMPK)去磷酸化,甾醇调节元件结合蛋白-1(SREBP-1)激活,和ER应激标记蛋白的上调,包括磷酸化的真核起始因子-2α(p-eIF2a),激活转录因子4(ATF4),损伤后三天,病灶周围皮质神经元中的C/EBP同源蛋白(CHOP)。值得注意的是,J147处理显著减弱AMPK去磷酸化,SERBP-1激活,和ER应激标志物的表达。总之,这项研究揭示了J147在减轻与TBI相关的继发性脑损伤和通过调节ER应激途径改善长期功能恢复方面的治疗前景.
    Endoplasmic reticulum (ER) stress is recognized as a crucial contributor to the progression of traumatic brain injury (TBI) and represents a potential target for therapeutic intervention. This study aimed to assess the potential of J147, a novel neurotrophic compound, in alleviating ER stress by modulating related signaling pathways, thereby promoting functional recovery in TBI. To this end, adult mice underwent controlled cortical impact (CCI) injury to induce TBI, followed by oral administration of J147 one-hour post-injury, with daily dosing for 3 to 7 days. Multiple behavioral assessments were conducted over 35 days, revealing a significant, dose-dependent improvement in neurofunctional recovery with J147 treatment. The neuropathological analysis demonstrated reduced acute neurodegeneration (observed at three days through FJC staining), enhanced long-term neuron survival (H&E and Nissl staining), and improved neuroplasticity (Golgi staining) at 35 days post-TBI. At the molecular level, TBI-induced AMP-activated protein kinase (AMPK) dephosphorylation, sterol regulatory element binding protein-1 (SREBP-1) activation, and upregulation of ER stress marker proteins, including phosphorylated eukaryotic initiation factor-2α (p-eIF2a), activating transcription factor 4 (ATF4), and C/EBP homologous protein (CHOP) in perilesional cortex neurons at three days post-injury. Notably, the J147 treatment significantly attenuated AMPK dephosphorylation, SERBP-1 activation, and expression of the ER stress markers. In summary, this study reveals the therapeutic promise of J147 in mitigating secondary brain damage associated with TBI and improving long-term functional recovery by modulating ER stress pathways.
    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
    CTCF是三维染色质折叠和转录控制的关键因素,已发现其通过尚不了解的机制影响癌细胞迁移。为了识别这种机制,我们使用CTCF部分功能丧失(pLoF)的小鼠黑色素瘤细胞。我们发现CTCFpLoF抑制细胞迁移速率,同时导致胆固醇生物合成途径中多种酶的表达增加以及细胞胆固醇水平的升高。与胆固醇变化一致,我们检测到CTCFpLoF细胞中膜动力学的改变,如通过减少迁移体的形成所测量的,在迁移细胞的后侧形成细胞外囊泡。抑制CTCFpLoF细胞中胆固醇合成可恢复细胞迁移速率和迁移体形成,表明CTCF通过抑制胆固醇合成来支持细胞迁移。详细分析了胆固醇合成途径中的早期酶Hmgcs1的启动子,揭示了CTCF防止在该启动子和另一个200kb的启动子之间形成环。CTCF还支持PRC2募集至启动子和H3K27me3的沉积。Hmgcs1启动子处的H3K27me3阻止SREBP2结合和转录激活。通过这种机制,CTCF微调胆固醇水平以支持细胞迁移。值得注意的是,全基因组关联研究表明,CTCF与胆固醇相关疾病之间存在联系,因此,CTCF成为胆固醇生物合成的新调节剂。
    CTCF is a key factor in three-dimensional chromatin folding and transcriptional control that was found to affect cancer cell migration by a mechanism that is still poorly understood. To identify this mechanism, we used mouse melanoma cells with a partial loss of function (pLoF) of CTCF. We found that CTCF pLoF inhibits cell migration rate while leading to an increase in the expression of multiple enzymes in the cholesterol biosynthesis pathway along with an elevation in the cellular cholesterol level. In agreement with the cholesterol change we detected altered membrane dynamics in CTCF pLoF cells as measured by reduced formation of migrasomes, extracellular vesicles formed at the rear side of migrating cells. Inhibition of cholesterol synthesis in CTCF pLoF cells restored the cellular migration rate and migrasome formation, suggesting that CTCF supports cell migration by suppressing cholesterol synthesis. Detailed analysis of the promoter of Hmgcs1, an early enzyme in the cholesterol synthesis pathway, revealed that CTCF prevents formation of a loop between that promoter and another promoter 200 kb away. CTCF also supports PRC2 recruitment to the promoter and deposition of H3K27me3. H3K27me3 at the promoter of Hmgcs1 prevents SREBP2 binding and activation of transcription. By this mechanism, CTCF fine-tunes cholesterol levels to support cell migration. Notably, genome wide association studies suggest a link between CTCF and cholesterol-associated diseases, thus CTCF emerges as a new regulator of cholesterol biosynthesis.
    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
    cGAS-STING先天免疫途径和SREBP激活的胆固醇和脂肪酸合成途径在神经退行性疾病中异常地共同调节。STING信号的激活发生在内质网(ER)膜上,当甾醇丰富时,STING被INSIG1锚定,与SREBP和固醇结合的SREBP裂解激活蛋白(SCAP)一起。当甾醇含量低时,INSIG依赖性STING途径失活,SREBP-SCAP复合物易位到高尔基体,在那里SREBP被切割并易位到细胞核,以反式激活胆固醇和脂肪酸合成的基因。因此,STING与STING的反向激活呼吸:当先天免疫活跃时,胆固醇和脂肪酸合成的途径被抑制,反之亦然。STING途径由外源病毒细胞质核酸与环GMP-AMP合酶(cGAS)DNA传感器或RIG-I和MDA5dsRNA传感器相互作用刺激,但是在神经变性时,先天免疫也被自身DNA和随着神经元死亡而积累的双链RNA激活。下游,激活的STING招募TBK1并刺激干扰素刺激的基因和自噬途径的反式激活,两者都是保护性的。然而,先天免疫的慢性激活有助于小胶质细胞的激活,神经炎症和自噬失败导致神经变性。STING也是质子通道,其在被激活时刺激质子从STING囊泡离开,导致细胞死亡。在这里,我们回顾了先天免疫和胆固醇和脂肪酸合成途径的显着特征,观察到神经退行性疾病中的STING和SREBP信号异常,和相关的治疗方法。
    The cGAS-STING innate immunity pathway and the SREBP-activated cholesterol and fatty acid synthesis pathway are abnormally co-regulated in neurodegenerative disease. Activation of STING signaling occurs at the endoplasmic reticulum (ER) membrane with STING anchored by INSIG1 along with SREBP and the sterol-bound SREBP cleavage activating protein (SCAP) when sterols are in abundance. When sterols are low, the INSIG-dependent STING pathway is inactivated and the SREBP-SCAP complex is translocated to the Golgi where SREBP is cleaved and translocated to the nucleus to transactivate genes for cholesterol and fatty acid synthesis. Thus, there is inverse activation of STING vs. SREBP: when innate immunity is active, pathways for cholesterol and fatty acid synthesis are suppressed, and vice versa. The STING pathway is stimulated by foreign viral cytoplasmic nucleic acids interacting with the cyclic GMP-AMP synthase (cGAS) DNA sensor or RIG-I and MDA5 dsRNA sensors, but with neurodegeneration innate immunity is also activated by self-DNAs and double-stranded RNAs that accumulate with neuronal death. Downstream, activated STING recruits TBK1 and stimulates the transactivation of interferon stimulated genes and the autophagy pathway, which are both protective. However, chronic activation of innate immunity contributes to microglia activation, neuroinflammation and autophagy failure leading to neurodegeneration. STING is also a proton channel that when activated stimulates proton exit from STING vesicles leading to cell death. Here we review the salient features of the innate immunity and cholesterol and fatty acid synthesis pathways, observations of abnormal STING and SREBP signaling in neurodegenerative disease, and relevant therapeutic approaches.
    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
    甾醇调节元件结合蛋白(SREBP)是控制脂质代谢的保守转录因子家族。当细胞胆固醇水平低时,SREBP2从内质网转运到高尔基体,在那里它经历蛋白水解活化以产生可溶性N末端片段,驱动脂质生物合成基因的表达。SREBP的功能异常激活与各种代谢异常有关。在这项研究中,我们发现,活性核形式SREBP2(nSREBP2)的过表达会导致各种类型细胞中caspase依赖性裂解细胞死亡。这些细胞表现出典型的变性和坏死特征,包括质膜膨胀和细胞内容物的释放。然而,该表型独立于gasdermin家族蛋白或混合谱系激酶结构域样(MLKL)。转录组分析鉴定nSREBP2诱导p73的表达,其进一步激活胱天蛋白酶。通过全基因组CRISPR-Cas9筛选,我们发现Pannexin-1(PANX1)作用于caspase下游促进膜破裂。半胱天冬酶-3或7在C末端尾部切割PANX1并增加渗透性。PANX1的孔形成活性的抑制减轻了裂解性细胞死亡。在TNF诱导的或化学治疗剂(阿霉素或顺铂)诱导的细胞死亡期间,PANX1可以介导不依赖汽油和MLKL的细胞裂解。一起,这项研究揭示了SREBPs作为程序性细胞死亡的增强剂的非规范功能,并表明PANX1可以直接促进裂解细胞死亡,而与gasdermins和MLKL无关。
    Sterol-regulatory element binding proteins (SREBPs) are a conserved transcription factor family governing lipid metabolism. When cellular cholesterol level is low, SREBP2 is transported from the endoplasmic reticulum to the Golgi apparatus where it undergoes proteolytic activation to generate a soluble N-terminal fragment, which drives the expression of lipid biosynthetic genes. Malfunctional SREBP activation is associated with various metabolic abnormalities. In this study, we find that overexpression of the active nuclear form SREBP2 (nSREBP2) causes caspase-dependent lytic cell death in various types of cells. These cells display typical pyroptotic and necrotic signatures, including plasma membrane ballooning and release of cellular contents. However, this phenotype is independent of the gasdermin family proteins or mixed lineage kinase domain-like (MLKL). Transcriptomic analysis identifies that nSREBP2 induces expression of p73, which further activates caspases. Through whole-genome CRISPR-Cas9 screening, we find that Pannexin-1 (PANX1) acts downstream of caspases to promote membrane rupture. Caspase-3 or 7 cleaves PANX1 at the C-terminal tail and increases permeability. Inhibition of the pore-forming activity of PANX1 alleviates lytic cell death. PANX1 can mediate gasdermins and MLKL-independent cell lysis during TNF-induced or chemotherapeutic reagents (doxorubicin or cisplatin)-induced cell death. Together, this study uncovers a noncanonical function of SREBPs as a potentiator of programmed cell death and suggests that PANX1 can directly promote lytic cell death independent of gasdermins and MLKL.
    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
    癌细胞必须维持其增殖的脂质供应,并通过上调脂肪生成基因程序来做到这一点。固醇调节元件结合蛋白(SREBPs)通过充当脂肪酸和胆固醇合成和摄取所需基因的转录激活因子而充当脂质稳态的调节剂。SREBPs已被认为是多种癌症的化疗靶点,然而,尚不清楚哪些SREBP靶基因对肿瘤发生至关重要。使用平行的体外和体内CRISPR基因敲除筛选,我们确定萜类骨架生物合成基因对胰腺导管腺癌(PDAC)肿瘤的发展至关重要.具体来说,我们鉴定了甲羟戊酸途径的非甾醇类异戊二烯产物,香叶基香叶基二磷酸(G3GPP),作为肿瘤生长的必需脂质。机械上,我们观察到,使用他汀类药物和SREBP抑制剂限制甲羟戊酸途径活性协同诱导细胞凋亡,并导致小G蛋白异戊烯化破坏,对细胞信号传导途径具有多效性作用.最后,我们证明了在原位异种移植小鼠模型中,香叶基香叶基二磷酸合成酶1(GGPS1)敲低可显著降低肿瘤负荷.这些发现表明PDAC肿瘤相对于其他脂质如胆固醇和脂肪酸选择性地需要GGb,并且这是胰腺癌细胞的可靶向脆弱性。
    OBJECTIVE: Cancer cells must maintain lipid supplies for their proliferation and do so by upregulating lipogenic gene programs. The sterol regulatory element-binding proteins (SREBPs) act as modulators of lipid homeostasis by acting as transcriptional activators of genes required for fatty acid and cholesterol synthesis and uptake. SREBPs have been recognized as chemotherapeutic targets in multiple cancers, however it is not well understood which SREBP target genes are essential for tumorigenesis. In this study, we examined the requirement of SREBP target genes for pancreatic ductal adenocarcinoma (PDAC) tumor growth.
    METHODS: Here we constructed a custom CRISPR knockout library containing known SREBP target genes and performed in vitro 2D culture and in vivo orthotopic xenograft CRISPR screens using a patient-derived PDAC cell line. In vitro, we grew cells in medium supplemented with 10% fetal bovine serum (FBS) or 10% lipoprotein-deficient serum (LPDS) to examine differences in gene essentiality in different lipid environments. In vivo, we injected cells into the pancreata of nude mice and collected tumors after 4 weeks.
    RESULTS: We identified terpenoid backbone biosynthesis genes as essential for PDAC tumor development. Specifically, we identified the non-sterol isoprenoid product of the mevalonate pathway, geranylgeranyl diphosphate (GGPP), as an essential lipid for tumor growth. Mechanistically, we observed that restricting mevalonate pathway activity using statins and SREBP inhibitors synergistically induced apoptosis and caused disruptions in small G protein prenylation that have pleiotropic effects on cellular signaling pathways. Finally, we demonstrated that geranylgeranyl diphosphate synthase 1 (GGPS1) knockdown significantly reduces tumor burden in an orthotopic xenograft mouse model.
    CONCLUSIONS: These findings indicate that PDAC tumors selectively require GGPP over other lipids such as cholesterol and fatty acids and that this is a targetable vulnerability of pancreatic cancer cells.
    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
    SREBP转录因子是脂质代谢的中心调节因子。它们的蛋白水解激活需要ER到高尔基体易位,然后通过位点1蛋白酶(S1P)进行切割。作为前蛋白产生,S1P经历从其前体S1PA到成熟S1PC形式的自催化裂解。这里,我们报道了SPRING(以前是C12ORF29)和S1P通过它们的胞外域相互作用,这有助于S1PA自催化裂解为其成熟的S1PC形式。相互,我们在SPRING中鉴定了S1P识别基序,并证明S1P介导的裂解导致细胞中SPRING胞外域的分泌,以及在用AAV-mSpring转导的肝脏特异性Spring敲除(LKO)小鼠中。通过将SPRING变体重建到SPRINGKO细胞中,我们显示SPRING胞外域支持S1P和SREBP信号的蛋白水解成熟,但是S1P介导的SPRING裂解对于这些过程不是必需的。缺乏SPRING会适度减少S1PA→C的蛋白水解成熟和S1PC向高尔基体的运输。然而,尽管在SPRINGKO细胞中到达高尔基体,S1PC未能挽救SREBP信令。值得注意的是,而SREBP信号在SPRINGKO细胞和LKO小鼠中严重减弱,ATF6,另一种S1P底物,在这些模型中没有受到影响。总的来说,我们的研究将SPRING定位为S1P特异性SREBP激活的专用许可因子.
    SREBP transcription factors are central regulators of lipid metabolism. Their proteolytic activation requires ER to the Golgi translocation and subsequent cleavage by site-1-protease (S1P). Produced as a proprotein, S1P undergoes autocatalytic cleavage from its precursor S1PA to mature S1PC form. Here, we report that SPRING (previously C12ORF29) and S1P interact through their ectodomains, and that this facilitates the autocatalytic cleavage of S1PA into its mature S1PC form. Reciprocally, we identified a S1P recognition-motif in SPRING and demonstrate that S1P-mediated cleavage leads to secretion of the SPRING ectodomain in cells, and in liver-specific Spring knockout (LKO) mice transduced with AAV-mSpring. By reconstituting SPRING variants into SPRINGKO cells we show that the SPRING ectodomain supports proteolytic maturation of S1P and SREBP signaling, but that S1P-mediated SPRING cleavage is not essential for these processes. Absence of SPRING modestly diminishes proteolytic maturation of S1PA→C and trafficking of S1PC to the Golgi. However, despite reaching the Golgi in SPRINGKO cells, S1PC fails to rescue SREBP signaling. Remarkably, whereas SREBP signaling was severely attenuated in SPRINGKO cells and LKO mice, that of ATF6, another S1P substrate, was unaffected in these models. Collectively, our study positions SPRING as a dedicated licensing factor for SREBP-specific activation by S1P.
    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
    脊髓小脑共济失调(SCA)包括一组遗传性神经退行性疾病。Machado-Joseph病(MJD)或脊髓小脑共济失调3(SCA3)是最常见的常染色体显性形式,由ataxin-3(ATXN3)基因内CAG重复序列的扩展引起。该突变导致含有长的聚谷氨酰胺(polyQ)延伸的异常蛋白质的表达,所述长的聚谷氨酰胺(polyQ)延伸赋予毒性功能增益,并导致ATXN3在神经元中的错误折叠和聚集。作为神经退行性过程的结果,SCA3患者严重残疾并过早死亡。几种筛查方法,例如,已经进行了药物全基因组和药物库筛选,集中于稳定过表达的ATXN3(polyQ)蛋白的减少和由此产生的毒性的改善。然而,毒性ATXN3的转基因过表达模型,错过了内源性ATXN3调节的潜在调节剂。在使用CRISPR/Cas9修饰的SK-N-SH野生型细胞系鉴定内源ATXN3表达的修饰剂的另一种方法中,在内源ATXN3启动子的控制下,使用GFP-T2A-荧光素酶(LUC)盒,四种他汀类药物被鉴定为潜在的表达激活剂。我们在这里提供了高通量筛选方法的概述,这些方法在不同的SCA3模型生物和细胞系中找到ATXN3(polyQ)毒性的化合物或基因组修饰剂,以改善和阻止患者的SCA3进展。此外,讨论了胆固醇在神经退行性疾病(NDDs)中的一般作用,特别是SCA3。
    The spinocerebellar ataxias (SCA) comprise a group of inherited neurodegenerative diseases. Machado-Joseph Disease (MJD) or spinocerebellar ataxia 3 (SCA3) is the most common autosomal dominant form, caused by the expansion of CAG repeats within the ataxin-3 (ATXN3) gene. This mutation results in the expression of an abnormal protein containing long polyglutamine (polyQ) stretches that confers a toxic gain of function and leads to misfolding and aggregation of ATXN3 in neurons. As a result of the neurodegenerative process, SCA3 patients are severely disabled and die prematurely. Several screening approaches, e.g., druggable genome-wide and drug library screenings have been performed, focussing on the reduction in stably overexpressed ATXN3(polyQ) protein and improvement in the resultant toxicity. Transgenic overexpression models of toxic ATXN3, however, missed potential modulators of endogenous ATXN3 regulation. In another approach to identify modifiers of endogenous ATXN3 expression using a CRISPR/Cas9-modified SK-N-SH wild-type cell line with a GFP-T2A-luciferase (LUC) cassette under the control of the endogenous ATXN3 promotor, four statins were identified as potential activators of expression. We here provide an overview of the high throughput screening approaches yet performed to find compounds or genomic modifiers of ATXN3(polyQ) toxicity in different SCA3 model organisms and cell lines to ameliorate and halt SCA3 progression in patients. Furthermore, the putative role of cholesterol in neurodegenerative diseases (NDDs) in general and SCA3 in particular is discussed.
    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
    作为癌症的重要标志,代谢重编程已被证明在肿瘤的发生中起关键作用,转移和耐药性。在涉及癌症代谢调节的各种信号分子和代谢酶中,氧化还原信号转导和代谢稳态需要两个关键转录因子Nrf1和Nrf2。然而,Nrf1和Nrf2(均分别由Nfe2l1和Nfe2l2编码)对肝癌细胞代谢重编程的调节作用至今尚未得到很好的理解.这里,我们发现HepG2细胞中Nrf1和Nrf2的基因缺失导致不同的代谢重编程。Nrf1α的缺失导致糖酵解增强,减少线粒体耗氧量,增强肝细胞癌细胞中的糖异生和戊糖磷酸途径的激活。对比鲜明,Nrf2的缺失减弱了糖酵解和糖异生途径,但对磷酸戊糖途径没有任何显著影响。此外,Nrf1α的敲除也导致脂肪沉积和氨基酸合成和转运增加,特别是丝氨酸合成,虽然Nrf2缺乏不会导致脂肪沉积,但削弱了氨基酸合成和运输。进一步的实验表明,Nrf1α-/-和Nrf2-/-之间的这种独特的代谢编程是由于Nrf1缺失后PI3K-AKT-mTOR信号通路的大量激活,导致葡萄糖摄取关键基因的表达增加。糖酵解,磷酸戊糖途径,和从头脂质合成,而Nrf2的缺乏通过抑制PI3K-AKT-mTOR途径而导致相反的现象。总之,这些为癌症代谢重编程提供了新的见解,并指导了靶向癌症治疗的新策略的探索。
    As a vital hallmarker of cancer, the metabolic reprogramming has been shown to play a pivotal role in tumour occurrence, metastasis and drug resistance. Amongst a vast variety of signalling molecules and metabolic enzymes involved in the regulation of cancer metabolism, two key transcription factors Nrf1 and Nrf2 are required for redox signal transduction and metabolic homeostasis. However, the regulatory effects of Nrf1 and Nrf2 (both encoded by Nfe2l1 and Nfe2l2, respectively) on the metabolic reprogramming of hepatocellular carcinoma cells have been not well understood to date. Here, we found that the genetic deletion of Nrf1 and Nrf2 from HepG2 cells resulted in distinct metabolic reprogramming. Loss of Nrf1α led to enhanced glycolysis, reduced mitochondrial oxygen consumption, enhanced gluconeogenesis and activation of the pentose phosphate pathway in the hepatocellular carcinoma cells. By striking contrast, loss of Nrf2 attenuated the glycolysis and gluconeogenesis pathways, but with not any significant effects on the pentose phosphate pathway. Moreover, knockout of Nrf1α also caused fat deposition and increased amino acid synthesis and transport, especially serine synthesis, whilst Nrf2 deficiency did not cause fat deposition, but attenuated amino acid synthesis and transport. Further experiments revealed that such distinctive metabolic programming of between Nrf1α-/- and Nrf2-/- resulted from substantial activation of the PI3K-AKT-mTOR signalling pathway upon the loss of Nrf1, leading to increased expression of critical genes for the glucose uptake, glycolysis, the pentose phosphate pathway, and the de novo lipid synthesis, whereas deficiency of Nrf2 resulted in the opposite phenomenon by inhibiting the PI3K-AKT-mTOR pathway. Altogether, these provide a novel insight into the cancer metabolic reprogramming and guide the exploration of a new strategy for targeted cancer therapy.
    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
    孕酮(P4)是一种重要的生殖激素,可作为所有其他内源性类固醇的前体。P4通过与孕酮受体(PR)结合来调节生殖过程中的转录活性。然而,P4在肝脏中的生理作用研究不足。这项研究调查了P4介导的肝脏脂质代谢,P4促进胰岛素抵抗并影响能量代谢。虽然外源性脂质主要来自食物,肝脏从碳水化合物饮食中合成内源性甘油三酯和胆固醇。肝脏从头脂肪生成(DNL)主要由乙酰辅酶A及其生物合成途径决定,其中涉及脂肪酸和胆固醇的合成。虽然P4增加了固醇调节元件结合蛋白1C(SREBP-1C)的肝脏水平,过氧化物酶体增殖物激活受体γ(PPARγ),乙酰辅酶A羧化酶(ACC),和CD36,与P4受体拮抗剂RU486共同处理阻断了这些蛋白质和P4介导的脂肪生成。RNA测序用于评估P4在脂肪生成事件中的作用,比如脂肪肝和脂肪酸代谢,脂蛋白信号,和胆固醇代谢。在高脂饮食的卵巢切除小鼠的肝脏或妊娠小鼠中证实了P4诱导的肝DNL和脂质合成代谢。P4在暴露于P4的小鼠中直接增加脂肪生成,在暴露于母体P4的胎儿中间接增加脂肪生成。脂肪生成和脂解之间的脂质平衡决定了脂肪的积累,并与脂质代谢功能障碍有关。其中涉及脂肪的分解和储存能量以及结构和功能脂质的合成。因此,P4可能影响妊娠期间的脂质代谢和生殖发育。
    Progesterone (P4) is a crucial reproductive hormone that acts as a precursor for all other endogenous steroids. P4 modulates transcriptional activity during reproduction by binding to progesterone receptors (PR). However, the physiological role of P4 in the liver is understudied. P4-mediated lipid metabolism in the liver was investigated in this study, as P4 facilitates insulin resistance and influences energy metabolism. While exogenous lipids are mainly obtained from food, the liver synthesizes endogenous triglycerides and cholesterol from a carbohydrate diet. Hepatic de novo lipogenesis (DNL) is primarily determined by acetyl-CoA and its biosynthetic pathways, which involve fatty acid and cholesterol synthesis. While P4 increased the hepatic levels of sterol regulatory element-binding protein 1 C (SREBP-1 C), peroxisome proliferator-activated receptor-gamma (PPARγ), acetyl-CoA carboxylase (ACC), and CD36, co-treatment with the P4 receptor antagonist RU486 blocked these proteins and P4-mediated lipogenesis. RNA sequencing was used to assess the role of P4 in lipogenic events, such as fatty liver and fatty acid metabolism, lipoprotein signaling, and cholesterol metabolism. P4 induced hepatic DNL and lipid anabolism were confirmed in the liver of ovarian resection mice fed a high-fat diet or in pregnant mice. P4 increased lipogenesis directly in mice exposed to P4 and indirectly in fetuses exposed to maternal P4. The lipid balance between lipogenesis and lipolysis determines fat build-up and is linked to lipid metabolism dysfunction, which involves the breakdown and storage of fats for energy and the synthesis of structural and functional lipids. Therefore, P4 may impact the lipid metabolism and reproductive development during gestation.
    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
    脂质合成受Scap的作用调节,内质网(ER)膜中结合胆固醇的多位膜蛋白。当ER胆固醇水平低时,Scap激活SREBPs,上调胆固醇合成基因的转录因子,脂肪酸,和甘油三酯。当ER胆固醇水平升高时,固醇与Scap结合,触发构象变化,阻止SREBPs活化和脂质合成停止。为了获得对胆固醇如何调节Scap/SREBP机器的分子理解,并确定脂质代谢失调的治疗方法,需要特异性结合和抑制Scap的胆固醇模拟化合物。为了实现这个目标,我们专注于AnthrolysinO(ALO),一种与胆固醇结合的成孔细菌毒素,其特异性和敏感性与Scap异常相似。我们推断结合和抑制ALO的小分子也可能抑制Scap。高通量筛选一个~300,000个化合物文库中发现的ALO结合一分子,称为UT-59,与Scap的胆固醇结合位点结合。在绑定时,UT-59触发与胆固醇诱导的Scap相同的构象变化,并阻断SREBP的激活和培养细胞中的脂肪生成。UT-59还抑制小鼠肝脏中的SREBP激活。与之前报道的5种SREBP激活抑制剂不同,UT-59是唯一通过结合Scap的胆固醇结合位点而特异性起作用的。我们确定特定Scap抑制剂如UT-59的方法在开发源于SREBP激活升高的人类疾病的治疗线索方面具有很大的希望。比如脂肪肝和某些癌症。
    Lipid synthesis is regulated by the actions of Scap, a polytopic membrane protein that binds cholesterol in membranes of the endoplasmic reticulum (ER). When ER cholesterol levels are low, Scap activates SREBPs, transcription factors that upregulate genes for synthesis of cholesterol, fatty acids, and triglycerides. When ER cholesterol levels rise, the sterol binds to Scap, triggering conformational changes that prevent activation of SREBPs and halting synthesis of lipids. To achieve a molecular understanding of how cholesterol regulates the Scap/SREBP machine and to identify therapeutics for dysregulated lipid metabolism, cholesterol-mimetic compounds that specifically bind and inhibit Scap are needed. To accomplish this goal, we focused on Anthrolysin O (ALO), a pore-forming bacterial toxin that binds cholesterol with a specificity and sensitivity that is uncannily similar to Scap. We reasoned that a small molecule that would bind and inhibit ALO might also inhibit Scap. High-throughput screening of a ~300,000-compound library for ALO-binding unearthed one molecule, termed UT-59, which binds to Scap\'s cholesterol-binding site. Upon binding, UT-59 triggers the same conformation changes in Scap as those induced by cholesterol and blocks activation of SREBPs and lipogenesis in cultured cells. UT-59 also inhibits SREBP activation in the mouse liver. Unlike five previously reported inhibitors of SREBP activation, UT-59 is the only one that acts specifically by binding to Scap\'s cholesterol-binding site. Our approach to identify specific Scap inhibitors such as UT-59 holds great promise in developing therapeutic leads for human diseases stemming from elevated SREBP activation, such as fatty liver and certain cancers.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号