cholesterol biosynthesis

胆固醇生物合成
  • 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
    目的:胆固醇代谢重编程被认为是癌症的一个新特征。胰腺导管腺癌(PDAC)是一种对胆固醇需求高的癌症。探讨了PDAC中胆固醇代谢抑制的潜在机制。
    方法:在TCGA数据库中证实了PDAC与胆固醇之间的相关性。在TCGA和GEO数据集中发现了表达和临床关联。采用敲低和过表达AGFG1进行功能研究。RNA测序,胆固醇检测,透射电子显微镜,免疫共沉淀,和免疫荧光等。被用来揭示潜在的机制。
    结果:生物信息学分析显示,AGFG1基因与PDAC中的胆固醇代谢呈正相关。然后发现AGFG1表达与PDAC的不良预后相关。AGFG1敲低导致肿瘤细胞在体外和体内的增殖降低。通过RNA测序,我们发现AGFG1上调表达导致细胞内胆固醇生物合成增强。AGFG1敲低抑制胆固醇生物合成和胆固醇在ER中的积累。机械上,我们证实AGFG1与CAV1相互作用以重新定位胆固醇进行胆固醇生物合成,因此导致细胞内胆固醇代谢紊乱。
    结论:我们的研究证明了AGFG1通过干扰PDAC中胆固醇代谢引起的肿瘤促进作用。我们的研究提出了基于PDAC中胆固醇代谢的癌症治疗方法的新视角。
    OBJECTIVE: Cholesterol metabolism reprograming has been acknowledged as a novel feature of cancers. Pancreatic ductal adenocarcinoma (PDAC) is a cancer with a high demand of cholesterol for rapid growth. The underlying mechanism of how cholesterol metabolism homestasis are disturbed in PDAC is explored.
    METHODS: The relevance between PDAC and cholesterol was confirmed in TCGA database. The expression and clinical association were discovered in TCGA and GEO datasets. Knockdown and overexpression of AGFG1 was adopted to perform function studies. RNA sequencing, cholesterol detection, transmission electron microscope, co-immunoprecipitation, and immunofluorescence et al. were utilized to reveal the underlying mechanism.
    RESULTS: AGFG1 was identified as one gene positively correlated with cholesterol metabolism in PDAC as revealed by bioinformatics analysis. AGFG1 expression was then found associated with poor prognosis in PDAC. AGFG1 knockdown led to decreased proliferation of tumor cells both in vitro and in vivo. By RNA sequencing, we found AGFG1 upregulated expression leads to enhanced intracellular cholesterol biosynthesis. AGFG1 knockdown suppressed cholesterol biosynthesis and an accumulation of cholesterol in the ER. Mechanistically, we confirmed that AGFG1 interacted with CAV1 to relocate cholesterol for the proceeding of cholesterol biosynthesis, therefore causing disorders in intracellular cholesterol metabolism.
    CONCLUSIONS: Our study demonstrates the tumor-promoting role of AGFG1 by disturbing cholesterol metabolism homestasis in PDAC. Our study has present a new perspective on cancer therapeutic approach based on cholerstrol metabolism in PDAC.
    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:慢性前列腺炎/慢性盆腔疼痛综合征(CP/CPPS)在世界范围内非常普遍,饮酒是一个显著的促成因素。研究表明,肠道菌群可以受到饮酒的影响,并且是调节Th17细胞免疫的重要介质。然而,目前尚不清楚酒精加剧CP/CPPS的确切机制以及肠道菌群在这一过程中的作用。
    方法:我们首先构建了最常用的CP/CPPS动物模型,实验性自身免疫性前列腺炎(EAP)模型,通过免疫测定。基于此,将小鼠分为EAP组和饮酒EAP组。通过16SrRNA测序和非靶向代谢组学分析,确定了两组之间不同的肠道微生物群及其代谢产物。随后,我们进行了针对胆固醇的代谢组学检测,以确定胆固醇的确切差异.此外,采用流式细胞术和免疫组织化学等多种方法检测Th17细胞的分化状态和27-羟胆固醇(差异胆固醇)及其上游调节因子-甾醇调节元件结合蛋白2(SREBP2)治疗前列腺炎的严重程度.最后,进行粪便移植以初步研究酒精摄入是否会加剧免疫受体小鼠的EAP。
    结果:酒精摄入增加了Th17细胞的比例和相关炎症因子的水平。它还导致改变的肠道细菌丰富度和增加的肠道通透性。进一步的代谢组学分析显示,EAP与酒精喂养的EAP小鼠在多种代谢产物上存在显著差异。代谢途径富集分析表明,与胆固醇合成和代谢相关的途径显著富集,随后通过检测代谢酶的表达来证实。通过靶向胆固醇合成,酒精喂养的EAP小鼠中27-羟基胆固醇显着增加。随后的机制研究表明,补充27-羟基胆固醇可以在体内和体外加重EAP并促进Th17细胞分化,它由SREBP2调节。此外,我们观察到,酒精摄入小鼠的粪便移植会加重正常饮食的免疫受体小鼠的EAP。
    结论:我们的研究首次表明酒精摄入促进Th17细胞分化,并通过微生物来源的胆固醇生物合成加剧EAP。
    BACKGROUND: Chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) is very common worldwide, and alcohol consumption is a notable contributing factor. Researches have shown that gut microbiota can be influenced by alcohol consumption and is an important mediator in regulating Th17 cell immunity. However, it is still unclear the exact mechanism by which alcohol exacerbates the CP/CPPS and the role of gut microbiota in this process.
    METHODS: We first constructed the most-commonly used animal model for CP/CPPS, the experimental autoimmune prostatitis (EAP) model, through immunoassay. Based on this, mice were divided into EAP group and alcohol-consuming EAP group. By 16S rRNA sequencing and non-targeted metabolomics analysis, differential gut microbiota and their metabolites between the two groups were identified. Subsequently, metabolomics detection targeting cholesterols was carried out to identify the exact difference in cholesterol. Furthermore, multiple methods such as flow cytometry and immunohistochemistry were used to detect the differentiation status of Th17 cells and severity of prostatitis treated with 27-hydroxycholesterol (the differential cholesterol) and its upstream regulatory factor-sterol regulatory element-binding protein 2 (SREBP2). Lastly, fecal transplantation was conducted to preliminary study on whether alcohol intake exacerbates EAP in immune receptor mice.
    RESULTS: Alcohol intake increased the proportion of Th17 cells and levels of related inflammatory factors. It also led to an altered gut bacterial richness and increased gut permeability. Further metabolomic analysis showed that there were significant differences in a variety of metabolites between EAP and alcohol-fed EAP mice. Metabolic pathway enrichment analysis showed that the pathways related to cholesterol synthesis and metabolism were significantly enriched, which was subsequently confirmed by detecting the expression of metabolic enzymes. By targeting cholesterol synthesis, 27-hydroxycholesterol was significantly increased in alcohol-fed EAP mice. Subsequent mechanistic research showed that supplementation with 27-hydroxycholesterol could aggravate EAP and promote Th17 cell differentiation both in vivo and in vitro, which is regulated by SREBP2. In addition, we observed that fecal transplantation from mice with alcohol intake aggravated EAP in immunized recipient mice fed a normal diet.
    CONCLUSIONS: Our study is the first to show that alcohol intake promotes Th17 cell differentiation and exacerbates EAP through microbiota-derived 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
    癌症干细胞(CSC)在癌症发作和复发中起重要作用。反常的铁和脂质代谢已被证明在CSC,表明铁性凋亡,最近发现的一种以脂质过氧化为特征的调节细胞死亡形式,可能对CSC产生重大影响。然而,铁凋亡在胃癌干细胞(GCSCs)中的确切作用尚不清楚.为了解决这个差距,我们使用癌症基因组图谱筛选了GCSCs中的铁凋亡相关基因,并通过定量聚合酶链反应和蛋白质印迹证实了我们的发现.这些结果表明,硬脂酰辅酶A去饱和酶(SCD1)是GCSC中铁凋亡调节的关键参与者。这项研究提供了证据,证明SCD1通过消除P53的转录抑制来正向调节角鲨烯环氧酶(SQLE)的转录。该机制增加胆固醇含量,并且由SCD1调节的升高的胆固醇通过mTOR信号通路抑制铁凋亡。此外,我们的体内研究表明,SCD1敲除或胆固醇摄入的调节会影响GCSCs的干性及其对铁凋亡诱导物的敏感性.因此,靶向SCD1/角鲨烯环氧酶/胆固醇信号轴与铁凋亡诱导物联合可能代表基于GCSC治疗胃癌的有希望的治疗方法.
    Cancer stem cells (CSCs) play a substantial role in cancer onset and recurrence. Anomalous iron and lipid metabolism have been documented in CSCs, suggesting that ferroptosis, a recently discovered form of regulated cell death characterised by lipid peroxidation, could potentially exert a significant influence on CSCs. However, the precise role of ferroptosis in gastric cancer stem cells (GCSCs) remains unknown. To address this gap, we screened ferroptosis-related genes in GCSCs using The Cancer Genome Atlas and corroborated our findings through quantitative polymerase chain reaction and western blotting. These results indicate that stearoyl-CoA desaturase (SCD1) is a key player in the regulation of ferroptosis in GCSCs. This study provides evidence that SCD1 positively regulates the transcription of squalene epoxidase (SQLE) by eliminating transcriptional inhibition of P53. This mechanism increases the cholesterol content and the elevated cholesterol regulated by SCD1 inhibits ferroptosis via the mTOR signalling pathway. Furthermore, our in vivo studies showed that SCD1 knockdown or regulation of cholesterol intake affects the stemness of GCSCs and their sensitivity to ferroptosis inducers. Thus, targeting the SCD1/squalene epoxidase/cholesterol signalling axis in conjunction with ferroptosis inducers may represent a promising therapeutic approach for the treatment of gastric cancer based on GCSCs.
    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
    肝细胞癌(HCC)是一种发生在肝脏的恶性肿瘤,恶性程度高,预后相对较差。绞股蓝皂苷L对肝癌细胞有抑制作用。然而,其作用机制尚不清楚。本研究旨在探讨绞股蓝皂苷L对肝癌的体外和体内抑制作用。并探索其潜在机制。结果表明,绞股蓝皂苷L降低了HepG2和Huh-7细胞的胆固醇和甘油三酯含量,抑制细胞增殖,侵袭和转移,细胞周期停滞在G0/G1期,促进细胞凋亡。机械上,靶向转录因子SREPB2抑制HMGCS1蛋白的表达,抑制下游蛋白HMGCR和MVK,从而调节甲羟戊酸(MVA)途径。过表达HMGCS1导致HCC胆固醇代谢途径的显著改变,它介导HCC细胞增殖并赋予对绞股蓝皂苷L的治疗作用的抗性。绞股蓝皂苷L通过减少胆固醇产生有效抑制荷瘤小鼠肝癌生长,表现出良好的安全性和最小的毒副作用。绞股蓝皂苷L调节胆固醇稳态,通过调节MHCI途径相关蛋白增强炎症因子的表达以增强抗癌免疫应答。来自HCC患者的临床样品在肿瘤组织中也显示出MVA途径相关基因的高表达水平。这些发现强调了绞股蓝皂苷L作为靶向HCC胆固醇代谢的有前途的药物,同时强调了调节SREBP2-HMGCS1轴作为治疗策略的有效性。
    Hepatocellular carcinoma (HCC) is a malignant tumor that occurs in the liver, with a high degree of malignancy and relatively poor prognosis. Gypenoside L has inhibitory effects on liver cancer cells. However, its mechanism of action is still unclear. This study aims to investigate the inhibitory effects of gypenoside L on HCC in vitro and in vivo, and explore its potential mechanisms. The results showed that gypenoside L reduced the cholesterol and triglyceride content in HepG2 and Huh-7 cells, inhibited cell proliferation, invasion and metastasis, arrested cell cycle at G0/G1 phase, promoted cell apoptosis. Mechanistically, it targeted the transcription factor SREPB2 to inhibit the expression of HMGCS1 protein and inhibited the downstream proteins HMGCR and MVK, thereby regulating the mevalonate (MVA) pathway. Overexpression HMGCS1 led to significant alterations in the cholesterol metabolism pathway of HCC, which mediated HCC cell proliferation and conferred resistance to the therapeutic effect of gypenoside L. In vivo, gypenoside L effectively suppressed HCC growth in tumor-bearing mice by reducing cholesterol production, exhibiting favorable safety profiles and minimal toxic side effects. Gypenoside L modulated cholesterol homeostasis, enhanced expression of inflammatory factors by regulating MHC I pathway-related proteins to augment anticancer immune responses. Clinical samples from HCC patients also exhibited high expression levels of MVA pathway-related genes in tumor tissues. These findings highlight gypenoside L as a promising agent for targeting cholesterol metabolism in HCC while emphasizing the effectiveness of regulating the SREBP2-HMGCS1 axis as a therapeutic strategy.
    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: Preprint
    不均匀(DVL)蛋白质家族在质膜的细胞质界面形成超分子蛋白质和脂质复合物,以调节组织模式,扩散,细胞极性,和通过DVL依赖性信号传导的致癌过程,如Wnt/β-catenin。虽然DVL与胆固醇结合是其膜缔合所必需的,DVL-甾醇联合的具体结构要求和细胞影响尚不清楚.我们报告说,在正常和病理条件下积累的细胞内固醇会导致DVL活性异常。计算机模拟和分子分析表明DVL-PDZ结构域内β-和α-甾醇面的取向调节DVL-甾醇结合。细胞内积累的自然发生的甾醇受损DVL2质膜结合,通过Foxk2诱导DVL2核定位。细胞内甾醇的变化也选择性受损DVL2蛋白-蛋白相互作用这项工作确定了甾醇特异性作为DVL信号的调节剂,表明细胞内甾醇对DVL活性产生不同的影响,并支持细胞内甾醇稳态在细胞信号传导中的作用。
    The Dishevelled (DVL) family of proteins form supramolecular protein and lipid complexes at the cytoplasmic interface of the plasma membrane to regulate tissue patterning, proliferation, cell polarity, and oncogenic processes through DVL-dependent signaling, such as Wnt/β-catenin. While DVL binding to cholesterol is required for its membrane association, the specific structural requirements and cellular impacts of DVL-sterol association are unclear. We report that intracellular sterols which accumulate within normal and pathological conditions cause aberrant DVL activity. In silico and molecular analyses suggested orientation of the β- and α-sterol face within the DVL-PDZ domain regulates DVL-sterol binding. Intracellular accumulation of naturally occurring sterols impaired DVL2 plasma membrane association, inducing DVL2 nuclear localization via Foxk2. Changes to intracellular sterols also selectively impaired DVL2 protein-protein interactions This work identifies sterol specificity as a regulator of DVL signaling, suggests intracellular sterols cause distinct impacts on DVL activity, and supports a role for intracellular sterol homeostasis in cell signaling.
    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
    溶血磷脂酸(LPA)物种,在肿瘤微环境(TME)中普遍存在,对各种癌症产生不利影响。在卵巢癌中,18:0和20:4LPA物种选择性地与较短的无复发生存期相关,表明对蜂窝信令网络的不同影响。巨噬细胞代表TME中高度相关的细胞类型,但是LPA对这些细胞的影响仍然不清楚。这里,我们通过无偏倚的磷酸化蛋白质组学发现了人类单核细胞衍生的巨噬细胞中不同的LPA物种特异性反应,87和161个磷酸位点上调20:4和18:0LPA,分别,只有24个共享网站。对于下调的磷酸位点(163对5个位点),特异性甚至更显著。考虑到TME中高水平的20:4LPA及其与低生存率的选择性关联,这一发现可能会产生重大影响。路径分析确定RHO/RAC1GTP酶信号传导为主要受影响的靶标,包括AHRGEF和DOCK鸟嘌呤交换因子,ARHGAPGTPase激活蛋白,和调节蛋白激酶。与这些发现一致,暴露于20:4导致肌动蛋白丝网络的强烈改变,并因此增强了巨噬细胞的迁移。此外,20:4LPA诱导p38磷酸化,18:0LPA未反映的响应,而AKT的模式逆转。此外,RNA谱分析将参与胆固醇/脂质代谢的基因鉴定为20:4LPA的选择性靶标。这些发现暗示两种LPA物种协同调节不同的途径以支持TME内的促肿瘤性巨噬细胞所必需的功能。这些包括通过AKT激活的细胞存活和通过RHO/RAC1和p38信号传导的迁移。
    Lysophosphatidic acid (LPA) species, prevalent in the tumor microenvironment (TME), adversely impact various cancers. In ovarian cancer, the 18:0 and 20:4 LPA species are selectively associated with shorter relapse-free survival, indicating distinct effects on cellular signaling networks. Macrophages represent a cell type of high relevance in the TME, but the impact of LPA on these cells remains obscure. Here, we uncovered distinct LPA-species-specific responses in human monocyte-derived macrophages through unbiased phosphoproteomics, with 87 and 161 phosphosites upregulated by 20:4 and 18:0 LPA, respectively, and only 24 shared sites. Specificity was even more pronounced for downregulated phosphosites (163 versus 5 sites). Considering the high levels 20:4 LPA in the TME and its selective association with poor survival, this finding may hold significant implications. Pathway analysis pinpointed RHO/RAC1 GTPase signaling as the predominantly impacted target, including AHRGEF and DOCK guanine exchange factors, ARHGAP GTPase activating proteins, and regulatory protein kinases. Consistent with these findings, exposure to 20:4 resulted in strong alterations to the actin filament network and a consequent enhancement of macrophage migration. Moreover, 20:4 LPA induced p38 phosphorylation, a response not mirrored by 18:0 LPA, whereas the pattern for AKT was reversed. Furthermore, RNA profiling identified genes involved in cholesterol/lipid metabolism as selective targets of 20:4 LPA. These findings imply that the two LPA species cooperatively regulate different pathways to support functions essential for pro-tumorigenic macrophages within the TME. These include cellular survival via AKT activation and migration through RHO/RAC1 and p38 signaling.
    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
    导管原位癌(DCIS)是浸润性乳腺癌的非强制性前体。然而,如果不及时治疗,约50%的DCIS进展。防止这种进展至关重要。累积证据表明甲羟戊酸级联,胆固醇生物合成的核心,有助于调节Hippo信号通路,提供GTP酶激活所需的类异戊二烯,Yes相关蛋白(YAP)/转录共激活因子与PDZ结合基序(TAZ)共激活因子的核积累,以及随后的基因转录以及这种合作的破坏与肿瘤进展有关。
    在这项计算机模拟研究中,我们调查了在正常乳腺上皮转化为DCIS的过程中是否已经发生了这种破坏.为了这个目标,我们询问了一个公开的数据集,我们在一组患者匹配的DCIS和相应的组织学正常(HN)上皮样本中,探索了参与从头胆固醇生物合成的基因之间的相互关系,以及与编码Hippo信号通路核心成分的基因之间的关联。
    大多数参与胆固醇生物合成的基因在DCIS中的表达高于相应的HN上皮。这种差异表达与它们的相关性谱的实质性变化有关。特别是,3-羟基-3-甲基戊二酰辅酶A还原酶(HMGCR)和INSIG1失去了HN上皮中显示的正相关,它们与LSS的负相关转变为正相关。此外,GGPS1在类异戊二烯生产中起着至关重要的作用,显著改变了其相关性。GGPS1和HMGCR或INSIG1之间的正相关消失,而与SQLE的正相关,这推动了对胆固醇的不可逆转的承诺,在DCIS中切换为负数。
    目前的发现证实了以下假设:功能失调的甲羟戊酸途径可能通过导致类异戊二烯的异常产生而与DCIS的发展相一致。进而激活GTP酶并促进YAP/TAZ核易位,并建议他汀类药物的安全和低成本治疗作为对比这种代谢功能障碍的可能的获胜策略。
    UNASSIGNED: Ductal carcinoma in situ (DCIS) is a non-obligate precursor to invasive breast cancer. However, if left untreated, about 50% of DCIS progress. Preventing such a progression is of paramount importance. Cumulative evidence indicated that the mevalonate cascade, the core of cholesterol biosynthesis, contributes to the regulation of the Hippo signaling pathway providing the isoprenoids required for GTPase activation, the nuclear accumulation of the Yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) coactivator, and the subsequent gene transcription and that the disruption of this cooperation associated with tumor progression.
    UNASSIGNED: In this in silico study, we investigated whether such a disruption occurred already during the transformation of the normal mammary epithelium into DCIS. To this aim, we interrogated a publicly available dataset, and we explored the interrelationship of the genes involved in the de novo cholesterol biosynthesis and the association with those coding for the core components of the Hippo signaling pathway in a set of patient-matched samples of DCIS and corresponding histologically normal (HN) epithelium.
    UNASSIGNED: Most genes involved in cholesterol biosynthesis were more expressed in DCIS than in the corresponding HN epithelium. This differential expression was associated with a substantial change in their correlation profile. In particular, 3-hydroxy-3-methylglutaryl coenzyme-A reductase (HMGCR) and INSIG1 lost the positive association shown in the HN epithelium, and their negative association with LSS switched to a positive one. Also, GGPS1, which plays a crucial role in isoprenoids production, significantly changed its correlation profile. The positive association between GGPS1 and HMGCR or INSIG1 disappeared, whereas the positive association with SQLE, which drives the irreversible commitment to cholesterol, switched to a negative one in DCIS.
    UNASSIGNED: Present findings corroborated the hypothesis that a dysfunctional mevalonate pathway possibly concurs with DCIS development by leading to abnormal production of isoprenoids, which in turn activate GTPases and promote YAP/TAZ nuclear translocation, and suggested the safe and low-cost treatment with statins as the possible winning strategy to contrast this metabolic dysfunction.
    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
    由于缺乏合适的小鼠模型,对急性髓细胞性白血病(AML)p53突变背后的机制的研究受到限制。历史上导致淋巴瘤而不是白血病。本研究介绍了两种新的AML小鼠模型。一个模型在早期发育中诱导突变型p53和Mdm2单倍体不足,显示Mdm2在骨髓偏向造血和AML易感性中的作用,独立于p53第二个模型通过在成人造血干细胞中诱导突变型p53来模拟克隆造血,证明p53突变的时间决定了AML与淋巴瘤的发展。在这种情况下,与年龄相关的造血干细胞(HSCs)的改变与突变型p53共同作用,导致髓样转化而不是淋巴瘤的发生.我们的研究揭示了HSC年龄合作影响的新见解,Trp53突变,和Mdm2单倍体不足对克隆造血和骨髓恶性肿瘤的发展。
    The investigation of the mechanisms behind p53 mutations in acute myeloid leukemia (AML) has been limited by the lack of suitable mouse models, which historically have resulted in lymphoma rather than leukemia. This study introduces two new AML mouse models. One model induces mutant p53 and Mdm2 haploinsufficiency in early development, showing the role of Mdm2 in myeloid-biased hematopoiesis and AML predisposition, independent of p53. The second model mimics clonal hematopoiesis by inducing mutant p53 in adult hematopoietic stem cells, demonstrating that the timing of p53 mutation determines AML vs. lymphoma development. In this context, age-related changes in hematopoietic stem cells (HSCs) collaborate with mutant p53 to predispose toward myeloid transformation rather than lymphoma development. Our study unveils new insights into the cooperative impact of HSC age, Trp53 mutations, and Mdm2 haploinsufficiency on clonal hematopoiesis and the development of myeloid malignancies.
    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
    2019年冠状病毒病(COVID-19)是由病原体严重急性呼吸综合征冠状病毒2(SARS-CoV-2)引起的。随着SARS-CoV-2新变种的反复流行,COVID-19仍然是一个全球公共卫生问题,并且仍然需要新的抗病毒药物。一些胆固醇衍生物,如25-羟基胆固醇,已知对多种包膜和无包膜病毒具有抗病毒活性,包括SARS-CoV-2.在SARS-CoV-2感染的进入阶段,病毒包膜与依赖病毒刺突(S)糖蛋白的宿主膜融合。从胆固醇衍生物的筛选,我们发现了一种新的化合物26,27-dinorcholest-5-en-24-yne-3β,在合胞体形成测定中抑制SARS-CoV-2S蛋白依赖性膜融合的20-二醇(Nat-20(S)-yne)。Nat-20(S)-yne以剂量依赖的方式表现出SARS-CoV-2假病毒进入和完整的SARS-CoV-2感染的抑制活性。在SARS-CoV-2的变种中,三角洲和武汉菌株对Nat-20(S)-yne感染的抑制作用更强,主要通过质膜融合侵入细胞,而不是omicron菌株。S蛋白的受体结合域与宿主受体ACE2之间的相互作用不受Nat-20(S)-yne的影响。不像25-羟基胆固醇,调节胆固醇代谢的各个步骤,Nat-20(S)-炔仅抑制从头胆固醇生物合成。因此,Nat-20(S)-yne处理细胞的质膜胆固醇含量显著降低,导致抑制SARS-CoV-2感染。具有新作用机制的Nat-20(S)-yne可能是COVID-19的潜在治疗候选药物。
    The coronavirus disease 2019 (COVID-19) is caused by the etiological agent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19, with the recurrent epidemics of new variants of SARS-CoV-2, remains a global public health problem, and new antivirals are still required. Some cholesterol derivatives, such as 25-hydroxycholesterol, are known to have antiviral activity against a wide range of enveloped and non-enveloped viruses, including SARS-CoV-2. At the entry step of SARS-CoV-2 infection, the viral envelope fuses with the host membrane dependent of viral spike (S) glycoproteins. From the screening of cholesterol derivatives, we found a new compound 26,27-dinorcholest-5-en-24-yne-3β,20-diol (Nat-20(S)-yne) that inhibited the SARS-CoV-2 S protein-dependent membrane fusion in a syncytium formation assay. Nat-20(S)-yne exhibited the inhibitory activities of SARS-CoV-2 pseudovirus entry and intact SARS-CoV-2 infection in a dose-dependent manner. Among the variants of SARS-CoV-2, inhibition of infection by Nat-20(S)-yne was stronger in delta and Wuhan strains, which predominantly invade into cells via fusion at the plasma membrane, than in omicron strains. The interaction between receptor-binding domain of S proteins and host receptor ACE2 was not affected by Nat-20(S)-yne. Unlike 25-hydroxycholesterol, which regulates various steps of cholesterol metabolism, Nat-20(S)-yne inhibited only de novo cholesterol biosynthesis. As a result, plasma membrane cholesterol content was substantially decreased in Nat-20(S)-yne-treated cells, leading to inhibition of SARS-CoV-2 infection. Nat-20(S)-yne having a new mechanism of action may be a potential therapeutic candidate for COVID-19.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号