UTR, Untranslated region

UTR,未翻译区域
  • 文章类型: Journal Article
    由P4-ATP酶调节的膜不对称性对于真核细胞的功能至关重要。特定脂质的潜在空间易位或翻转通常通过与符合的非催化亚基偶联的相应P4-ATP酶来确保。我们以前的工作已经确定了细胞内原生动物病原体中的五种P4-ATPases(TgP4-ATPase1-5)和三种非催化伴侣蛋白(TgLem1-3),弓形虫.然而,他们的翻转活动,生理相关性和功能偶联仍然未知。在这里,我们证明了TgP4-ATPase1和TgLem1在弓形虫的裂解周期中共同作用以转运磷脂酰丝氨酸(PtdSer)。两种蛋白质均位于其急性感染速殖子阶段的侵入性(顶端)末端的质膜中。速殖子中P4-ATPase1的遗传敲除和Lem1的条件耗竭严重破坏了PtdSer的无性繁殖和跨质膜易位。此外,个体突变体的表型分析揭示了脂质翻转对运动性的要求,速殖子的出口和入侵。并非最不重要的,邻近依赖的生物素化和相互免疫沉淀试验证明了P4-ATPase1和Lem1的物理相互作用。我们的发现揭示了裂解周期中PtdSer翻转的机制和意义,并确定了P4-ATPase1-Lem1异质复合物作为弓形虫的潜在药物靶标。
    The membrane asymmetry regulated by P4-ATPases is crucial for the functioning of eukaryotic cells. The underlying spatial translocation or flipping of specific lipids is usually assured by respective P4-ATPases coupled to conforming non-catalytic subunits. Our previous work has identified five P4-ATPases (TgP4-ATPase1-5) and three non-catalytic partner proteins (TgLem1-3) in the intracellular protozoan pathogen, Toxoplasma gondii. However, their flipping activity, physiological relevance and functional coupling remain unknown. Herein, we demonstrate that TgP4-ATPase1 and TgLem1 work together to translocate phosphatidylserine (PtdSer) during the lytic cycle of T. gondii. Both proteins localize in the plasma membrane at the invasive (apical) end of its acutely-infectious tachyzoite stage. The genetic knockout of P4-ATPase1 and conditional depletion of Lem1 in tachyzoites severely disrupt the asexual reproduction and translocation of PtdSer across the plasma membrane. Moreover, the phenotypic analysis of individual mutants revealed a requirement of lipid flipping for the motility, egress and invasion of tachyzoites. Not least, the proximity-dependent biotinylation and reciprocal immunoprecipitation assays demonstrated the physical interaction of P4-ATPase1 and Lem1. Our findings disclose the mechanism and significance of PtdSer flipping during the lytic cycle and identify the P4-ATPase1-Lem1 heterocomplex as a potential drug target in T. gondii.
    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
    磷酸三糖异构酶缺乏症(TPIDf)是一种罕见的,侵袭性遗传疾病,通常影响幼儿,目前还没有确定的治疗方法。TPIDf的特征是溶血性贫血,进行性神经肌肉变性,寿命明显缩短。该疾病主要使用无脊椎动物和体外模型进行研究,缺乏人类疾病的关键方面。虽然其他群体已经产生了哺乳动物Tpi1突变株,特别是老鼠的musmusculus,这些没有概括人类疾病的关键特征表型。这里报道的是TPIDf的新型鼠模型的产生。CRISPR-Cas9被用来设计最常见的人类致病突变,Tpi1E105D,和Tpi1空小鼠也被分离为移码缺失。Tpi1E105D/null小鼠的寿命明显缩短,姿势异常与广泛的神经肌肉功能障碍一致,溶血性贫血,脾脏病理变化,体重下降。与野生型同窝动物相比,Tpi1E105D/null动物的TPI蛋白水平降低了95%,与TPI蛋白稳定性下降一致,TPIDf的已知原因。这项工作说明了Tpi1E105D/空小鼠作为人TPIDf的哺乳动物模型的能力。这项工作将允许在具有类似于人类的生理学的模型中推进TPIDf的研究。本文报道的模型的开发将使疾病发病机理的机理研究成为可能,重要的是,用于新兴TPIDf治疗的哺乳动物系统中的功效测试。
    Triosephosphate isomerase deficiency (TPI Df) is a rare, aggressive genetic disease that typically affects young children and currently has no established treatment. TPI Df is characterized by hemolytic anemia, progressive neuromuscular degeneration, and a markedly reduced lifespan. The disease has predominately been studied using invertebrate and in vitro models, which lack key aspects of the human disease. While other groups have generated mammalian Tpi1 mutant strains, specifically with the mouse mus musculus, these do not recapitulate key characteristic phenotypes of the human disease. Reported here is the generation of a novel murine model of TPI Df. CRISPR-Cas9 was utilized to engineer the most common human disease-causing mutation, Tpi1 E105D , and Tpi1 null mice were also isolated as a frame-shifting deletion. Tpi1 E105D/null mice experience a markedly shortened lifespan, postural abnormalities consistent with extensive neuromuscular dysfunction, hemolytic anemia, pathological changes in spleen, and decreased body weight. There is a ∼95% reduction in TPI protein levels in Tpi1 E105D/null animals compared to wild-type littermates, consistent with decreased TPI protein stability, a known cause of TPI Df. This work illustrates the capability of Tpi1 E105D/null mice to serve as a mammalian model of human TPI Df. This work will allow for advancement in the study of TPI Df within a model with physiology similar to humans. The development of the model reported here will enable mechanistic studies of disease pathogenesis and, importantly, efficacy testing in a mammalian system for emerging TPI Df treatments.
    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
    未经证实:皮瓣部分坏死在整形和重建手术中仍然是一个重大问题。在这项研究中,研究了miR-590-3p在脂肪干细胞(ADSCs)移植中对提高小鼠模型皮瓣存活率的作用.
    UNASSIGNED:在小鼠中建立腹部穿支皮瓣模型。ADSC移植后小鼠皮肤组织的组织病理学检查使用苏木精和伊红(H&E)染色进行。使用免疫组织化学(IHC)或免疫荧光(IF)染色来评估PCNA或CD31水平。使用VEGFAELISA试剂盒定量培养基中VEGFA的浓度。
    UNASSIGNED:ADSCs移植可显着缓解皮瓣组织损伤。MiR-590-3p过表达显著抑制,而miR-590-3p敲低通过调节PCNA促进皮瓣存活,VCAM-1和VEGFA水平。MiR-590-3p靶向VEGFA以调节其表达。VEGFA的敲低显著抑制,而VEGFA的过表达显著促进了皮瓣的存活。
    未经证实:ADSCs移植通过促进血管生成促进皮瓣存活。miR-590-3p/VEGFA轴调节ADSC中的皮瓣血管生成和存活。这些结果表明,干扰ADSC中的miR-590-3p可能是改善皮瓣存活的新治疗靶标。
    UNASSIGNED: Partial necrosis of skin flaps is still a substantial problem in plastic and reconstructive surgery. In this study, the role of miR-590-3p in adipose-derived stem cells (ADSCs) transplantation in improving the survival of skin flap in a mouse model was delved into.
    UNASSIGNED: An abdominal perforator flap model was established in mice. The histopathological examination of mice skin tissues after ADSCs transplantation was implemented using Hematoxylin & eosin (H&E) staining. Immunohistochemistry (IHC) or immunofluorescence (IF) staining was utilized to assess the PCNA or CD31 levels. The concentrations of VEGFA in the culture medium were quantified using a VEGFA ELISA kit.
    UNASSIGNED: The damage of tissue in the skin flap was dramatically relieved by ADSCs transplantation. MiR-590-3p overexpression notably suppressed, while miR-590-3p knockdown facilitated skin flap survival by regulating PCNA, VCAM-1, and VEGFA levels. MiR-590-3p targeted VEGFA to regulate its expression. The knockdown of VEGFA significantly inhibited, while overexpression of VEGFA notably promoted the survival of skin flap.
    UNASSIGNED: ADSCs transplantation promotes skin flap survival by boosting angiogenesis. The miR-590-3p/VEGFA axis modulates skin flap angiogenesis and survival in ADSCs. These results reveal that interfering with miR-590-3p in ADSCs could potentially be a novel therapeutic target for the improvement of skin flap survival.
    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
    未经证实:干扰素基因(STING)/TANK结合激酶1(TBK1)途径的刺激因子在介导氧化/内质网(ER)应激期间的先天免疫和炎症反应中至关重要。然而,目前尚不清楚巨噬细胞硫氧还蛋白相互作用蛋白(TXNIP)是否在氧化应激/ER应激过程中调节TBK1功能和细胞死亡途径.
    未经证实:肝缺血/再灌注损伤(IRI)小鼠模型,原代肝细胞,和骨髓来源的巨噬细胞用于骨髓特异性TXNIP敲除(TXNIPM-KO)和TXNIP熟练(TXNIPFL/FL)小鼠。
    UNASSIGNED:TXNIPM-KO小鼠对缺血/再灌注(IR)应激诱导的肝损伤具有抗性,血清丙氨酸氨基转移酶(ALT)/天冬氨酸氨基转移酶(AST)水平降低,巨噬细胞/中性粒细胞浸润,和促炎介质与TXNIPFL/FL对照相比。IR应力增加TXNIP,p-STING,缺血肝脏中p-TBK1的表达。然而,TXNIPM-KO抑制STING,TBK1,干扰素调节因子3(IRF3),和NF-κB激活与干扰素-β(IFN-β)表达。有趣的是,TXNIPM-KO增强核因子(红系衍生的2)样2(NRF2)活性,抗氧化基因表达增加,并减少IR应激肝脏中巨噬细胞活性氧(ROS)的产生和肝细胞凋亡/坏死。机械上,巨噬细胞TXNIP缺乏促进圆柱瘤病(CYLD),与NADPH氧化酶4(NOX4)共定位并相互作用,以通过去泛素化NOX4来增强NRF2活性。巨噬细胞NRF2或其靶基因2'的破坏,5'寡腺苷酸合成酶样1(OASL1)增强RasGTP酶激活蛋白结合蛋白1(G3BP1)和TBK1介导的炎症反应。值得注意的是,巨噬细胞OASL1缺乏诱导肝细胞凋亡肽酶活化因子1(APAF1),细胞色素c,和caspase-9激活,导致caspase-3引发的凋亡和受体相互作用的丝氨酸/苏氨酸蛋白激酶3(RIPK3)介导的坏死性凋亡增加。
    未经证实:巨噬细胞TXNIP缺乏增强CYLD活性并激活NRF2-OASL1信号,控制IR应激诱导的肝损伤。受NRF2调控的靶基因OASL1对于调节STING介导的TBK1激活和Apaf1/细胞色素c/caspase-9触发的凋亡/坏死细胞死亡途径至关重要。我们的发现强调了巨噬细胞TXNIP介导的CYLD-NRF2-OASL1轴在应激诱导的肝脏炎症和细胞死亡中的新作用,暗示肝脏炎症性疾病的潜在治疗靶点。
    UNASSIGNED:由缺血和再灌注引起的肝脏炎症和损伤(缺乏血液流向肝脏组织,然后再供应血液)是肝移植后肝功能障碍和肝功能衰竭的重要原因,切除,失血性休克.在这里,我们揭示了在这种情况下导致肝脏炎症和细胞死亡的潜在机制,并且可能是应激诱导的肝脏炎症损伤的治疗靶标。
    UNASSIGNED: The stimulator of interferon genes (STING)/TANK-binding kinase 1 (TBK1) pathway is vital in mediating innate immune and inflammatory responses during oxidative/endoplasmic reticulum (ER) stress. However, it remains unknown whether macrophage thioredoxin-interacting protein (TXNIP) may regulate TBK1 function and cell death pathways during oxidative/ER stress.
    UNASSIGNED: A mouse model of hepatic ischaemia/reperfusion injury (IRI), the primary hepatocytes, and bone marrow-derived macrophages were used in the myeloid-specific TXNIP knockout (TXNIPM-KO) and TXNIP-proficient (TXNIPFL/FL) mice.
    UNASSIGNED: The TXNIPM-KO mice were resistant to ischaemia/reperfusion (IR) stress-induced liver damage with reduced serum alanine aminotransferase (ALT)/aspartate aminotransferase (AST) levels, macrophage/neutrophil infiltration, and pro-inflammatory mediators compared with the TXNIPFL/FL controls. IR stress increased TXNIP, p-STING, and p-TBK1 expression in ischaemic livers. However, TXNIPM-KO inhibited STING, TBK1, interferon regulatory factor 3 (IRF3), and NF-κB activation with interferon-β (IFN-β) expression. Interestingly, TXNIPM-KO augmented nuclear factor (erythroid-derived 2)-like 2 (NRF2) activity, increased antioxidant gene expression, and reduced macrophage reactive oxygen species (ROS) production and hepatic apoptosis/necroptosis in IR-stressed livers. Mechanistically, macrophage TXNIP deficiency promoted cylindromatosis (CYLD), which colocalised and interacted with NADPH oxidase 4 (NOX4) to enhance NRF2 activity by deubiquitinating NOX4. Disruption of macrophage NRF2 or its target gene 2\',5\' oligoadenylate synthetase-like 1 (OASL1) enhanced Ras GTPase-activating protein-binding protein 1 (G3BP1) and TBK1-mediated inflammatory response. Notably, macrophage OASL1 deficiency induced hepatocyte apoptotic peptidase activating factor 1 (APAF1), cytochrome c, and caspase-9 activation, leading to increased caspase-3-initiated apoptosis and receptor-interacting serine/threonine-protein kinase 3 (RIPK3)-mediated necroptosis.
    UNASSIGNED: Macrophage TXNIP deficiency enhances CYLD activity and activates the NRF2-OASL1 signalling, controlling IR stress-induced liver injury. The target gene OASL1 regulated by NRF2 is crucial for modulating STING-mediated TBK1 activation and Apaf1/cytochrome c/caspase-9-triggered apoptotic/necroptotic cell death pathway. Our findings underscore a novel role of macrophage TXNIP-mediated CYLD-NRF2-OASL1 axis in stress-induced liver inflammation and cell death, implying the potential therapeutic targets in liver inflammatory diseases.
    UNASSIGNED: Liver inflammation and injury induced by ischaemia and reperfusion (the absence of blood flow to the liver tissue followed by the resupply of blood) is a significant cause of hepatic dysfunction and failure following liver transplantation, resection, and haemorrhagic shock. Herein, we uncover an underlying mechanism that contributes to liver inflammation and cell death in this setting and could be a therapeutic target in stress-induced liver inflammatory injury.
    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)被认为是卫生系统的一项挑战。几项研究评估了这种疾病的免疫相关方面,以确定影响COVID-19病程的宿主相关因素。microRNAs(miRNAs)作为免疫应答的有效调节因子在这方面获得了很多关注。最近的研究表明,miRNA在COVID-19中的异常表达与疾病进程有关。差异表达的miRNA已在与炎症和抗病毒免疫应答相关的途径中富集。miRNA也被认为是COVID-19的潜在治疗靶标,特别是用于管理COVID-19的病理后果。在当前的审查中,我们总结了COVID-19中miRNA失调的数据。
    Coronavirus disease 2019 (COVID-19) is regarded as a challenge in health system. Several studies have assessed the immune-related aspect of this disorder to identify the host-related factors that affect the course of COVID-19. microRNAs (miRNAs) as potent regulators of immune responses have gained much attention in this regard. Recent studies have shown aberrant expression of miRNAs in COVID-19 in association with disease course. Differentially expressed miRNAs have been enriched in pathways related with inflammation and antiviral immune response. miRNAs have also been regarded as potential therapeutic targets in COVID-19, particularly for management of pathological consequences of COVID-19. In the current review, we summarize the data about dysregulation of miRNAs in COVID-19.
    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
    寨卡病毒(ZIKV)在没有特殊治疗的情况下引起重大的人类疾病。以前我们发现了红色素B,FDA批准的食品添加剂,抑制病毒NS2B-NS3相互作用,导致细胞培养中ZIKV感染的抑制。在这项研究中,我们进行了药代动力学和体内研究,以证明在3D微型脑类器官和小鼠模型中,红色素B对ZIKV的疗效。我们的结果表明,在3D类器官模型中,红色素B在消除ZIKV复制方面非常有效。虽然药代动力学研究表明,红色素B具有低吸收曲线,用致死剂量的ZIKV攻击的小鼠在口服红血素B后显示出显着提高的存活率,与车辆控制相比。有限的结构-活性关系研究表明,大多数在黄吨环上修饰的红色素B类似物导致对病毒NS2B-NS3相互作用的抑制活性丧失或降低,蛋白酶活性和抗病毒功效。相比之下,在异苯并呋喃环上引入氯取代导致活性略有增加,这表明异苯并呋喃环对修饰的耐受性良好。细胞毒性研究表明所有衍生物对人细胞无毒。总的来说,我们的研究表明,在体外和体内,红色素B是一种有效的抗ZIKV的抗病毒药物。
    Zika virus (ZIKV) causes significant human diseases without specific therapy. Previously we found erythrosin B, an FDA-approved food additive, inhibited viral NS2B-NS3 interactions, leading to inhibition of ZIKV infection in cell culture. In this study, we performed pharmacokinetic and in vivo studies to demonstrate the efficacy of erythrosin B against ZIKV in 3D mini-brain organoid and mouse models. Our results showed that erythrosin B is very effective in abolishing ZIKV replication in the 3D organoid model. Although pharmacokinetics studies indicated that erythrosin B had a low absorption profile, mice challenged by a lethal dose of ZIKV showed a significantly improved survival rate upon oral administration of erythrosin B, compared to vehicle control. Limited structure-activity relationship studies indicated that most analogs of erythrosin B with modifications on the xanthene ring led to loss or reduction of inhibitory activities towards viral NS2B-NS3 interactions, protease activity and antiviral efficacy. In contrast, introducing chlorine substitutions on the isobenzofuran ring led to slightly increased activities, suggesting that the isobenzofuran ring is well tolerated for modifications. Cytotoxicity studies indicated that all derivatives are nontoxic to human cells. Overall, our studies demonstrated erythrosin B is an effective antiviral against ZIKV both in vitro and in vivo.
    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
    蜱传脑炎病毒(TBEV),欧亚大陆医学上最相关的蜱传播黄病毒,针对宿主中枢神经系统,并经常引起严重的脑炎。TBEV诱导的神经发病机制的严重程度是高度细胞类型特异性的,造成这种差异的确切机制尚未完全描述。因此,我们对TBEV体外感染人类原代神经元(高细胞病变效应)和星形胶质细胞(低细胞病变效应)后宿主poly-(A)/miRNA/lncRNA表达的变化进行了综合分析.严重但不轻度的TBEV菌株感染导致较高的神经元死亡率。相比之下,人星形胶质细胞中任何一种TBEV菌株的感染都没有。通过miRNA/mRNA/lncRNA/vd-sRNA网络的计算机预测进行差异表达和剪接分析,发现炎症和免疫应答途径发生了显着变化。TBEVHypr感染神经元的神经系统发育和有丝分裂调节。负责上述现象的候选机制包括通过模仿内源性miRNA的差异表达的miRNA/lncRNA或vd-sRNA对宿主mRNA水平的特异性调节和病毒驱动的宿主前mRNA剪接的调节。我们建议这些因素是在不同细胞系中观察到的两种TBEV菌株的毒力表现差异的原因。这项工作带来了人类星形胶质细胞和神经元转录组变化的第一个复杂的概述在感染过程中由两个不同毒力的TBEV菌株。所得数据可作为进一步研究TBEV-宿主相互作用机制和TBEV发病机理相关过程的起点。
    Tick-borne encephalitis virus (TBEV), the most medically relevant tick-transmitted flavivirus in Eurasia, targets the host central nervous system and frequently causes severe encephalitis. The severity of TBEV-induced neuropathogenesis is highly cell-type specific and the exact mechanism responsible for such differences has not been fully described yet. Thus, we performed a comprehensive analysis of alterations in host poly-(A)/miRNA/lncRNA expression upon TBEV infection in vitro in human primary neurons (high cytopathic effect) and astrocytes (low cytopathic effect). Infection with severe but not mild TBEV strain resulted in a high neuronal death rate. In comparison, infection with either of TBEV strains in human astrocytes did not. Differential expression and splicing analyses with an in silico prediction of miRNA/mRNA/lncRNA/vd-sRNA networks found significant changes in inflammatory and immune response pathways, nervous system development and regulation of mitosis in TBEV Hypr-infected neurons. Candidate mechanisms responsible for the aforementioned phenomena include specific regulation of host mRNA levels via differentially expressed miRNAs/lncRNAs or vd-sRNAs mimicking endogenous miRNAs and virus-driven modulation of host pre-mRNA splicing. We suggest that these factors are responsible for the observed differences in the virulence manifestation of both TBEV strains in different cell lines. This work brings the first complex overview of alterations in the transcriptome of human astrocytes and neurons during the infection by two TBEV strains of different virulence. The resulting data could serve as a starting point for further studies dealing with the mechanism of TBEV-host interactions and the related processes of TBEV pathogenesis.
    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
    SARS-CoV-2的新变种,即omicron的出现,由于其基因组中的高传播率和突变率,引起了全球关注。全世界的研究人员都在试图了解这种变异的进化和出现,以了解突变级联事件。
    我们已经考虑了直到2021年12月2日在GISAID的公共存储库中提供的所有omicron基因组(n=302基因组)以及关注变体(VOC)的代表,即,阿尔法,beta,gamma,delta,和omicron;感兴趣的变体(VOI)mu和lambda;和受监测的变体(VUM)。进行了基于全基因组的系统发育和突变分析,以了解导致omicron变体出现的SARSCoV-2的进化。
    基于全基因组的系统发育描绘了两个系统群(PG-I和PG-II)形成变体特异性进化枝,除了γ和VUGH。突变分析在omicron变体中检测到18,261个突变,其中大多数是刺突中的非同义突变(A67,T547K,D614G,H655Y,N679K,P681H,D796Y,N856K,Q954H),其次是RNA依赖性RNA聚合酶(rdrp)(A1892T,I189V,P314L,K38R,T492I,V57V),ORF6(M19M)和核衣壳卵白(RG203KR)。
    Delta和omicron已经进化成不同的系统组,并且没有共同的祖先。同时,omicron与VOIlambda有着共同的祖先,其进化主要是由非同义突变产生的。
    Emergence of new variant of SARS-CoV-2, namely omicron, has posed a global concern because of its high rate of transmissibility and mutations in its genome. Researchers worldwide are trying to understand the evolution and emergence of such variants to understand the mutational cascade events.
    We have considered all omicron genomes (n = 302 genomes) available till 2nd December 2021 in the public repository of GISAID along with representatives of variants of concern (VOC), i.e., alpha, beta, gamma, delta, and omicron; variant of interest (VOI) mu and lambda; and variant under monitoring (VUM). Whole genome-based phylogeny and mutational analysis were performed to understand the evolution of SARS CoV-2 leading to emergence of omicron variant.
    Whole genome-based phylogeny depicted two phylogroups (PG-I and PG-II) forming variant specific clades except for gamma and VUM GH. Mutational analysis detected 18,261 mutations in the omicron variant, majority of which were non-synonymous mutations in spike (A67, T547K, D614G, H655Y, N679K, P681H, D796Y, N856K, Q954H), followed by RNA dependent RNA polymerase (rdrp) (A1892T, I189V, P314L, K38R, T492I, V57V), ORF6 (M19M) and nucleocapsid protein (RG203KR).
    Delta and omicron have evolutionary diverged into distinct phylogroups and do not share a common ancestry. While, omicron shares common ancestry with VOI lambda and its evolution is mainly derived by the non-synonymous mutations.
    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
    一旦吸入,SARS-CoV-2颗粒通过与血管紧张素转换酶2(ACE2)相互作用进入呼吸道纤毛细胞。了解气道组织内ACE2的性质已成为最近的焦点,特别是考虑到COVID-19大流行。使用原代人鼻上皮细胞和分化的气液界面(ALI)模型在体外产生气道粘膜纤毛组织。使用ALI组织,鉴定了ACE2的三种不同的转录变体。一种转录物编码记录的全长ACE2蛋白。另外两个转录物是独特的截短同种型,直到最近才通过序列分析软件预测存在。定量PCR显示,所有三种转录物变体在气道粘液纤毛上皮的整个分化过程中都表达。在细胞系中外源表达的单个ACE2蛋白同种型的免疫荧光分析显示,在质膜中定位并与SARSCoV2尖峰受体结合域相互作用的能力相似。使用ACE2的N期或C期抗体对分化的ALI组织进行的免疫组织化学显示细胞中重叠和不同的信号,最值得注意的是只有ACE2C-term抗体显示质膜定位。我们还证明,与ACE2转染的细胞系相比,ALI组织中的ACE2蛋白脱落是不同的。ACE2从ALI组织的顶端和基底表面释放。一起,我们的数据强调了气道粘膜纤毛组织中ACE2转录物和蛋白质的各个方面,这些可能代表影响个体对SARS-CoV-2感染易感性的变量,或者新冠肺炎的严重程度。
    Once inhaled, SARS-CoV-2 particles enter respiratory ciliated cells by interacting with angiotensin converting enzyme 2 (ACE2). Understanding the nature of ACE2 within airway tissue has become a recent focus particularly in light of the COVID-19 pandemic. Airway mucociliary tissue was generated in-vitro using primary human nasal epithelial cells and the air-liquid interface (ALI) model of differentiation. Using ALI tissue, three distinct transcript variants of ACE2 were identified. One transcript encodes the documented full-length ACE2 protein. The other two transcripts are unique truncated isoforms, that until recently had only been predicted to exist via sequence analysis software. Quantitative PCR revealed that all three transcript variants are expressed throughout differentiation of airway mucociliary epithelia. Immunofluorescence analysis of individual ACE2 protein isoforms exogenously expressed in cell-lines revealed similar abilities to localize in the plasma membrane and interact with the SARS CoV 2 spike receptor binding domain. Immunohistochemistry on differentiated ALI tissue using antibodies to either the N-term or C-term of ACE2 revealed both overlapping and distinct signals in cells, most notably only the ACE2 C-term antibody displayed plasma-membrane localization. We also demonstrate that ACE2 protein shedding is different in ALI Tissue compared to ACE2-transfected cell lines, and that ACE2 is released from both the apical and basal surfaces of ALI tissue. Together, our data highlights various facets of ACE2 transcripts and protein in airway mucociliary tissue that may represent variables which impact an individual\'s susceptibility to SARS-CoV-2 infection, or the severity of Covid-19.
    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
    迄今为止,COVID-19大流行的爆发已经夺走了500万人的生命,是由一种名为SARS-CoV2的阳性RNA病毒引起的。缺乏针对SARS-CoV2的特异性药物,导致我们寻找有效且特定的治疗方法。小干扰RNA(siRNA)能够激活RNA干扰(RNAi)通路,沉默特定的靶基因,抑制病毒复制,作为SARS-CoV2抗病毒剂还没有引起足够的重视。在全面完成之前,它可能是对抗这种流行病的潜在武器,有效的大规模疫苗接种。对于这项研究,使用基于网络的生物信息学工具(siDirect2.0)针对14个靶序列设计特异性siRNA。这些可能具有沉默SARS-CoV2的必需蛋白的高概率。例如:3CLpro/Mpro/nsp5,nsp7,Rd-Rp/nsp12,ZD,NTPase/HEL或nsp13,PLpro/nsp3,包膜蛋白(E),刺突糖蛋白(S),核衣壳磷蛋白(N),膜糖蛋白(M),ORF8,ORF3a,nsp2及其各自的5'和3'-UTR。在这些潜在的药物靶点中,它们中的大多数包含高度保守的序列;其余的是根据它们在病毒复制和存活中的作用来选择的。使用SARS-CoV2蛋白的传统疫苗开发技术需要6-8个月;同时,该病毒在用于疫苗开发的候选蛋白中经历了几次突变。当基于蛋白质的疫苗进入市场时,病毒会经历几次突变,这样,针对病毒序列的抗体可能不能有效地限制新突变的病毒。然而,siRNA技术可以根据实时病毒突变状态做出序列。这有可能抑制SARS-CoV2病毒复制,通过RNAi技术。
    The outbreak of the COVID-19 pandemic has cost five million lives to date, and was caused by a positive-sense RNA virus named SARS-CoV2. The lack of drugs specific to SARS-CoV2, leads us to search for an effective and specific therapeutic approach. Small interfering RNA (siRNA) is able to activate the RNA interference (RNAi) pathway to silence the specific targeted gene and inhibit the viral replication, and it has not yet attracted enough attention as a SARS-CoV2 antiviral agent. It could be a potential weapon to combat this pandemic until the completion of full scale, effective mass vaccination. For this study, specific siRNAs were designed using a web-based bioinformatics tool (siDirect2.0) against 14 target sequences. These might have a high probability of silencing the essential proteins of SARS-CoV2. such as: 3CLpro/Mpro/nsp5, nsp7, Rd-Rp/nsp12, ZD, NTPase/HEL or nsp13, PLpro/nsp3, envelope protein (E), spike glycoprotein (S), nucleocapsid phosphoprotein (N), membrane glycoprotein (M), ORF8, ORF3a, nsp2, and its respective 5\' and 3\'-UTR. Among these potential drug targets, the majority of them contain highly conserved sequences; the rest are chosen on the basis of their role in viral replication and survival. The traditional vaccine development technology using SARS-CoV2 protein takes 6-8 months; meanwhile the virus undergoes several mutations in the candidate protein chosen for vaccine development. By the time the protein-based vaccine reaches the market, the virus would have undergone several mutations, such that the antibodies against the viral sequence may not be effective in restricting the newly mutated viruses. However, siRNA technology can make sequences based on real time viral mutation status. This has the potential for suppressing SARS-CoV2 viral replication, through RNAi technology.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号