Murine hepatitis virus

鼠肝炎病毒
  • 文章类型: Journal Article
    由病原病毒引起的流行病对全球公共卫生构成严重威胁。电磁波是一种非接触和非电离辐射技术,已成为灭活细菌病原体的有效工具。在这项研究中,我们使用9.375GHz的电磁波研究了电磁波对致病性人类冠状病毒替代病毒MHV-A59的灭活效果和机制,并评估不同表面材料的失活效率。我们表明,9.375GHz电磁波通过破坏病毒颗粒使MHV-A59失活,信封或基因组。我们还发现,9.375GHz电磁波可以降低病毒在无生命材料如塑料表面的感染性,玻璃,布,和木头。总之,我们的结果表明,9.375GHz电磁波是一种有前途的消毒技术,可以防止病原病毒的传播和感染。
    Epidemics caused by pathogenic viruses are a severe threat to public health worldwide. Electromagnetic waves are a type of noncontact and nonionizing radiation technology that has emerged as an effective tool for inactivating bacterial pathogens. In this study, we used a 9.375 GHz electromagnetic wave to study the inactivation effect and mechanism of electromagnetic waves on MHV-A59, a substitute virus for pathogenic human coronavirus, and to evaluate the inactivation efficiency on different surface materials. We showed that 9.375 GHz electromagnetic waves inactivate MHV-A59 by destroying viral particles, envelopes, or genomes. We also found that 9.375 GHz electromagnetic waves can decrease the infectivity of viruses on the surface of inanimate materials such as plastic, glass, cloth, and wood. In conclusion, our results suggested that the 9.375 GHz electromagnetic wave is a promising disinfection technique for preventing the spread and infection of pathogenic viruses.
    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
    小鼠肝炎病毒(MHV)感染是实验室中最常见的小鼠感染类型之一。MHV可能导致小鼠死亡,甚至干扰动物实验的结果。在这里,我们开发了两种基于多酶等温快速扩增(MIRA)的等温方法,用于保守M基因中MHV的快速检测。我们设计并筛选了几对引物和探针,并将等温荧光检测器用于外切核酸酶Ⅲ逆转录MIRA(exo-RT-MIRA)测定。为了进一步简化工作流程,便携式荧光可视化仪器,也作为一个手掌大小的手持系统,用于裸眼exo-RT-MIRA测定。优化扩增温度和时间。对于exo-RT-MIRA和裸眼exo-RT-MIRA测定,该测定可以在42°C下处理20分钟。外-RT-MIRA测定的检测限(LoD)为43.4拷贝/μL。裸眼exo-RT-MIRA测定的LoD为68.2拷贝/μL。在两个测定中未观察到非特异性扩增。通过qPCR检查了总共107个样本,并开发了两个测定。实验结果统计学分析表明,具有qPCR的exo-RT-MIRA测定产生与1.000的卡伯值足够一致(p<0.0001)。结果也表现出良好的一致性(卡伯值,0.961)(p<0.0001)在裸眼外切RT-MIRA测定和qPCR测定之间。在我们的研究中,exo-RT-MIRA测定和裸眼exo-RT-MIRA测定为MHV点诊断提供了新方法的可能性。
    Murine hepatitis virus (MHV) infection is one of the most prevalent types of mice infection in laboratory. MHV could cause death in mice and even interfere with the results in animal experiments. Herein, we developed two isothermal approaches based on the Multienzyme Isothermal Rapid Amplification (MIRA), for rapid detection of MHV in conserved M gene. We designed and screened several pairs of primers and probes and the isothermal fluorescence detector was applied for the exonuclease Ⅲ reverse transcription MIRA (exo-RT-MIRA) assay. To further simplify the workflow, the portable fluorescence visualization instrument, also as a palm-sized handheld system, was used for the naked-eye exo-RT-MIRA assay. The amplification temperature and time were optimized. The assay could be processed well at 42 °C 20 min for the exo-RT-MIRA and the naked-eye exo-RT-MIRA assay. The limit of detection (LoD) of the exo-RT-MIRA assay was 43.4 copies/μL. The LoD of the naked-eye exo-RT-MIRA assay was 68.2 copies/μL. No nonspecific amplifications were observed in the two assays. A total of 107 specimens were examined by qPCR and two assays developed. The experimental results statistical analysis demonstrated that the exo-RT-MIRA assay with the qPCR yielded sufficient agreement with a kappa value of 1.000 (p < 0.0001). The results also exhibited a good agreement (kappa value, 0.961) (p < 0.0001) between the naked-eye exo-RT-MIRA assay and the qPCR assay. In our study, the exo-RT-MIRA assay and the naked-eye exo-RT-MIRA assay presented the possibility of new methods in MHV point-of-testing diagnosis.
    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
    已提出冷链条件以促进SARS-CoV-2的远距离传播,但尚不清楚该病毒在冷链包装材料上的可行性。
    本研究以鼠肝炎病毒MHV-JHM株作为模型生物,研究SARS-CoV-2在泡沫上的生存力,塑料,纸板,和不同温度下的木板(-40°C,-20°C,和4°C)。此外,还评估了过乙酸和次氯酸钠消除塑料和纸板上的MHV-JHM的能力。
    结果表明MHV-JHM可以在泡沫上存活,塑料,或纸板在-40°C和-20°C下最多28天,在4°C下在泡沫和塑料表面上长达14天。尽管病毒核酸在4°C储存28天后仍可检测到,相应的病毒滴度低于定量限(LOQ).
    该研究强调,核酸检测阳性结果可能不表明病毒仍然存活,并证实过乙酸和次氯酸钠能有效消除冷链条件下包装材料上的MHV-JHM。
    The cold chain conditions have been suggested to facilitate long-distance transmission of SARS-CoV-2, but it is unclear how viable the virus is on cold chain packaging materials.
    This study used the MHV-JHM strain of murine hepatitis virus as a model organism to investigate the viability of SARS-CoV-2 on foam, plastic, cardboard, and wood sheets at different temperatures (-40°C, -20°C, and 4°C). In addition, the ability of peracetic acid and sodium hypochlorite to eliminate the MHV-JHM on plastic and cardboard sheets were also evaluated.
    The results indicate that MHV-JHM can survive on foam, plastic, or cardboard sheets for up to 28 days at -40°C and -20°C, and up to 14 days on foam and plastic surfaces at 4°C. Although viral nucleic acids were still detectable after storing at 4°C for 28 days, the corresponding virus titer was below the limit of quantification (LOQ).
    The study highlights that a positive nucleic acid test result may not indicate that the virus is still viable, and confirms that peracetic acid and sodium hypochlorite can effectively eliminate MHV-JHM on packaging materials under cold chain conditions.
    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
    目标:冠状病毒,像其他正义RNA病毒一样,可以重塑宿主膜以形成双膜囊泡(DMV)作为其复制细胞器。目前,参与DMV形成的宿主因素尚未明确。在这项研究中,我们使用传染性胃肠炎病毒(TGEV)作为病毒模型,研究双特异性酪氨酸磷酸化调节激酶1A(DYRK1A)对冠状病毒的调节机制.结果显示DYRK1A以不依赖激酶的方式显著抑制TGEV复制。DYRK1A基因敲除(KO)可通过调节受体氨肽酶N(ANPEP)和内吞相关基因的表达来抑制病毒进入。更重要的是,我们的结果表明,DYRK1AKO显着抑制DMV的形成以调节病毒的复制。进一步的数据证明,DYRK1A在小鼠肝炎病毒的复制中也是必不可少的,猪三角洲冠状病毒,和猪沙普病毒。一起来看,我们的发现表明,DYRK1A是一个保守的因子为正义性RNA病毒,并提供了新的见解,其转录调控活性,揭示了其作为治疗设计候选靶标的潜力。
    Coronaviruses (CoVs) pose a major threat to human and animal health worldwide, which complete viral replication by hijacking host factors. Identifying host factors essential for the viral life cycle can deepen our understanding of the mechanisms of virus-host interactions. Based on our previous genome-wide CRISPR screen of α-CoV transmissible gastroenteritis virus (TGEV), we identified the host factor dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A), but not DYRK1B, as a critical factor in TGEV replication. Rescue assays and kinase inhibitor experiments revealed that the effect of DYRK1A on viral replication is independent of its kinase activity. Nuclear localization signal modification experiments showed that nuclear DYRK1A facilitated virus replication. Furthermore, DYRK1A knockout significantly downregulated the expression of the TGEV receptor aminopeptidase N (ANPEP) and inhibited viral entry. Notably, we also demonstrated that DYRK1A is essential for the early stage of TGEV replication. Transmission electron microscopy results indicated that DYRK1A contributes to the formation of double-membrane vesicles in a kinase-independent manner. Finally, we validated that DYRK1A is also a proviral factor for mouse hepatitis virus, porcine deltacoronavirus, and porcine sapelovirus. In conclusion, our work demonstrated that DYRK1A is an essential host factor for the replication of multiple viruses, providing new insights into the mechanism of virus-host interactions and facilitating the development of new broad-spectrum antiviral drugs.IMPORTANCECoronaviruses, like other positive-sense RNA viruses, can remodel the host membrane to form double-membrane vesicles (DMVs) as their replication organelles. Currently, host factors involved in DMV formation are not well defined. In this study, we used transmissible gastroenteritis virus (TGEV) as a virus model to investigate the regulatory mechanism of dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) on coronavirus. Results showed that DYRK1A significantly inhibited TGEV replication in a kinase-independent manner. DYRK1A knockout (KO) can regulate the expression of receptor aminopeptidase N (ANPEP) and endocytic-related genes to inhibit virus entry. More importantly, our results revealed that DYRK1A KO notably inhibited the formation of DMV to regulate the virus replication. Further data proved that DYRK1A is also essential in the replication of mouse hepatitis virus, porcine deltacoronavirus, and porcine sapelovirus. Taken together, our findings demonstrated that DYRK1A is a conserved factor for positive-sense RNA viruses and provided new insights into its transcriptional regulation activity, revealing its potential as a candidate target for therapeutic design.
    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:先前的研究表明,自然杀伤(NK)细胞从外周器官迁移到肝脏中,并在病毒诱导的肝衰竭中对肝细胞产生细胞毒性作用。
    目的:本研究旨在探讨趋化因子受体在小鼠肝炎病毒3株(MHV-3)诱导的暴发性肝衰竭(MHV-3-FHF)模型中NK细胞迁移中的潜在治疗作用及其机制。
    结果:通过基因阵列分析,发现MHV-3感染后,趋化因子(C-C基序)受体5(CCR5)在肝NK细胞中的表达水平显着升高。在MHV-3感染后48h,肝脏CCR5+常规NK(cNK)细胞数量增加并达到峰值,而肝脏常驻NK(rNK)细胞的数量稳步下降。此外,CCR5相关趋化因子的表达,包括巨噬细胞炎性蛋白(MIP)-1α,MIP-1β并在激活时受到调节,正常T细胞表达和分泌(RANTES)在MHV-3感染的肝细胞中显著上调。在体外Transwell迁移试验中,CCR5阻断的脾cNK细胞显示向MHV-3感染的肝细胞迁移减少,抑制MIP-1β或RANTES而不是MIP-1α会降低cNK细胞的迁移。此外,CCR5基因敲除(KO)小鼠显示MHV-3感染后肝脏cNK细胞浸润减少,伴随着减轻肝损伤和改善小鼠生存时间。将野生型小鼠的cNK细胞过继转移到CCR5KO小鼠中,导致肝脏cNK细胞大量积累并加重肝损伤。此外,在MHV-3-FHF模型中,maraviroc对CCR5的药理学抑制减少了肝脏中cNK细胞的浸润和肝损伤。
    结论:在MHV-3诱导的肝损伤期间,CCR5-MIP-1β/RANTES轴在cNK细胞募集到肝脏中起关键作用。CCR5的靶向抑制提供了在病毒诱导的急性肝损伤期间改善肝损伤的治疗方法。
    Previous studies have demonstrated that natural killer (NK) cells migrated into the liver from peripheral organs and exerted cytotoxic effects on hepatocytes in virus-induced liver failure.
    This study aimed to investigate the potential therapeutic role of chemokine receptors in the migration of NK cells in a murine hepatitis  virus strain 3 (MHV-3)-induced fulminant hepatic failure (MHV-3-FHF) model and its mechanism.
    By gene array analysis, chemokine (C-C motif) receptor 5 (CCR5) was found to have remarkably elevated expression levels in hepatic NK cells after MHV-3 infection. The number of hepatic CCR5+ conventional NK (cNK) cells increased and peaked at 48 h after MHV-3 infection, while the number of hepatic resident NK (rNK) cells steadily declined. Moreover, the expression of CCR5-related chemokines, including macrophage inflammatory protein (MIP)-1α, MIP-1β and regulated on activation, normal T-cell expressed and secreted (RANTES) was significantly upregulated in MHV-3-infected hepatocytes. In an in vitro Transwell migration assay, CCR5-blocked splenic cNK cells showed decreased migration towards MHV-3-infected hepatocytes, and inhibition of MIP-1β or RANTES but not MIP-1α decreased cNK cell migration. Moreover, CCR5 knockout (KO) mice displayed reduced infiltration of hepatic cNK cells after MHV-3 infection, accompanied by attenuated liver injury and improved mouse survival time. Adoptive transfer of cNK cells from wild-type mice into CCR5 KO mice resulted in the abundant accumulation of hepatic cNK cells and aggravated liver injury. Moreover, pharmacological inhibition of CCR5 by maraviroc reduced cNK cell infiltration in the liver and liver injury in the MHV-3-FHF model.
    The CCR5-MIP-1β/RANTES axis played a critical role in the recruitment of cNK cells to the liver during MHV-3-induced liver injury. Targeted inhibition of CCR5 provides a therapeutic approach to ameliorate liver damage during virus-induced acute liver 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: Comparative Study
    目标:贝塔克隆病毒,包括严重急性呼吸道综合征冠状病毒-2(SARS-CoV-2)和小鼠肝炎病毒(MHV),利用溶酶体胞吐途径进行出口。然而,目前尚不清楚是否所有β病毒成员都使用相同的途径退出细胞.这里,我们证明了猪血凝性脑脊髓炎病毒(PHEV)通过Arl8b依赖性溶酶体胞吐作用发生,SARS-CoV-2和MHV共有的细胞出口机制。值得注意的是,PHEV酸化溶酶体并激活溶酶体降解酶,SARS-CoV-2和MHV使溶酶体脱酸并限制溶酶体降解酶的激活。此外,PHEV释放取决于V-ATP酶介导的溶酶体pH。此外,这是第一项使用溶酶体评估βCoV在体内传播的研究,我们发现溶酶体在PHEV神经传递和中枢神经系统病毒感染引起的脑损伤中起着关键作用。一起来看,不同的β冠状病毒可以不同地破坏溶酶体功能以退出细胞。
    OBJECTIVE: Betacoronaviruses, including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and mouse hepatitis virus (MHV), exploit the lysosomal exocytosis pathway for egress. However, whether all betacoronaviruses members use the same pathway to exit cells remains unknown. Here, we demonstrated that porcine hemagglutinating encephalomyelitis virus (PHEV) egress occurs by Arl8b-dependent lysosomal exocytosis, a cellular egress mechanism shared by SARS-CoV-2 and MHV. Notably, PHEV acidifies lysosomes and activates lysosomal degradative enzymes, while SARS-CoV-2 and MHV deacidify lysosomes and limit the activation of lysosomal degradative enzymes. In addition, PHEV release depends on V-ATPase-mediated lysosomal pH. Furthermore, this is the first study to evaluate βCoV using lysosome for spreading through the body, and we have found that lysosome played a critical role in PHEV neural transmission and brain damage caused by virus infection in the central nervous system. Taken together, different betacoronaviruses could disrupt lysosomal function differently to exit 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: Letter
    暂无摘要。
    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
    鼠肝炎病毒(MHV)是一种高度传染性的鼠冠状病毒,具有对宿主动物造成伤害的高潜力。本研究旨在建立实时逆转录重组酶聚合酶扩增(RT-RPA)快速检测实验小鼠MHV的方法。
    UNASSIGNED:设计了针对MHV参考菌株M基因中保守区域的RT-RPA测定的特异性引物和探针(登录号:FJ6647223)根据TwistDx手册说明。特异性,灵敏度,评估RT-RPA方法的可重复性,并与标准RT-qPCR方法进行比较。使用68个现场样品评估该测定的临床适用性。
    UNASSIGNED:使用新开发的RT-RPA测定法在37°C下在20分钟内完成扩增,而使用RT-qPCR方法需要近60分钟。RT-RPA方法具有明显的省时优势。RT-RPA和RT-PCR方法的检测限相同,为4.45×101拷贝/μL。特异性表现为缺乏与MHV的交叉反应,小鼠肺炎病毒,仙台病毒,汉坦病毒,微小的小鼠病毒,和III型呼肠孤病毒。RT-RPA检测的MHV检出率为13.63%(9/66),RT-qPCR检测为15.15%(10/66)。Cohen的“kappa”(κ)分析结果在两种方法之间表现出非常好的一致性,κ≥0.750(因为κ=0.939)和p<0.0005(因为p=0.000)。
    UASSIGNED:RT-RPA检测提供了一种简单的替代工具,快速,在实验室小鼠中可靠地检测MHV,具有在实验室中应用的巨大潜力。
    Murine hepatitis virus (MHV) is a highly infectious murine coronavirus that has a high potential for causing harm to host animals. This study aimed to develop a real-time reverse transcription recombinase polymerase amplification (RT-RPA) method for rapid detection of MHV in laboratory mice.
    UNASSIGNED: Specific primers and probes for RT-RPA assay were designed targeting the conserved region in the M gene of the MHV reference strain (accession no. FJ6647223) according to the TwistDx manual instructions. The specificity, sensitivity, and reproducibility of the RT-RPA method were evaluated and compared with those of the standard RT-qPCR method. The clinical applicability of this assay was evaluated using 68 field samples.
    UNASSIGNED: Amplification using the newly developed RT-RPA assay was completed within 20 min at 37°C, while that using the RT-qPCR method required nearly 60 min. The RT-RPA method exhibited an obvious time-saving advantage. Both RT-RPA and RT-PCR methods had the same limit of detection, which was 4.45 × 101 copies/μL. The specificity was indicated by a lack of cross-reaction with MHV, pneumonia virus of mice, Sendai virus, hantavirus, minute virus of mice, and reovirus type III. The MHV detection rate of RT-RPA assays was 13.63% (9/66) and RT-qPCR assays was 15.15% (10/66). Cohen\'s \"kappa\" (κ) analysis results exhibited a very good agreement between two methods with the value of κ ≥ 0.750(since κ = 0.939) and p < 0.0005 (since p = 0.000).
    UNASSIGNED: The RT-RPA assay offers an alternative tool for simple, rapid, and reliable detection of MHV in laboratory mice and has significant potential for application in laboratories.
    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
    病毒后细菌感染是冠状病毒感染的主要医疗保健挑战,包括COVID-19;然而,宿主对继发感染的易感性增加的冠状病毒特异性机制仍然未知.在人类中,冠状病毒,包括SARS-CoV-2,感染肺部免疫细胞,包括肺泡巨噬细胞,在SARS-CoV-2的小鼠模型中复制不良的表型。为了克服这一点,我们使用同时感染免疫细胞和结构细胞的天然鼠β-冠状病毒小鼠模型来研究冠状病毒增强对细菌感染的易感性.我们的数据表明,冠状病毒感染会损害宿主清除入侵的细菌病原体的能力,并增强小鼠的肺组织损伤。机械上,冠状病毒通过损害溶酶体酸化和与吞噬的细菌融合来限制巨噬细胞的细菌杀伤能力。此外,冠状病毒诱导的溶酶体功能障碍促进细胞凋亡和IL-1β的释放。抑制组织蛋白酶B可减少冠状病毒细菌感染后小鼠的细胞死亡和IL-1β释放,并促进细菌清除。
    Postviral bacterial infections are a major health care challenge in coronavirus infections, including COVID-19; however, the coronavirus-specific mechanisms of increased host susceptibility to secondary infections remain unknown. In humans, coronaviruses, including SARS-CoV-2, infect lung immune cells, including alveolar macrophages, a phenotype poorly replicated in mouse models of SARS-CoV-2. To overcome this, we used a mouse model of native murine β-coronavirus that infects both immune and structural cells to investigate coronavirus-enhanced susceptibility to bacterial infections. Our data show that coronavirus infection impairs the host ability to clear invading bacterial pathogens and potentiates lung tissue damage in mice. Mechanistically, coronavirus limits the bacterial killing ability of macrophages by impairing lysosomal acidification and fusion with engulfed bacteria. In addition, coronavirus-induced lysosomal dysfunction promotes pyroptotic cell death and the release of IL-1β. Inhibition of cathepsin B decreased cell death and IL-1β release and promoted bacterial clearance in mice with postcoronavirus bacterial infection.
    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
    重型病毒性肝炎(FVH)是一种威胁生命的疾病,但其发病机制尚不完全清楚。中性粒细胞胞外诱捕网(NETs)是炎症和凝血之间未被识别的联系,这是FVH的2个主要特征。这里,我们研究了NETs在FVH发病机制中的作用和机制。
    通过鼠肝炎病毒株-3感染建立FVH小鼠模型。感染或未感染小鼠的肝白细胞用于单细胞RNA测序和全转录组测序。使用DNase1实现NET消耗。对乙酰氨基酚用于建立非病毒引起的急性肝衰竭小鼠模型。临床上,肝脏中与NETs相关的标志物,等离子体,在乙型肝炎病毒(HBV)相关的急性肝损伤患者中评估外周血中性粒细胞。
    在鼠肝炎病毒株3感染的小鼠中观察到肝NETs形成增加,但在对乙酰氨基酚治疗的小鼠中没有。NETs耗竭通过抑制肝纤维蛋白沉积和炎症改善FVH的肝损伤和存活率。过继转移实验表明,中性粒细胞特异性纤维蛋白原样蛋白2(FGL2)促进了NETs的形成。发现FGL2直接与粘磷脂3相互作用,粘磷脂3调节钙内流并启动自噬,导致网络形成。临床上,HBV急性肝损伤患者血浆NETs水平升高与凝血功能障碍相关。FGL2、NET、在这些患者中观察到肝脏中的纤维蛋白。
    NETs通过促进纤维蛋白沉积和炎症加重FVH的肝损伤。NETs的形成受FGL2-粘磷脂3-自噬轴的调节。靶向NETs可能为FVH的治疗提供新的策略。
    Fulminant viral hepatitis (FVH) is a life-threatening disease, but its pathogenesis is not fully understood. Neutrophil extracellular traps (NETs) were an unrecognized link between inflammation and coagulation, which are 2 main features of FVH. Here, we investigated the role and mechanism of NETs in the pathogenesis of FVH.
    A mouse model of FVH was established by murine hepatitis virus strain-3 infection. Liver leukocytes of infected or uninfected mice were used for single-cell RNA sequencing and whole-transcriptome sequencing. NETs depletion was achieved using DNase 1. Acetaminophen was used to establish a mouse model of non-virus-caused acute liver failure. Clinically, NETs-related markers in liver, plasma, and peripheral neutrophils were assessed in patients with hepatitis B virus (HBV)-related acute liver injury.
    Increased hepatic NETs formation was observed in murine hepatitis virus strain-3-infected mice, but not in acetaminophen-treated mice. NETs depletion improved the liver damage and survival rate in FVH by inhibiting hepatic fibrin deposition and inflammation. An adoptive transfer experiment showed that neutrophil-specific fibrinogen-like protein 2 (FGL2) promoted NETs formation. FGL2 was found to directly interact with mucolipin 3, which regulated calcium influx and initiated autophagy, leading to NETs formation. Clinically, increased plasma NETs level was associated with coagulation dysfunction in patients with HBV acute liver injury. Colocalization of FGL2, NETs, and fibrin in liver was observed in these patients.
    NETs aggravated liver injury in FVH by promoting fibrin deposition and inflammation. NETs formation was regulated by the FGL2-mucolipin 3-autophagy axis. Targeting NETs may provide a new strategy for the treatment of FVH.
    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