antivirals

抗病毒药物
  • 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

    求助全文

  • 文章类型: Journal Article
    背景:急性COVID期间的治疗是否能对长期COVID发病率产生保护作用尚不清楚。
    目的:评估抗病毒药物急性COVID治疗之间的关系,皮质类固醇,单克隆抗体(mAb)和长期COVID发病率,以及它们对不同人群和个体症状的影响。
    方法:直到2024年1月29日在PubMed进行了搜索,Medline,WebofScience,和Embase。
    方法:报道急性COVID后COVID长期发病率的文章,随访至少30天,无语言限制。
    方法:有COVID-19诊断史的患者。
    方法:接受抗病毒药物治疗的患者,皮质类固醇或单克隆抗体。
    质量评估基于纽卡斯尔-渥太华量表,ROBINS-I和Cochrane偏差工具的风险。
    记录每个研究的基本特征。随机森林模型和元回归用于评估治疗与长期COVID之间的相关性。
    结果:我们的搜索确定了2363条记录,其中32项纳入定性综合,25项纳入荟萃分析。来自14篇研究急性COVID抗病毒治疗的论文的效果大小得出结论,其对长期COVID的保护功效(OR0.61,95%CI:0.48-0.79,p=0.0002);然而,皮质类固醇(OR1.57,95%CI:0.80-3.09,p=0.1913)和mAb治疗(OR0.94,95%CI:0.56-1.56,p=0.8012)未产生这种效果.随后的亚组分析显示,抗病毒药物在老年人中提供了更强的保护,男性,未接种疫苗和非糖尿病人群。此外,抗病毒药物有效地减少了22例分析的长期COVID症状中的8例。
    结论:我们的荟萃分析确定,抗病毒药物降低了人群的长期covid发病率,因此应推荐用于急性COVID治疗。单克隆抗体治疗与长期COVID之间没有关系,但应进行研究以阐明急性COVID皮质类固醇对COVID急性期的潜在有害影响。
    BACKGROUND: Whether treatment during acute COVID-19 results in protective efficacy against long COVID incidence remains unclear.
    OBJECTIVE: To assess the relationship between acute COVID-19 treatments of antivirals, corticosteroids, and monoclonal antibodies (mAbs) and long COVID incidence, and their effects in different populations and individual symptoms.
    METHODS: A systematic review and meta-analysis.
    METHODS: Searches were conducted up to January 29, 2024 in PubMed, Medline, Web of Science, and Embase.
    METHODS: Articles that reported long COVID incidence post-acute COVID with a follow-up of at least 30 days with no language restrictions.
    METHODS: Patients with a COVID-19 diagnosis history.
    METHODS: Patients treated with antivirals, corticosteroids or mAbs.
    UNASSIGNED: Quality assessment was based on the Newcastle-Ottawa scale, risk of bias in nonrandomized studies of interventions-I and Cochrane risk of bias tool.
    UNASSIGNED: Basic characteristics were documented for each study. Random forest model and meta-regression were used to evaluate the correlation between treatments and long COVID.
    RESULTS: Our search identified 2363 records, 32 of which were included in the qualitative synthesis and 25 included into the meta-analysis. Effect size from 14 papers investigating acute COVID-19 antiviral treatment concluded its protective efficacy against long COVID (OR, 0.61; 95% CI, 0.48-0.79; p 0.0002); however, corticosteroid (OR, 1.57; 95% CI, 0.80-3.09; p 0.1913), and mAbs treatments (OR, 0.94; 95% CI, 0.56-1.56; p 0.8012) did not generate such effect. Subsequent subgroup analysis revealed that antivirals provided stronger protection in the aged, male, unvaccinated and nondiabetic populations. Furthermore, antivirals effectively reduced 8 out of the 22 analysed long COVID symptoms.
    CONCLUSIONS: Our meta-analysis determined that antivirals reduced long COVID incidence across populations and should thus be recommended for acute COVID-19 treatment. There was no relationship between mAbs treatment and long COVID, but studies should be conducted to clarify acute COVID-19 corticosteroids\' potential harmful effects on the post-acute phase of COVID-19.
    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
    寨卡病毒(ZIKV)是蚊媒黄病毒之一,对神经系统表现出独特的嗜性,并与格林-巴利综合征和先天性寨卡综合征(CZS)有关。登革热病毒(DENV)和黄热病病毒(YFV),另外两种蚊媒黄病毒,也已经传播了很长时间并导致严重的疾病,如登革热出血热和黄热病,分别。然而,目前还没有安全有效的抗病毒药物被批准用于治疗这些黄病毒的感染或合并感染。这里,我们发现扎鲁司特,一种怀孕安全的白三烯受体拮抗剂,在不同细胞系中对来自不同谱系的ZIKV菌株的感染表现出有效的抗病毒活性,以及针对DENV-2和YFV17D的感染。机制研究表明,扎鲁司特通过破坏病毒体的完整性,直接和不可逆地灭活这些黄病毒,导致病毒感染性的丧失,从而抑制病毒感染的进入步骤。考虑到它对黄病毒的功效,它对孕妇的安全性,以及它的神经保护作用,扎鲁司特是预防和治疗ZIKV感染或合并感染的有前途的候选药物,DENV,YFV,即使是孕妇。
    Zika virus (ZIKV) is one of the mosquito-borne flaviviruses that exhibits a unique tropism to nervous systems and is associated with Guillain-Barre syndrome and congenital Zika syndrome (CZS). Dengue virus (DENV) and yellow fever virus (YFV), the other two mosquito-borne flaviviruses, have also been circulating for a long time and cause severe diseases, such as dengue hemorrhagic fever and yellow fever, respectively. However, there are no safe and effective antiviral drugs approved for the treatment of infections or coinfections of these flaviviruses. Here, we found that zafirlukast, a pregnancy-safe leukotriene receptor antagonist, exhibited potent antiviral activity against infections of ZIKV strains from different lineages in different cell lines, as well as against infections of DENV-2 and YFV 17D. Mechanistic studies demonstrated that zafirlukast directly and irreversibly inactivated these flaviviruses by disrupting the integrity of the virions, leading to the loss of viral infectivity, hence inhibiting the entry step of virus infection. Considering its efficacy against flaviviruses, its safety for pregnant women, and its neuroprotective effect, zafirlukast is a promising candidate for prophylaxis and treatment of infections or coinfections of ZIKV, DENV, and YFV, even in pregnant women.
    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
    猪繁殖与呼吸综合征(PRRS)由PRRS病毒(PRRSV)引起,由于缺乏有效的商业疫苗和药物,给全球养猪业造成了巨大的经济损失。迫切需要制定PRRS预防和控制的替代战略,如抗病毒药物。在这项研究中,我们确定了ursonicacid(UNA),一种来自草药的天然五环三萜类化合物,作为一种具有体外抗PRRSV活性的新药。机械上,添加时间测定显示,UNA在之前添加时抑制PRRSV复制,同时,诱导PRRSV感染后。化合物靶标预测和分子对接分析表明UNA与PTPN1的活性口袋相互作用,这进一步通过靶标蛋白干扰测定和磷酸酶活性测定得到证实。此外,UNA通过靶向抑制IFN-β产生的PTPN1来抑制PRRSV复制。此外,UNA在体外表现出对猪流行性腹泻病毒(PEDV)和Seneca病毒A(SVA)复制的抗病毒活性。这些发现将有助于开发针对PRRS和其他猪病毒感染的新型预防和治疗剂。
    Porcine reproductive and respiratory syndrome (PRRS), caused by the PRRS virus (PRRSV), has caused substantial economic losses to the global swine industry due to the lack of effective commercial vaccines and drugs. There is an urgent need to develop alternative strategies for PRRS prevention and control, such as antiviral drugs. In this study, we identified ursonic acid (UNA), a natural pentacyclic triterpenoid from medicinal herbs, as a novel drug with anti-PRRSV activity in vitro. Mechanistically, a time-of-addition assay revealed that UNA inhibited PRRSV replication when it was added before, at the same time as, and after PRRSV infection was induced. Compound target prediction and molecular docking analysis suggested that UNA interacts with the active pocket of PTPN1, which was further confirmed by a target protein interference assay and phosphatase activity assay. Furthermore, UNA inhibited PRRSV replication by targeting PTPN1, which inhibited IFN-β production. In addition, UNA displayed antiviral activity against porcine epidemic diarrhoea virus (PEDV) and Seneca virus A (SVA) replication in vitro. These findings will be helpful for developing novel prophylactic and therapeutic agents against PRRS and other swine virus infections.
    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
    由于过量消费而导致地表水中抗病毒药物的增加对水生生物构成了严重威胁。我们最近的研究发现,在藻类水华中输入抗病毒药物阿比多可对铜绿微囊藻的生长和代谢产生急性毒性,导致生长抑制,以及叶绿素和ATP含量的降低。然而,所涉及的毒性机制仍然不清楚,在这项研究中通过转录组学分析进一步研究。结果表明,暴露于0.01-10.0mg/L的阿比妥后,藻类中的885-1248个基因差异表达。大多数被下调。对常见下调基因的分析发现,细胞对氧化应激的反应和受损的DNA结合受到影响,暗示藻类的应激防御系统和DNA修复功能可能受到损害。卟啉代谢基因的下调,光合作用,碳固定,糖酵解,三羧酸循环,氧化磷酸化可能会抑制叶绿素合成,光合作用,和ATP供应,从而阻碍藻类的生长和代谢。此外,与核苷酸代谢和DNA复制相关的基因的下调可能会影响藻类的繁殖。这些发现为阐明藻类水华水中污染物对藻类的毒性机制提供了有效的策略。
    Increasing antivirals in surface water caused by their excessive consumption pose serious threats to aquatic organisms. Our recent research found that the input of antiviral drug arbidol to algal bloom water can induce acute toxicity to the growth and metabolism of Microcystis aeruginosa, resulting in growth inhibition, as well as decrease in chlorophyll and ATP contents. However, the toxic mechanisms involved remained obscure, which were further investigated through transcriptomic analysis in this study. The results indicated that 885-1248 genes in algae were differentially expressed after exposure to 0.01-10.0 mg/L of arbidol, with the majority being down-regulated. Analysis of commonly down-regulated genes found that the cellular response to oxidative stress and damaged DNA bonding were affected, implying that the stress defense system and DNA repair function of algae might be damaged. The down-regulation of genes in porphyrin metabolism, photosynthesis, carbon fixation, glycolysis, tricarboxylic acid cycle, and oxidative phosphorylation might inhibit chlorophyll synthesis, photosynthesis, and ATP supply, thereby hindering the growth and metabolism of algae. Moreover, the down-regulation of genes related to nucleotide metabolism and DNA replication might influence the reproduction of algae. These findings provided effective strategies to elucidate toxic mechanisms of contaminants on algae in algal bloom water.
    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
    COVID-19的动物模型有助于开发针对SARS-CoV-2的疫苗和抗病毒药物。抗病毒剂或疫苗的功效在具有不同程度疾病的不同动物模型中可能不同。这里,我们介绍了一个表达人血管紧张素转换酶2(ACE2)的小鼠模型。在这个模型中,通过CRISPR-Cas9(K18-hACE2KI)将具有人细胞角蛋白18启动子的ACE2敲入C57BL/6J小鼠的Hipp11基因座。鼻内接种高(3×105PFU)或低(2.5×102PFU)剂量的SARS-CoV-2野生型(WT),Delta,OmicronBA.1或OmicronBA.2变体,所有小鼠都表现出明显的感染症状,包括减肥,肺部病毒载量高,和间质性肺炎.在所有变体感染的K18-hACE2KI小鼠中观察到100%的致死率,延迟了Delta和BA.1的终点,并且观察到BA.2的致病性显着减弱。感染小鼠的肺炎伴随着中性粒细胞的浸润和肺纤维化。与K18-hACE2Tg小鼠和HFH4-hACE2Tg小鼠相比,K18-hACE2KI小鼠更容易感染SARS-CoV-2。在抗病毒药物测试中,REGN10933和Remdesivir在SARS-CoV-2感染的挑战下,K18-hACE2KI小鼠的抗病毒功效有限,而Nirmatrelvir,单克隆抗体4G4和mRNA疫苗有效保护小鼠免于死亡。我们的结果表明,K18-hACE2KI小鼠模型对SARS-CoV-2感染是致命且稳定的,并且对抗病毒开发是可行和严格的。
    Animal models of COVID-19 facilitate the development of vaccines and antivirals against SARS-CoV-2. The efficacy of antivirals or vaccines may differ in different animal models with varied degrees of disease. Here, we introduce a mouse model expressing human angiotensin-converting enzyme 2 (ACE2). In this model, ACE2 with the human cytokeratin 18 promoter was knocked into the Hipp11 locus of C57BL/6J mouse by CRISPR - Cas9 (K18-hACE2 KI). Upon intranasal inoculation with high (3 × 105 PFU) or low (2.5 × 102 PFU) dose of SARS-CoV-2 wildtype (WT), Delta, Omicron BA.1, or Omicron BA.2 variants, all mice showed obvious infection symptoms, including weight loss, high viral loads in the lung, and interstitial pneumonia. 100% lethality was observed in K18-hACE2 KI mice infected by variants with a delay of endpoint for Delta and BA.1, and a significantly attenuated pathogenicity was observed for BA.2. The pneumonia of infected mice was accompanied by the infiltration of neutrophils and pulmonary fibrosis in the lung. Compared with K18-hACE2 Tg mice and HFH4-hACE2 Tg mice, K18-hACE2 KI mice are more susceptible to SARS-CoV-2. In the antivirals test, REGN10933 and Remdesivir had limited antiviral efficacies in K18-hACE2 KI mice upon the challenge of SARS-CoV-2 infections, while Nirmatrelvir, monoclonal antibody 4G4, and mRNA vaccines potently protected the mice from death. Our results suggest that the K18-hACE2 KI mouse model is lethal and stable for SARS-CoV-2 infection, and is practicable and stringent to antiviral development.
    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
    即使使用有效的疫苗,与没有CKD的患者相比,患有CKD的患者在COVID-19感染后住院和死亡的风险更高.Molnupiravir和nirmatrelvir-ritonavir已被批准用于紧急用途,但它们对CKD人群的有效性仍然未知。进行这项研究是为了确定这些药物在降低CKD人群死亡率和严重COVID-19方面的有效性。
    这是一项使用香港电子健康数据库的目标试验模拟研究。纳入因COVID-19住院的18岁或以上CKD患者。COVID-19口服抗病毒引发剂的平均治疗效果,包括全因死亡率,重症监护病房(ICU)入院,和28天内的通气支持,与非发起者进行了比较。
    已发现抗病毒药物可降低全因死亡的风险,Molnupiravir的危险比(HR)为0.85(95%置信区间[CI],0.77至0.95]和nirmatrelvir-利托那韦,HR为0.78[95%CI,0.60至1.00]。然而,它们并不能显着降低ICU入院的风险(molnupiravir:HR,0.88[95%CI,0.59至1.30];尼马特雷韦-利托那韦:HR,0.86[95%CI,0.56至1.32])或通气支持(莫努比拉韦:HR,1.00[95%CI,0.76至1.33];尼马特雷韦-利托那韦:HR,1.01[95%CI,0.74至1.37])。男性和Charlson合并症指数(CCI)较高的男性风险降低更大。nirmatrelvir-ritonavir试验还显示,那些接受抗病毒治疗并接受3次或更多疫苗剂量的人的风险降低。
    莫那普拉韦和尼马特雷韦-利托那韦都降低了住院的COVID-19CKD患者的死亡率。
    UNASSIGNED: Even with effective vaccines, patients with CKD have a higher risk of hospitalization and death subsequent to COVID-19 infection than those without CKD. Molnupiravir and nirmatrelvir-ritonavir have been approved for emergency use, but their effectiveness for the CKD population is still unknown. This study was conducted to determine the effectiveness of these drugs in reducing mortality and severe COVID-19 in the CKD population.
    UNASSIGNED: This was a target trial emulation study using electronic health databases in Hong Kong. Patients with CKD aged 18 years or older who were hospitalized with COVID-19 were included. The per-protocol average treatment effect among COVID-19 oral antiviral initiators, including all-cause mortality, intensive care unit (ICU) admission, and ventilatory support within 28 days, were compared to noninitiators.
    UNASSIGNED: Antivirals have been found to lower the risk of all-cause mortality, with Molnupiravir at a hazard ratio (HR) of 0.85 (95% confidence interval [CI], 0.77 to 0.95] and nirmatrelvir-ritonavir at an HR of 0.78 [95% CI, 0.60 to 1.00]. However, they do not significantly reduce the risk of ICU admission (molnupiravir: HR, 0.88 [95% CI, 0.59 to 1.30]; nirmatrelvir-ritonavir: HR, 0.86 [95% CI, 0.56 to 1.32]) or ventilatory support (molnupiravir: HR, 1.00 [95% CI, 0.76 to 1.33]; nirmatrelvir-ritonavir: HR, 1.01 [95% CI, 0.74 to 1.37]). There was a greater risk reduction in males and those with higher Charlson Comorbidity Index (CCI). The nirmatrelvir-ritonavir trial also showed reduced risk for those who had antiviral treatment and received 3 or more vaccine doses.
    UNASSIGNED: Both molnupiravir and nirmatrelvir-ritonavir reduced mortality rates for hospitalized COVID-19 patients with CKD.
    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
    全球重新出现的呼吸道病原体肠道病毒D68(EV-D68)与严重呼吸道疾病的爆发有关,并与急性弛缓性脊髓炎有关。然而,对于EV-D68感染仍然缺乏有效的治疗方法.在这项工作中,我们发现宿主Toll样受体7(TLR7)蛋白,作为强大的先天免疫传感器,在响应EV-D68感染时表达选择性升高。随后,我们调查了维沙莫德(GS-9620)的影响,Toll样受体7激动剂,在EV-D68复制上。我们的发现揭示EV-D68感染导致TLR7的mRNA水平增加。用Vesatolimod治疗可显着抑制EV-D68复制[最大有效浓度的一半(EC50)=0.1427µM],而在杀病毒浓度下不会诱导明显的细胞毒性。尽管Vesatolimod对EV-D68附件的影响有限,病毒进入后抑制RNA复制和病毒蛋白合成。Vesatolimod广泛抑制了循环分离的EV-D68菌株的复制。此外,我们的研究结果表明,Vesatolimod治疗赋予了呼吸道和神经细胞对EV-D68感染的抵抗.总的来说,这些结果为药物开发提供了一种有前景的策略,即通过药理学激活TLR7,选择性地在EV-D68感染的细胞中启动抗病毒状态.重要的抗病毒药物开发的常规策略主要集中在直接靶向病毒蛋白酶或关键成分,以及参与病毒复制的宿主蛋白。在这项研究中,基于我们有趣的发现,肠道病毒D68(EV-D68)感染特异性上调免疫传感器Toll样受体7(TLR7)蛋白的表达,在呼吸细胞中不存在或低水平表达,我们提出了一种潜在的抗病毒方法,利用TLR7激动剂激活EV-D68感染的细胞进入抗病毒防御状态。值得注意的是,我们的研究结果表明,TLR7的药理激活通过TLR7/MyD88依赖性机制有效抑制呼吸道细胞中EV-D68的复制.这项研究不仅提出了一种有前途的药物候选物和针对EV-D68传播的靶标,而且还强调了利用病毒感染诱导的细胞先天免疫反应的独特改变的潜力。在受感染的细胞中选择性地诱导防御状态,同时保护未感染的正常细胞免受与治疗干预相关的潜在不利影响。
    The globally reemerging respiratory pathogen enterovirus D68 (EV-D68) is implicated in outbreaks of severe respiratory illness and associated with acute flaccid myelitis. However, there remains a lack of effective treatments for EV-D68 infection. In this work, we found that the host Toll-like receptor 7 (TLR7) proteins, which function as powerful innate immune sensors, were selectively elevated in expression in response to EV-D68 infection. Subsequently, we investigated the impact of Vesatolimod (GS-9620), a Toll-like receptor 7 agonist, on EV-D68 replication. Our findings revealed that EV-D68 infection resulted in increased mRNA levels of TLR7. Treatment with Vesatolimod significantly inhibited EV-D68 replication [half maximal effective concentration (EC50) = 0.1427 µM] without inducing significant cytotoxicity at virucidal concentrations. Although Vesatolimod exhibited limited impact on EV-D68 attachment, it suppressed RNA replication and viral protein synthesis after virus entry. Vesatolimod broadly inhibited the replication of circulating isolated strains of EV-D68. Furthermore, our findings demonstrated that treatment with Vesatolimod conferred resistance to both respiratory and neural cells against EV-D68 infection. Overall, these results present a promising strategy for drug development by pharmacologically activating TLR7 to initiate an antiviral state in EV-D68-infected cells selectively.IMPORTANCEConventional strategies for antiviral drug development primarily focus on directly targeting viral proteases or key components, as well as host proteins involved in viral replication. In this study, based on our intriguing discovery that enterovirus D68 (EV-D68) infection specifically upregulates the expression of immune sensor Toll-like receptor 7 (TLR7) protein, which is either absent or expressed at low levels in respiratory cells, we propose a potential antiviral approach utilizing TLR7 agonists to activate EV-D68-infected cells into an anti-viral defense state. Notably, our findings demonstrate that pharmacological activation of TLR7 effectively suppresses EV-D68 replication in respiratory tract cells through a TLR7/MyD88-dependent mechanism. This study not only presents a promising drug candidate and target against EV-D68 dissemination but also highlights the potential to exploit unique alterations in cellular innate immune responses induced by viral infections, selectively inducing a defensive state in infected cells while safeguarding uninfected normal cells from potential adverse effects associated with therapeutic interventions.
    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
    在复杂的动物生物学领域,许多重要的过程严重依赖于精确协调的蛋白酶级联,但是潜在的破坏使蛋白酶抑制剂不可或缺,丝氨酸蛋白酶抑制剂(serpin)处于最前沿,充当体内平衡的保管人,并参与各种关键的生物过程。重要的是,serpin功能仍有许多未开发的方面。在这项研究中,我们专注于日本浮对虾的serpin家族蛋白,利用微调预训练的蛋白质语言模型。这种方法导致了28个serpin的鉴定和进化验证,其中之一,Mjserpin-1被称为Mjserpin-1,在计算和实验上都证明了其作为抗病毒和凋亡抑制剂的潜力。我们的研究揭示了最先进的人工智能和丰富的生物信息学融合的令人兴奋的前景,拥有重大发现的希望,可以为未来的治疗进展铺平道路。
    In the intricate realm of animal biology, a multitude of vital processes heavily rely on precisely orchestrated proteinase cascades, but the potential for havoc makes proteinase inhibitors indispensable, with serine proteinase inhibitors (serpins) at the forefront, serving as custodians of homeostasis and participating in various critical biological processes. Importantly, there are still many unexplored facets of serpin functionality. In this study, we focused on the serpin family proteins from Marsupenaeus japonicus, utilizing a fine-tuned pretrained protein language model. This approach led to the identification and evolutionary validation of 28 serpins, one of which, referred to as Mjserpin-1, was both computationally and experimentally demonstrated to show potential as an antiviral and apoptosis inhibitor. Our research unveils exciting prospects for the fusion of state-of-the-art artificial intelligence and rich bioinformatics, holding the promise of significant discoveries that could pave the way for future therapeutic advancements.
    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
    慢性病毒性肝炎是由乙型肝炎病毒引起的,丙型肝炎病毒或丁型肝炎病毒(HBV,HCV,和HDV)。尽管复制策略不同,所有这些病毒都依赖于宿主内质网-高尔基途径的分泌,为抗病毒治疗提供潜在的宿主靶标。病毒细胞培养模型中跨膜6超家族成员2(TM6SF2)的敲减减少了感染性HCV病毒粒子的分泌,HDV病毒体和HBV亚病毒颗粒。此外,在一组乙型肝炎患者中,TM6SF2多态性(rs58542926CT/TT,导致肝脏中蛋白质错误折叠和TM6SF2减少)与血液中亚病毒颗粒浓度降低相关,补充了我们以前的工作,表明具有这种多态性的人的HCV病毒载量降低。总之,宿主蛋白TM6SF2在HBV的分泌中起关键作用,HCV和HDV,提供新型泛病毒药物治疗慢性病毒性肝炎的潜力。
    Chronic viral hepatitis is caused by hepatitis B virus, hepatitis C virus or hepatitis D virus (HBV, HCV, and HDV). Despite different replication strategies, all these viruses rely on secretion through the host endoplasmic reticulum-Golgi pathway, providing potential host targets for antiviral therapy. Knockdown of transmembrane 6 superfamily member 2 (TM6SF2) in virus cell culture models reduced secretion of infectious HCV virions, HDV virions and HBV subviral particles. Moreover, in a cohort of people with hepatitis B a TM6SF2 polymorphism (rs58542926 CT/TT, which causes protein misfolding and reduced TM6SF2 in the liver) correlated with lower concentrations of subviral particles in blood, complementing our previous work showing decreased HCV viral load in people with this polymorphism. In conclusion, the host protein TM6SF2 plays a key role in secretion of HBV, HCV and HDV, providing the potential for novel pan-viral agents to treat people with chronic viral hepatitis.
    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