cross-immunity

交叉免疫
  • 文章类型: Journal Article
    猴痘病毒(MPXV)的人传人增多令人担忧,和针对牛痘病毒(VACV)的抗体已知赋予针对水痘的交叉保护。我们使用了来自苏格兰患者群体的430份血清样品来研究抗体介导的针对MPXV的交叉中和。通过将电化学发光免疫测定与活病毒中和测定相结合,我们显示,在英国常规接种天花疫苗时出生的人,交叉中和MPXV的抗体水平升高.我们的结果表明,年龄是水痘感染的危险因素,1971年以后出生的人在接触后感染的风险更高。
    Increased human-to-human transmission of monkeypox virus (MPXV) is cause for concern, and antibodies directed against vaccinia virus (VACV) are known to confer cross-protection against Mpox. We used 430 serum samples derived from the Scottish patient population to investigate antibody-mediated cross-neutralization against MPXV. By combining electrochemiluminescence immunoassays with live-virus neutralization assays, we show that people born when smallpox vaccination was routinely offered in the United Kingdom have increased levels of antibodies that cross-neutralize MPXV. Our results suggest that age is a risk factor of Mpox infection, and people born after 1971 are at higher risk of infection upon exposure.
    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
    对严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)的免疫力现在很普遍;但是,SARS-CoV-2与地方病的交叉免疫程度,季节性人类冠状病毒(HCoV)仍不清楚。
    SARS-CoV-2和HCoV交叉免疫是在参加美国III期子研究的成年参与者中评估的,AZD1222(ChAdOx1nCoV-19)主要系列疫苗接种1年的随机对照试验(NCT04516746)。抗HCoV-229E的抗HCoV刺突结合抗体,HCoV-HKU1,HCoV-OC43和HCoV-NL63在研究给药后的参与者中进行了评估,在AZD1222组中,在非研究第三剂加强后。通过基线抗HCoV滴度评估了接受AZD1222初级系列治疗的患者的SARS-CoV-2血清转化时间(通过抗核衣壳抗体水平评估)和COVID-19的发生率。
    我们评估了AZD1222组的2,020/21,634名参与者和安慰剂组的1,007/10,816名参与者。在一年的数据截止时间(2022年3月11日),AZD1222(n=1,940)和安慰剂(n=962)组的参与者的平均随访时间为230.9(SD:106.36,范围:1-325)和94.3(74.12,1-321)天,分别。我们观察到研究给药后或加强后的抗HCoV体液滴度几乎没有升高,也没有证据表明随着时间的推移。SARS-CoV-2血清转换的发生和时间以及COVID-19的发生率在很大程度上不受基线抗HCoV滴度的影响。
    我们发现在AZD1222主要系列和加强疫苗接种后SARS-CoV-2和HCoV之间交叉免疫的证据有限。尽管全球人群中SARS-CoV-2免疫的流行率很高,但对未来新型冠状病毒出现的易感性可能会持续存在。
    UNASSIGNED: Immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is now widespread; however, the degree of cross-immunity between SARS-CoV-2 and endemic, seasonal human coronaviruses (HCoVs) remains unclear.
    UNASSIGNED: SARS-CoV-2 and HCoV cross-immunity was evaluated in adult participants enrolled in a US sub-study in the phase III, randomized controlled trial (NCT04516746) of AZD1222 (ChAdOx1 nCoV-19) primary-series vaccination for one-year. Anti-HCoV spike-binding antibodies against HCoV-229E, HCoV-HKU1, HCoV-OC43, and HCoV-NL63 were evaluated in participants following study dosing and, in the AZD1222 group, after a non-study third-dose booster. Timing of SARS-CoV-2 seroconversion (assessed via anti-nucleocapsid antibody levels) and incidence of COVID-19 were evaluated in those who received AZD1222 primary-series by baseline anti-HCoV titers.
    UNASSIGNED: We evaluated 2,020/21,634 participants in the AZD1222 group and 1,007/10,816 in the placebo group. At the one-year data cutoff (March 11, 2022) mean duration of follow up was 230.9 (SD: 106.36, range: 1-325) and 94.3 (74.12, 1-321) days for participants in the AZD1222 (n = 1,940) and placebo (n = 962) groups, respectively. We observed little elevation in anti-HCoV humoral titers post study-dosing or post-boosting, nor evidence of waning over time. The occurrence and timing of SARS-CoV-2 seroconversion and incidence of COVID-19 were not largely impacted by baseline anti-HCoV titers.
    UNASSIGNED: We found limited evidence for cross-immunity between SARS-CoV-2 and HCoVs following AZD1222 primary series and booster vaccination. Susceptibility to future emergence of novel coronaviruses will likely persist despite a high prevalence of SARS-CoV-2 immunity in global populations.
    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
    许多临床参数与2019年严重冠状病毒疾病有关,但预防有症状疾病的因素仍然未知。我们调查了严重急性呼吸综合征冠状病毒2型(SARS-CoV-2)和地方性人类冠状病毒(HCoV)抗体反应对纵向儿童队列(n=2,917)和包括儿童和成人的横断面队列(n=882)症状的影响。所有人都是在瑞士首次接触SARS-CoV-2(2020年3月至2021年3月)。唾液(n=4,993)和血浆(n=7,486)抗体对四个HCoV(亚基S1[S1])和SARS-CoV-2(S1,受体结合域,S2亚基[S2],核衣壳蛋白)的测定以及对SARS-CoV-2武汉的中和活性,阿尔法,Delta,和Omicron(BA.2)在一个子集的个体中。最近推断的SARS-CoV-2感染与粘膜和全身性SARS-CoV-2抗尖峰反应之间的强相关性相关。具有预先存在的HCoV-S1反应性的个体在两种血浆中均表现出对SARS-CoV-2的抗体应答显着升高(IgG回归系数=0.20,95%CI=[0.09,0.32],P<0.001)和唾液(IgG回归系数=0.60,95%CI=[0.088,1.11],P=0.025)。唾液中和活性适中,但令人惊讶的是广泛,对武汉的保留活性(NT50中位数=32.0,1Q-3Q=[16.4,50.2]),Alpha(NT50中位数=34.9,1Q-3Q=[26.0,46.6]),和Delta(NT50中位数=28.0,1Q-3Q=[19.9,41.7])。与交叉反应性HCoV免疫引发的快速粘膜防御一致,无症状个体血浆中预先存在的HCoV-S1活性较高(IgGHKU1,比值比[OR]=0.53,95%CI=[0.29,0.97],P=0.038)和唾液(总HCoV,OR=0.55,95%CI=[0.33,0.91],P=0.019)和唾液中更高的SARS-CoV-2反应性(IgGS2倍数变化=1.26,95%CI=[1.03,1.54],P=0.030)。通过调查未事先接触SARS-CoV-2或接种疫苗的人群对SARS-CoV-2和HCoV的全身和粘膜免疫反应,我们确定了与症状缺乏相关的特异性抗体反应性.重要信息了解人类冠状病毒(HCoV)免疫与严重急性呼吸道综合症冠状病毒2型(SARS-CoV-2)感染之间的相互作用对于了解当前地方性冠状病毒的共存以及建立潜在的未来人畜共患冠状病毒传播的知识至关重要。这项研究,该研究回顾性分析了2020-2021年在瑞士首次接触SARS-CoV-2的一大群人,揭示了几个关键发现。预先存在的HCoV免疫力,特别是粘膜抗体反应,在改善SARS-CoV-2感染后的免疫反应和减少症状发展方面发挥了重要作用。对SARS-CoV-2的粘膜中和活性,尽管幅度较低,针对SARS-CoV-2变体的保留活性强调了维持对SARS-CoV-2的局部粘膜免疫的重要性。尽管HCoV免疫的交叉保护作用不足以阻止SARS-CoV-2感染,但本研究表明对限制症状性疾病具有显着影响。这些发现支持通过诱导有效的粘膜免疫反应来产生泛保护性冠状病毒疫苗的可行性。
    Numerous clinical parameters link to severe coronavirus disease 2019, but factors that prevent symptomatic disease remain unknown. We investigated the impact of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) and endemic human coronavirus (HCoV) antibody responses on symptoms in a longitudinal children cohort (n = 2,917) and a cross-sectional cohort including children and adults (n = 882), all first exposed to SARS-CoV-2 (March 2020 to March 2021) in Switzerland. Saliva (n = 4,993) and plasma (n = 7,486) antibody reactivity to the four HCoVs (subunit S1 [S1]) and SARS-CoV-2 (S1, receptor binding domain, subunit S2 [S2], nucleocapsid protein) was determined along with neutralizing activity against SARS-CoV-2 Wuhan, Alpha, Delta, and Omicron (BA.2) in a subset of individuals. Inferred recent SARS-CoV-2 infection was associated with a strong correlation between mucosal and systemic SARS-CoV-2 anti-spike responses. Individuals with pre-existing HCoV-S1 reactivity exhibited significantly higher antibody responses to SARS-CoV-2 in both plasma (IgG regression coefficients = 0.20, 95% CI = [0.09, 0.32], P < 0.001) and saliva (IgG regression coefficient = 0.60, 95% CI = [0.088, 1.11], P = 0.025). Saliva neutralization activity was modest but surprisingly broad, retaining activity against Wuhan (median NT50 = 32.0, 1Q-3Q = [16.4, 50.2]), Alpha (median NT50 = 34.9, 1Q-3Q = [26.0, 46.6]), and Delta (median NT50 = 28.0, 1Q-3Q = [19.9, 41.7]). In line with a rapid mucosal defense triggered by cross-reactive HCoV immunity, asymptomatic individuals presented with higher pre-existing HCoV-S1 activity in plasma (IgG HKU1, odds ratio [OR] = 0.53, 95% CI = [0.29,0.97], P = 0.038) and saliva (total HCoV, OR = 0.55, 95% CI = [0.33, 0.91], P = 0.019) and higher SARS-CoV-2 reactivity in saliva (IgG S2 fold change = 1.26, 95% CI = [1.03, 1.54], P = 0.030). By investigating the systemic and mucosal immune responses to SARS-CoV-2 and HCoVs in a population without prior exposure to SARS-CoV-2 or vaccination, we identified specific antibody reactivities associated with lack of symptom development.IMPORTANCEKnowledge of the interplay between human coronavirus (HCoV) immunity and severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection is critical to understanding the coexistence of current endemic coronaviruses and to building knowledge potential future zoonotic coronavirus transmissions. This study, which retrospectively analyzed a large cohort of individuals first exposed to SARS-CoV-2 in Switzerland in 2020-2021, revealed several key findings. Pre-existing HCoV immunity, particularly mucosal antibody responses, played a significant role in improving SARS-CoV-2 immune response upon infection and reducing symptoms development. Mucosal neutralizing activity against SARS-CoV-2, although low in magnitude, retained activity against SARS-CoV-2 variants underlining the importance of maintaining local mucosal immunity to SARS-CoV-2. While the cross-protective effect of HCoV immunity was not sufficient to block infection by SARS-CoV-2, the present study revealed a remarkable impact on limiting symptomatic disease. These findings support the feasibility of generating pan-protective coronavirus vaccines by inducing potent mucosal immune responses.
    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:人类针对不同冠状病毒的一般免疫反应相似性可能反映了一定程度的交叉免疫,因此,暴露于一种冠状病毒可能会赋予另一种冠状病毒部分免疫力。目的是确定先前的MERS-CoV感染是否与随后的COVID-19疾病及其相关结局的较低风险相关。
    方法:我们对2012年至2020年初在沙特阿拉伯一家三级医院筛查MERS-CoV的所有患者进行了回顾性队列研究。从2020年初至2021年9月,MERS-CoV阳性和阴性患者均接受了RT-PCR检测证实的COVID-19感染的随访。
    结果:共有397名参与者在COVID-19大流行期间平均随访15个月(距MERS-CoV感染4.9年)。在397名参与者中,基线时,93例(23.4%)的MERS-CoV阳性;61例(65.6%)的阳性病例是有症状的。在397名参与者中,48名(12.1%)参与者在随访期结束时发展为COVID-19。Cox回归分析调整了年龄,性别,和主要合并症显示,在MERS-CoV阳性的患者中,COVID-19疾病(风险比=0.533,p=0.085)和住院(风险比=0.411,p=0.061)的风险略显着降低。此外,在有症状的MERS-CoV感染(风险比=0.324,p=0.034)和住院(风险比=0.317,p=0.042)的患者中,COVID-19疾病的风险进一步降低,并且变得显著.
    结论:当前的发现可能表明部分交叉免疫,有症状的MERS-CoV患者未来发生COVID-19感染和相关住院的风险较低。目前的结果可能需要使用免疫标记在全国范围内进行进一步检查。
    BACKGROUND: The general human immune responses similarity against different coronaviruses may reflect some degree of cross-immunity, whereby exposure to one coronavirus may confer partial immunity to another. The aim was to determine whether previous MERS-CoV infection was associated with a lower risk of subsequent COVID-19 disease and its related outcomes.
    METHODS: We conducted a retrospective cohort study among all patients screened for MERS-CoV at a tertiary care hospital in Saudi Arabia between 2012 and early 2020. Both MERS-CoV positive and negative patients were followed up from early 2020 to September 2021 for developing COVID-19 infection confirmed by RT-PCR testing.
    RESULTS: A total of 397 participants followed for an average 15 months during COVID-19 pandemic (4.9 years from MERS-CoV infection). Of the 397 participants, 93 (23.4%) were positive for MERS-CoV at baseline; 61 (65.6%) of the positive cases were symptomatic. Out of 397, 48 (12.1%) participants developed COVID-19 by the end of the follow-up period. Cox regression analysis adjusted for age, gender, and major comorbidity showed a marginally significant lower risk of COVID-19 disease (hazard ratio = 0.533, p = 0.085) and hospital admission (hazard ratio = 0.411, p = 0.061) in patients with positive MERS-CoV. Additionally, the risk of COVID-19 disease was further reduced and became significant in patients with symptomatic MERS-CoV infection (hazard ratio = 0.324, p = 0.034) and hospital admission (hazard ratio = 0.317, p = 0.042).
    CONCLUSIONS: The current findings may indicate a partial cross-immunity, where patients with symptomatic MERS-CoV have a lower risk of future COVID-19 infection and related hospitalization. The present results may need further examination nationally using immunity markers.
    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
    霍乱弧菌严重依赖水生水库作为传播途径,在最近的许多疫情中观察到两种不同的血清型。在本文中,我们扩展了先前研究的常微分方程流行病学模型,以创建一个两菌株SIRP(易感感染-恢复病原体)系统,该系统同时包含疾病菌株和环境病原体传播之间的部分交叉免疫.特别感兴趣的是无阻尼反相位周期解,因为这些表现出一种共存的类型,菌株通常会切换主导地位,了解是什么推动了这种转变,可以优化宿主群体对疾病的控制措施的效率。我们得出基本繁殖数R0,并使用稳定性分析来检查无病和单菌株平衡。我们制定了独特的共存平衡,并证明了当R0>1时两种菌株的均匀持久性。此外,我们模拟这个系统的解决方案,以及有和没有宿主共感染的模型的季节性强制版本。交叉免疫和传播途径影响阻尼或持续振荡动力学,季节性的存在可以改变,扩增或同步血清型的周期和阶段,推动流行病浪潮。在大的时间间隔内循环血清型,与观察到的数据相似,发现了一系列交叉免疫水平,并且在模型中包含共感染有助于持续的反相位周期解。
    The bacteria Vibrio cholerae relies heavily upon an aquatic reservoir as a transmission route with two distinct serotypes observed in many recent outbreaks. In this paper, we extend previously studied ordinary differential equation epidemiological models to create a two-strain SIRP (susceptible-infectious-recovered-pathogen) system which incorporates both partial cross-immunity between disease strains and environmental pathogen transmission. Of particular interest are undamped anti-phase periodic solutions, as these display a type of coexistence where strains routinely switch dominance, and understanding what drives this switch can optimize the efficiency of the host population\'s control measures against the disease. We derive the basic reproduction number R0 and use stability analysis to examine the disease free and single-strain equilibria. We formulate a unique coexistence equilibrium and prove uniform persistence of both strains when R0>1. In addition, we simulate solutions to this system, along with seasonally forced versions of the model with and without host coinfection. Cross-immunity and transmission pathways influence damped or sustained oscillatory dynamics, where the presence of seasonality can modify, amplify or synchronize the period and phase of serotypes, driving epidemic waves. Cycling of serotypes over large time intervals, similar to observed data, is found for a range of cross-immunity levels, and the inclusion of coinfection in the model contributes to sustained anti-phase periodic solutions.
    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
    毒力进化理论中历史悠久的范式假定传染性和危害性之间存在正相关关系。然而,呼吸系统疾病的病因产生了负相关,下呼吸道疾病的感染性较低,危害更大。我们在一个简单的模型中探索进化结果,该模型包含疾病菌株之间的交叉免疫,这些疾病菌株随着它们在呼吸道中的距离而减少,假设对接速率遵循细胞表面类型的局部混合与病原体表面之间的匹配,和交叉免疫病原体表面的相似性。健身成分之间的假定关系导致感染下气道的毒株进化到更温和,更多可传播的变体。有限的交叉免疫通常会导致多样化的准备,这随着宿主人口密度的增加而增加。在呼吸系统疾病中,上呼吸道的多样性最高。试探性地,新出现的呼吸系统疾病可能开始低毒,进化并变得更温和。我们的结果扩展到疾病生态学的现实概括,并包括其他表位轴。这些扩展使我们能够定量地应用我们的结果来阐明鼻和冠状病毒引起的普通感冒之间的多样化差异。
    The time-honored paradigm in the theory of virulence evolution assumes a positive relation between infectivity and harmfulness. However, the etiology of respiratory diseases yields a negative relation, with diseases of the lower respiratory tract being less infective and more harmful. We explore the evolutionary consequences in a simple model incorporating cross-immunity between disease strains that diminishes with their distance in the respiratory tract, assuming that docking rate follows the match between the local mix of cell surface types and the pathogen\'s surface and cross-immunity the similarity of the pathogens\' surfaces. The assumed relation between fitness components causes virulent strains infecting the lower airways to evolve to milder more transmissible variants. Limited cross-immunity, generally, causes a readiness to diversify that increases with host population density. In respiratory diseases that diversity will be highest in the upper respiratory tract. More tentatively, emerging respiratory diseases are likely to start low and virulent, to evolve up, and become milder. Our results extend to a panoply of realistic generalizations of the disease\'s ecology to including additional epitope axes. These extensions allow us to apply our results quantitatively to elucidate the differences in diversification between rhino- and coronavirus caused common colds.
    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
    多品系疾病导致人之间一定程度的交叉免疫力的发展。在本论文中,我们提出了具有潜伏期和免疫时滞的多时滞SIRC传染病模型。在这里,我们旨在研究和研究孵化延迟的影响[公式:参见正文]以及提供部分/交叉免疫与免疫延迟参数([公式:参见正文])的疫苗对疾病动力学的影响。此外,我们研究了交叉免疫强度[公式:见正文]对疾病患病率的影响。建立了传染病模型解的积极性和有界性。已推导出两种不同类型的平衡点(无病和地方性)。已导出基本再现数的表达式。已经讨论了在不存在和存在两个延迟的情况下两个平衡点的稳定性条件和Hopf分叉。建立了关于特有平衡点的Lyapunov稳定性条件。已经进行了数值模拟以支持我们的分析结果。我们定量地证明了振荡和Hopf分叉如何允许时间延迟来改变系统的动力学。已经研究了两种延误对疾病患病率的综合影响。通过参数敏感性分析,我们观察到,感染人群随着疫苗接种率的增加而减少,并且随着交叉免疫率的增加,系统在早期开始稳定.已使用拉丁超立方体采样和部分秩相关系数技术对基本再现数进行了全局敏感性分析。已讨论了疫苗接种率与传播率和疫苗接种率与再感染概率(即交叉免疫强度)对[公式:见正文]的综合影响。我们的研究强调需要在流行病模型中考虑交叉免疫和时间延迟,以便更好地了解疾病动态。
    Multi-strain diseases lead to the development of some degree of cross-immunity among people. In the present paper, we propose a multi-delayed SIRC epidemic model with incubation and immunity time delays. Here we aim to examine and investigate the effects of incubation delay [Formula: see text] and the impact of vaccine which provides partial/cross-immunity with immunity delay parameter ([Formula: see text]) on the disease dynamics. Also, we study the impact of the strength of cross-immunity [Formula: see text] on the disease prevalence. The positivity and boundedness of the solutions of the epidemic model have been established. Two different types of equilibrium points (disease-free and endemic) have been deduced. Expression for basic reproduction number has been derived. The stability conditions and Hopf-bifurcation about both the equilibrium points in the absence and presence of both delays have been discussed. The Lyapunov stability conditions about the endemic equilibrium point have been established. Numerical simulations have been performed to support our analytical results. We quantitatively demonstrate how oscillations and Hopf-bifurcation allow time delays to alter the dynamics of the system. The combined impacts of both the delays on disease prevalence has been studied. Through parameter sensitivity analysis, we observe that the infected population decreases with an increase in vaccination rate and the system starts to stabilize early with the increase in cross-immunity rate. Global sensitivity analysis for the basic reproduction number has been performed using Latin hypercube sampling and partial rank correlation coefficients techniques. The combined effect of vaccination rate with transmission rate and vaccination rate with re-infection probability (i.e. strength of cross-immunity) on [Formula: see text] have been discussed. Our research underlines the need to take cross-immunity and time delays into account in the epidemic model in order to better understand disease dynamics.
    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
    自2019年底首次报道SARS-CoV-2以来,原始病毒出现了多种变异。每个变种都有突变的积累,特别是在尖峰糖蛋白中,与增加的病毒传播性和逃避免疫力有关。不同变体的刺突蛋白中的不同突变塑造了随后的抗体和T细胞应答,这样暴露于不同的刺突蛋白可以导致对异源变体的反应降低或增强。全球范围内,人们已经暴露并重新暴露于祖先菌株的多种变体,包括关注的五种变体。研究表明,个体的保护性免疫应答受它们所暴露的菌株或菌株组合的影响。最初暴露于特定毒株还可以塑造其随后的免疫模式和对随后的异源病毒感染的反应。大多数免疫学观察是在大流行期间祖先菌株循环时进行的。然而,SARS-CoV-2变体表现出不同的疾病严重程度模式,免疫力下降,免疫逃避和对治疗的敏感性。在这里,我们研究了先前感染了关注变体(VOC)并随后再次感染了异源变体的仓鼠的交叉保护。我们还确定了交叉保护和免疫力是否依赖于仓鼠首次暴露的特定病毒。我们进一步分析了每种SARS-CoV-2变体以及再次感染后诱导的宿主细胞因子反应。对三种VOC的比较分析表明,Alpha变体是最致病的VOC。我们表明,自然获得性免疫保护仓鼠免受随后与异源SARS-CoV-2变体的再感染,无论动物最初接触的是哪种变种。我们的研究支持观察到,不同SARS-CoV-2变体的异源感染不会在随后的再感染中加剧疾病。新的SARS-CoV-2变体的不断出现要求更好地理解感染个体的交叉保护和免疫印迹。这些信息对于指导新出现的SARS-CoV-2VOC和未来新型大流行冠状病毒的疫苗策略和公共政策至关重要。
    Since SARS-CoV-2 was first reported in late 2019, multiple variations of the original virus have emerged. Each variant harbors accumulations of mutations, particularly within the spike glycoprotein, that are associated with increased viral transmissibility and escape immunity. The different mutations in the spike protein of different variants shape the subsequent antibody and T cell responses, such that exposure to different spike proteins can result in reduced or enhanced responses to heterologous variants further down the line. Globally, people have been exposed and re-exposed to multiple variations of the Ancestral strain, including the five variants of concerns. Studies have shown that the protective immune response of an individual is influenced by which strain or combination of strains they are exposed to. The initial exposure to a specific strain may also shape their subsequent immune patterns and response to later infections with a heterologous virus. Most immunological observations were carried out early during the pandemic when the Ancestral strain was circulating. However, SARS-CoV-2 variants exhibit varying patterns of disease severity, waning immunity, immune evasion and sensitivity to therapeutics. Here we investigated the cross-protection in hamsters previously infected with a variant of concern (VOC) and subsequently re-infected with a heterologous variant. We also determined if cross-protection and immunity were dependent on the specific virus to which the hamster was first exposed. We further profiled the host cytokine response induced by each SARS-CoV-2 variants as well as subsequent to re-infection. A comparative analysis of the three VOCs revealed that Alpha variant was the most pathogenic VOC to emerge. We showed that naturally acquired immunity protected hamsters from subsequent re-infection with heterologous SARS-CoV-2 variant, regardless which variant the animal was first exposed to. Our study supports observations that heterologous infection of different SARS-CoV-2 variants do not exacerbate disease in subsequent re-infections. The continual emergence of new SARS-CoV-2 variants mandates a better understanding of cross-protection and immune imprinting in infected individuals. Such information is essential to guide vaccine strategy and public policy to emerging SARS-CoV-2 VOCs and future novel pandemic coronaviruses.
    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
    RNA病毒的高突变率导致新的突变体的出现,其可以逃脱由原始(野生型)毒株激活的免疫。然而,由于最初感染细胞数量很少,它们中的许多都因随机性而灭绝。在之前的一篇论文中,我们研究了当新型突变体具有更快的感染率并且对抑制野生型病毒的药物具有抗性时逃避随机灭绝的可能性。在这项研究中,我们研究逃避宿主免疫反应的效果。基于具有时间依赖性速率的连续时间分支过程,我们得出的结论是,当突变体产生时,由于野生型感染,[公式:见文本]随着时间的推移,建立突变菌株的机会[公式:见文本]减少。能够逃脱灭绝风险的新突变体的数量在野生型感染后不久达到峰值。对于非常强和非常弱的免疫反应,每个患者在宿主感染早期产生的新逃逸突变数量很少。当免疫活性达到中等强度时,它达到最大值。
    A high mutation rate of the RNA virus results in the emergence of novel mutants that may escape the immunity activated by the original (wild-type) strain. However, many of them go extinct because of the stochasticity due to the small initial number of infected cells. In a previous paper, we studied the probability of escaping stochastic extinction when the novel mutant has a faster rate of infection and when it is resistant to a drug that suppresses the wild-type virus. In this study, we examine the effect of escaping the immune reaction of the host. Based on a continuous-time branching process with time-dependent rates, we conclude the chance for a mutant strain to be established [Formula: see text] decreases with time [Formula: see text] since the wild-type infection when the mutant is produced. The number of novel mutants that can escape extinction risk has a peak soon after the wild-type infection. The number of novel escape mutations produced per patient in the early phase of host infection is small both for very strong and very weak immune responses, and it attains its maximum value when immune activity is of an intermediate strength.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    The WHO regularly updates influenza vaccine recommendations to maximize their match with circulating strains. Nevertheless, the effectiveness of the influenza A vaccine, specifically its H3N2 component, has been low for several seasons. The aim of the study is to develop a mathematical model of cross-immunity based on the array of published WHO hemagglutination inhibition assay (HAI) data.
    In this study, a mathematical model was proposed, based on finding, using regression analysis, the dependence of HAI titers on substitutions in antigenic sites of sequences. The computer program we developed can process data (GISAID, NCBI, etc.) and create real-time databases according to the set tasks.
    Based on our research, an additional antigenic site F was identified. The difference in 1.6 times the adjusted R2, on subsets of viruses grown in cell culture and grown in chicken embryos, demonstrates the validity of our decision to divide the original data array by passage histories. We have introduced the concept of a degree of homology between two arbitrary strains, which takes the value of a function depending on the Hamming distance, and it has been shown that the regression results significantly depend on the choice of function. The provided analysis showed that the most significant antigenic sites are A, B, and E. The obtained results on predicted HAI titers showed a good enough result, comparable to similar work by our colleagues.
    The proposed method could serve as a useful tool for future forecasts, with further study to confirm its sustainability.
    Введение. Всемирная организация здравоохранения (ВОЗ) регулярно обновляет рекомендации по вакцинам против гриппа с целью достижения их максимального соответствия очередным циркулирующим штаммам. Тем не менее на протяжении нескольких сезонов эффективность вакцины против гриппа А, а именно её компоненты H3N2, определялась как низкая. Цель исследования разработка математической модели перекрёстного иммунитета на основании имеющегося массива опубликованных ВОЗ данных реакции торможения гемагглютинации (РТГА). Материалы и методы. В настоящей работе представлена математическая модель, основанная на нахождении с помощью регрессионного анализа зависимости титров РТГА от замен в антигенных сайтах последовательностей. Разработанная нами компьютерная программа имеет возможность обрабатывать данные (GISAID, NCBI и др.) и формировать в режиме реального времени базы данных согласно поставленным задачам. Результаты. На основе наших исследований был вычленен дополнительный антигенный сайт F. Разница в 1,6 раза скорректированного R2 на подмножествах вирусов, выращенных в культуре клеток и культивируемых в куриных эмбрионах, демонстрирует обоснованность нашего решения о разделении первоначального массива данных по пассажным историям. Нами введено понятие степени гомологичности между двумя произвольными штаммами, которая принимает значение функции, зависящей от дистанции Хэмминга, и показано, что результаты регрессии существенно зависят от выбора функции. Проведённый анализ показал, что наиболее значимыми антигенными сайтами являются A, B и E. Полученные результаты прогноза титров РТГА показали достаточно хороший результат, сопоставимый с аналогичными работами наших коллег. Заключение. Предложенный метод может послужить хорошим инструментом для будущих прогнозов с дальнейшим изучением для подтверждения его устойчивости.
    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