SARS‐COV‐2

SARS - CoV - 2
  • 文章类型: Journal Article
    本工作报道了源自没食子酸(没食子胺)的酰胺对严重急性呼吸综合征冠状病毒2(SARS-CoV-2)主要蛋白酶(Mpro)的抑制作用,以及细胞毒性评估和分子对接研究。除了没食子胺,还合成了其他相关化合物,并对Mpro进行了评估,共25个化合物。八种化合物在抑制测定期间呈现溶解度问题,且一种化合物未显示抑制活性。化合物3a,3b,和3f显示出最高的酶抑制,IC50=0.26±0.19µM,0.80±0.38µM,和2.87±1.17µM,分别。硒代胍6a的IC50值为5.42±2.89µM,与其非硒化同源物3c的比较表明,硫属元素的插入使化合物的抑制能力提高了约10倍。关于THP-1和Vero细胞的细胞毒性,化合物3e和3g,在Vero细胞中显示中等的细胞毒性,而对于THP-1,两者都是无毒的,CC50>150µM。衍生的3d对两种细胞系都显示出中等的细胞毒性,而6d对THP-1有中等毒性。所分析的其它化合物在所测试的浓度下不诱导显著的细胞毒性。化合物3a的分子对接结果,3b,和3f显示涉及没食子酸酯部分的羟基(OH)的氢键相互作用是相关的,以及羰基。
    The present work reports the inhibitory effect of amides derived from gallic acid (gallamides) against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease (Mpro), along with cytotoxicity evaluation and molecular docking studies. In addition to gallamides, other relevant compounds were also synthesized and evaluated against Mpro, making a total of 25 compounds. Eight compounds presented solubility issues during the inhibitory assay and one showed no inhibitory activity. Compounds 3a, 3b, and 3f showed the highest enzymatic inhibition with IC50 = 0.26 ± 0.19 µM, 0.80 ± 0.38 µM, and 2.87 ± 1.17 µM, respectively. Selenogallamide 6a exhibited IC50 values of 5.42 ± 2.89 µM and a comparison with its nonselenylated congener 3c shows that the insertion of the chalcogen moiety improved the inhibitory capacity of the compound by approximately 10 times. Regarding the cellular toxicity in THP-1 and Vero cells, compounds 3e and 3g, showed moderate cytotoxicity in Vero cells, while for THP-1 both were nontoxic, with CC50 > 150 µM. Derivative 3d showed moderate cytotoxicity against both cell lines, whereas 6d was moderatly toxic to THP-1. Other compounds analyzed do not induce substantial cellular toxicity at the concentrations tested. The molecular docking results for compounds 3a, 3b, and 3f show that hydrogen bonding interactions involving the hydroxyl groups (OH) of the gallate moiety are relevant, as well as the carbonyl group.
    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SARS-CoV-2出现以来,该病毒的RNA依赖性RNA聚合酶(RdRp)的所有亚基中的突变已被反复报道。尽管RdRp是抗病毒药物的主要靶标,探索这些突变的表型效应的实验研究是有限的。这项研究的重点是三个RdRp亚基nsp7,nsp8和nsp12中取代的表型效应,根据它们的发生率和潜在影响进行选择。我们使用纳米差示扫描荧光法和微尺度热电泳来检查这些突变对蛋白质稳定性和RdRp复合物组装的影响。我们观察到不同的影响;特别是,nsp8中的单个突变显着增加了其稳定性,如解链温度升高13°C所证明的那样,而nsp7和nsp8中的某些突变在RdRp复合物形成期间降低了它们对nsp12的结合亲和力。使用荧光酶测定法,我们评估了对RNA聚合酶活性的总体影响.我们发现大多数检测的突变改变了聚合酶的活性,通常作为对RdRp复合物的其他组分的稳定性或亲和力变化的直接结果。有趣的是,nsp8A21V和nsp12P323L突变的组合导致聚合酶活性增加50%.据我们所知,这是第一项生化研究,旨在证明在新兴的SARS-CoV-2亚变体中构成RdRp复合物的所有组分中氨基酸突变的影响.
    Since the emergence of SARS-CoV-2, mutations in all subunits of the RNA-dependent RNA polymerase (RdRp) of the virus have been repeatedly reported. Although RdRp represents a primary target for antiviral drugs, experimental studies exploring the phenotypic effect of these mutations have been limited. This study focuses on the phenotypic effects of substitutions in the three RdRp subunits: nsp7, nsp8, and nsp12, selected based on their occurrence rate and potential impact. We employed nano-differential scanning fluorimetry and microscale thermophoresis to examine the impact of these mutations on protein stability and RdRp complex assembly. We observed diverse impacts; notably, a single mutation in nsp8 significantly increased its stability as evidenced by a 13°C increase in melting temperature, whereas certain mutations in nsp7 and nsp8 reduced their binding affinity to nsp12 during RdRp complex formation. Using a fluorometric enzymatic assay, we assessed the overall effect on RNA polymerase activity. We found that most of the examined mutations altered the polymerase activity, often as a direct result of changes in stability or affinity to the other components of the RdRp complex. Intriguingly, a combination of nsp8 A21V and nsp12 P323L mutations resulted in a 50% increase in polymerase activity. To our knowledge, this is the first biochemical study to demonstrate the impact of amino acid mutations across all components constituting the RdRp complex in emerging SARS-CoV-2 subvariants.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Systematic Review
    卡波西肉瘤(KS)是一种血管增殖性疾病,具有病毒性病因和由免疫功能障碍引起的多因素发病机制。在受疱疹病毒等潜伏病毒感染影响的患者中,SARS-CoV-2感染可能导致宿主细胞的裂解周期重新激活。强大的免疫系统反应对于消除病原体和解决潜伏和非潜伏病毒感染至关重要。我们报告了一系列以SARS-CoV-2感染后肿瘤进展为特征的KS病例。我们对PubMed/MEDLINE和EMBASE数据库进行了系统的文献综述。关键词包括\"SARS-CoV-2,\"HHV-8,\"\"卡波西肉瘤,\“\”IL-6,\“和\”COVID-19。\"应用了英语语言限制。我们的研究未涵盖的项目被排除在外。KS是一种与免疫系统受损有关的复杂疾病。导致暂时性或永久性免疫缺陷的病症可触发病毒再激活或加重现有疾病。COVID-19患者细胞因子水平升高是可行的,再加上淋巴细胞下调和诱导疱疹病毒裂解再激活的治疗,可能有助于SARS-CoV-2感染后KS的进展。这些观察结果表明,应在SARS-CoV-2感染期间和之后对KS患者进行临床监测。然而,应收集前瞻性数据以验证这一假设,并增强我们对KS发病或进展相关机制的理解.
    Kaposi\'s sarcoma (KS) is an angio-proliferative disease with a viral etiology and a multifactorial pathogenesis that results from immune dysfunction. In patients affected by latent viral infections such as herpesviruses, SARS-CoV-2 infection may result in lytic cycle reactivation in host cells. A robust immune system response is crucial for eliminating pathogens and resolving both latent and non-latent viral infections. We report a case series of KS characterized by tumor progression after SARS-CoV-2 infection. We performed a systematic literature review of the PubMed/MEDLINE and EMBASE databases. The keyword terms included \"SARS-CoV-2,\" \"HHV-8,\" \"Kaposi\'s sarcoma,\" \"IL-6,\" and \"COVID-19.\" English language restriction was applied. Items not covered by our study were excluded. KS is a complex disease linked to an impaired immune system. Conditions that result in temporary or permanent immunodeficiency can trigger viral reactivation or exacerbate an existing disease. It is feasible that the increase in cytokine levels in COVID-19 patients, coupled with lymphocyte downregulation and treatment that induces herpesvirus lytic reactivation, may contribute to the progression of KS after SARS-CoV-2 infection. These observations suggest that patients with KS should be clinically monitored both during and after SARS-CoV-2 infection. Nevertheless, prospective data should be collected to validate this hypothesis and enhance our understanding of the mechanisms implicated in the onset or progression of KS.
    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
    严重急性呼吸系统综合症冠状病毒-2(SARS-CoV-2)的出现导致了全球COVID-19大流行,严重影响孕妇的健康。产科人群,已经脆弱,面临与COVID-19相关的发病率和死亡率增加,并因先前存在的合并症而加剧。最近的研究揭示了COVID-19与先兆子痫(PE)之间的潜在相关性,全世界孕产妇和围产期发病率的主要原因,强调探索这两个条件之间关系的意义。这里,我们回顾了PE与COVID-19的病理生理相似性,特别关注严重的COVID-19病例和与SARS-CoV-2感染相关的PE样综合征病例。我们强调了这两种情况之间的细胞和分子机制的相互联系,例如,肾素-血管紧张素系统的调节,紧密连接和屏障完整性,和补充系统。最后,我们讨论了COVID-19大流行的动态,包括变种的出现和疫苗接种的努力,塑造了临床情景,并影响了COVID-19和PE的严重程度和管理。有必要继续研究怀孕期间SARS-CoV-2感染的机制以及先前感染导致PE的潜在风险,以描述COVID-19和PE相互作用的复杂性,并改善这两种情况的临床管理。
    The emergence of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has led to the global COVID-19 pandemic, significantly impacting the health of pregnant women. Obstetric populations, already vulnerable, face increased morbidity and mortality related to COVID-19, aggravated by preexisting comorbidities. Recent studies have shed light on the potential correlation between COVID-19 and preeclampsia (PE), a leading cause of maternal and perinatal morbidity worldwide, emphasizing the significance of exploring the relationship between these two conditions. Here, we review the pathophysiological similarities that PE shares with COVID-19, with a particular focus on severe COVID-19 cases and in PE-like syndrome cases related with SARS-CoV-2 infection. We highlight cellular and molecular mechanistic inter-connectivity between these two conditions, for example, regulation of renin-angiotensin system, tight junction and barrier integrity, and the complement system. Finally, we discuss how COVID-19 pandemic dynamics, including the emergence of variants and vaccination efforts, has shaped the clinical scenario and influenced the severity and management of both COVID-19 and PE. Continued research on the mechanisms of SARS-CoV-2 infection during pregnancy and the potential risk of developing PE from previous infections is warranted to delineate the complexities of COVID-19 and PE interactions and to improve clinical management of both conditions.
    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
    这里,我们对不同临床表现的恢复期COVID-19患者的S1和受体结合域蛋白特异性B细胞进行了单细胞RNA测序.本研究旨在评估非典型记忆B细胞(MBC)在应对严重急性呼吸综合征冠状病毒2(SARS-CoV-2)感染中的作用和发育途径。结果揭示了与疾病严重程度相关的B细胞亚群的促炎特征,正如GADD45B等基因的上调所证明的那样,MAP3K8和NFKBIA在危重和严重个体中的应用。此外,非典型MBCs的分析表明,通过生发中心与常规MBCs相似的发育途径,正如参与生发中心过程的几个基因的表达所表明的那样,包括CXCR4、CXCR5、BCL2和MYC。此外,COVID-19中免疫反应特征性基因的上调,如ZFP36和DUSP1,表明非典型MBCs的分化和激活可能受SARS-CoV-2暴露的影响,这些基因可能有助于COVID-19恢复的免疫反应。我们的研究有助于更好地了解COVID-19中的非典型MBCs以及其他B细胞亚群在不同临床表现中的作用。
    Here, we performed single-cell RNA sequencing of S1 and receptor binding domain protein-specific B cells from convalescent COVID-19 patients with different clinical manifestations. This study aimed to evaluate the role and developmental pathway of atypical memory B cells (MBCs) in response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The results revealed a proinflammatory signature across B cell subsets associated with disease severity, as evidenced by the upregulation of genes such as GADD45B, MAP3K8, and NFKBIA in critical and severe individuals. Furthermore, the analysis of atypical MBCs suggested a developmental pathway similar to that of conventional MBCs through germinal centers, as indicated by the expression of several genes involved in germinal center processes, including CXCR4, CXCR5, BCL2, and MYC. Additionally, the upregulation of genes characteristic of the immune response in COVID-19, such as ZFP36 and DUSP1, suggested that the differentiation and activation of atypical MBCs may be influenced by exposure to SARS-CoV-2 and that these genes may contribute to the immune response for COVID-19 recovery. Our study contributes to a better understanding of atypical MBCs in COVID-19 and the role of other B cell subsets across different clinical manifestations.
    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
    将新药从最初发现带到患者床边的平均成本估计超过20亿美元,需要十多年的研究和开发。需要新的药物筛选技术,其可以在开发早期解析具有增加的临床应用可能性的候选药物,以便增加该管线的成本效益。例如,在COVID-19大流行期间,资源被迅速调动以确定有效的治疗性治疗,但许多主要抗病毒化合物在进展到人体试验时未能证明疗效.为了解决缺乏预测性临床前药物筛选工具的问题,PREDICT96-ALI,一种高通量(n=96)微生理系统(MPS),概括了初级人类气管支气管组织,适用于评估受关注的天然SARS-CoV-2变体的差异抗病毒功效。这里,PREDICT96-ALI解决了变体之间的差异病毒动力学和抗病毒化合物在药物剂量范围内的功效。PREDICT96-ALI能够将临床上有效的抗病毒疗法(如雷德西韦和尼马特雷韦)与未显示临床疗效的有希望的先导化合物区分开来。重要的是,来自这个概念验证研究跟踪的结果与已知的临床结果,证明该技术作为预后药物发现工具的可行性。
    The average cost to bring a new drug from its initial discovery to a patient\'s bedside is estimated to surpass $2 billion and requires over a decade of research and development. There is a need for new drug screening technologies that can parse drug candidates with increased likelihood of clinical utility early in development in order to increase the cost-effectiveness of this pipeline. For example, during the COVID-19 pandemic, resources were rapidly mobilized to identify effective therapeutic treatments but many lead antiviral compounds failed to demonstrate efficacy when progressed to human trials. To address the lack of predictive preclinical drug screening tools, PREDICT96-ALI, a high-throughput (n = 96) microphysiological system (MPS)  that recapitulates primary human tracheobronchial tissue,is adapted for the evaluation of differential antiviral efficacy of native SARS-CoV-2 variants of concern. Here, PREDICT96-ALI resolves both the differential viral kinetics between variants and the efficacy of antiviral compounds over a range of drug doses. PREDICT96-ALI is able to distinguish clinically efficacious antiviral therapies like remdesivir and nirmatrelvir from promising lead compounds that do not show clinical efficacy. Importantly, results from this proof-of-concept study track with known clinical outcomes, demonstrate the feasibility of this technology as a prognostic drug discovery tool.
    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
    首批批准的人类使用的针对严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)的疫苗是基于纳米技术的。虽然它们是模块化的,迅速生产,可以减轻疾病的严重程度,目前可用的疫苗在预防感染方面受到限制,强调全球对新型预防性疫苗技术的需求。记住这一点,我们着手开发一种灵活的纳米疫苗平台,用于鼻腔给药以诱导粘膜免疫,这是防止呼吸道病毒感染的最佳保护的基础。下一代多表位纳米疫苗共同递送免疫原性肽,通过免疫信息学工作流程选择,以及PD-L1表达的佐剂和调节剂。作为一个案例研究,我们以SARS-CoV-2肽作为相关抗原来验证该方法。该平台可以引起针对SARS-CoV-2的局部和系统性细胞和体液特异性反应。这导致免疫球蛋白A(IgA)的分泌,能够中和SARS-CoV-2,包括关注的变种,遵循异源免疫策略。考虑到当前基于纳米技术的疫苗所需的冷链分布的局限性,这表明冻干的纳米疫苗在室温下是长期稳定的,并且在重建时保持其体内功效。这使其与发展中国家特别相关,并提供了适应未来病毒威胁的模块化系统。
    The first approved vaccines for human use against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are nanotechnology-based. Although they are modular, rapidly produced, and can reduce disease severity, the currently available vaccines are restricted in preventing infection, stressing the global demand for novel preventive vaccine technologies. Bearing this in mind, we set out to develop a flexible nanovaccine platform for nasal administration to induce mucosal immunity, which is fundamental for optimal protection against respiratory virus infection. The next-generation multiepitope nanovaccines co-deliver immunogenic peptides, selected by an immunoinformatic workflow, along with adjuvants and regulators of the PD-L1 expression. As a case study, we focused on SARS-CoV-2 peptides as relevant antigens to validate the approach. This platform can evoke both local and systemic cellular- and humoral-specific responses against SARS-CoV-2. This led to the secretion of immunoglobulin A (IgA), capable of neutralizing SARS-CoV-2, including variants of concern, following a heterologous immunization strategy. Considering the limitations of the required cold chain distribution for current nanotechnology-based vaccines, it is shown that the lyophilized nanovaccine is stable for long-term at room temperature and retains its in vivo efficacy upon reconstitution. This makes it particularly relevant for developing countries and offers a modular system adaptable to future viral threats.
    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:2019年爆发的严重急性呼吸综合征冠状病毒(SARS-CoV-2)有必要调查其对妊娠结局和胎儿发育的潜在不利影响。
    目的:本研究旨在回顾妊娠期SARS-CoV-2感染对胎儿结局影响的证据。
    方法:这篇叙述性综述总结了PubMed和WebofScience自COVID-19爆发以来的文献,显示母亲在怀孕期间感染SARS-CoV-2对胎儿发育的影响。
    结果:妊娠期SARS-CoV-2感染可通过胎盘垂直传播,在子宫内和围产期,影响母胎免疫界面和胎盘功能。怀孕期间的病毒感染与中枢神经系统发育障碍和自闭症等疾病有关。呼吸道结构和功能的变化,免疫,和内脏系统也有报道。怀孕期间SARS-CoV-2感染与死产和早产风险增加有关。然而,所涉及的机制尚不清楚,可能包括细胞因子风暴,巨噬细胞介导,基因突变,甲基化,和其他表观遗传变化。在动物和临床研究中探索抗病毒治疗和其他干预措施的保护作用可能有助于改善结果。
    结论:妊娠期SARS-CoV-2感染通过垂直传播激活母胎免疫界面,对胎儿发育有短期和长期影响,包括中枢神经系统.未来的长期研究可能有助于提供证据,为干预措施提供信息,以降低不良后果的风险。
    BACKGROUND: The severe acute respiratory syndrome coronavirus (SARS-CoV-2) outbreak in 2019 has necessitated investigating its potential adverse effects on pregnancy outcomes and fetal development.
    OBJECTIVE: This study aimed to review the evidence on the impact of SARS-CoV-2 infection during pregnancy on fetal outcomes.
    METHODS: Literatures since the outbreak of COVID-19 from PubMed and Web of Science were summarized in this narrative review, to show the effects of maternal SARS-CoV-2 infection during pregnancy on fetal development.
    RESULTS: SARS-CoV-2 infection during pregnancy can be transmitted vertically through the placenta, both in utero and perinatally, affecting the maternal-fetal immune interface and placental function. Viral infections during pregnancy have been linked to central nervous system development impairments and disorders such as autism. Changes in the structure and function of the respiratory, immune, and visceral systems have also been reported. SARS-CoV-2 infection during pregnancy has been linked with increased risks of stillbirth and preterm birth. However, the mechanisms involved remain unclear and may include cytokine storms, macrophage mediation, genetic mutations, methylation, and other epigenetic changes. Exploring the protective effects of antiviral treatment and other interventions in animal and clinical studies may help improve outcomes.
    CONCLUSIONS: SARS-CoV-2 infection during pregnancy activates the maternal-fetal immune interface through vertical transmission, and has short- and long-term effects on fetal development, including the central nervous system. Future long-term studies may help provide evidence that can inform interventions to reduce the risk of adverse outcomes.
    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
    阐明日本儿童中严重急性呼吸综合征冠状病毒2(SARS-CoV-2)的血清阳性率和无症状感染率,血清学分析使用从2020年3月至2023年2月收集的血清样本进行.在第一个研究期间(2020年3月至2021年2月)共收集了1493份血清样品。血清样品中没有SARS-CoV-2抗体阳性。在第二阶段(2021年3月至2022年2月),1055名患者中有7名(0.7%)经历了SARS-CoV-2感染。第三阶段(2022年3月至2023年2月)分为三个期限:2022年3月至6月30日;2022年7月至10月;以及2022年11月至2023年2月。在此期间血清阳性率逐渐增加,比率为6.0%,18.6%,在三项中占30.4%,分别。无症状的SARS-CoV-2感染的儿科病例发生在Omicron变体激增之后。由于SARS-CoV-2抗体阳性患者均无2019年冠状病毒病史,因此本研究中的血清阳性率可能代表无症状感染率。
    To elucidate the seroprevalence and rate of asymptomatic infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in Japanese children, serological analysis was performed using serum samples collected from March 2020 to February 2023. A total of 1493 serum samples were collected during the first study period (March 2020 to February 2021). None of the serum samples was positive for SARS-CoV-2 antibody. In the second period (March 2021 to February 2022), seven of the 1055 patients (0.7%) experienced SARS-CoV-2 infection. The third period (March 2022 to February 2023) was divided into three terms: from March to June 30, 2022; from July to October 2022; and from November 2022 to February 2023. The seroprevalence gradually increased throughout this period, with rates of 6.0%, 18.6%, and 30.4% in the three terms, respectively. Pediatric cases of asymptomatic SARS-CoV-2 infection occurred after the surge of Omicron variants. Since none of the SARS-CoV-2 antibody-positive patients had a previous history of coronavirus disease 2019, the seroprevalence rate in this study may represent the rate of asymptomatic 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
    由于需要高度的专业知识,业余爱好者经常难以检测和定量体液中的蛋白质生物标志物。本研究引入了一个实验室在小瓶(LV)快速诊断平台,具有绣球花状铂纳米酶(PtNH),为了快速,在15分钟内现场准确检测和定量蛋白质生物标志物。这种方法显着提高了体液中各种生物标志物的检测灵敏度,超过传统方法,例如酶联免疫吸附测定(ELISA)和侧流测定(LFA)约250至1300倍。LV平台使用涂有特定生物受体(如抗原或抗体)的玻璃小瓶。能够从少量液体样本中快速体外评估疾病风险,类似于个人ELISA样护理点测试(POCT)。它克服了现场生物标志物检测的挑战,允许通过用于医疗保健物联网(H-IoT)的便携式无线光谱仪进行检测和量化。使用SARS-CoV-2感染患者的IgG/IgM抗体和尿路上皮癌(UC)患者的核基质蛋白(NMP22)作为生物标志物,证实了该平台的有效性和适应性。这些测试证明了它的准确性和灵活性。这种方法为各种疾病的应用提供了巨大的潜力,前提是体液中的相关蛋白质生物标志物被鉴定。
    Amateurs often struggle with detecting and quantifying protein biomarkers in body fluids due to the high expertise required. This study introduces a Lab-in-a-Vial (LV) rapid diagnostic platform, featuring hydrangea-like platinum nanozymes (PtNH), for rapid, accurate detection and quantification of protein biomarkers on-site within 15 min. This method significantly enhances detection sensitivity for various biomarkers in body fluids, surpassing traditional methods such as enzyme-linked immunosorbent assays (ELISA) and lateral flow assays (LFA) by ≈250 to 1300 times. The LV platform uses a glass vial coated with specific bioreceptors such as antigens or antibodies, enabling rapid in vitro evaluation of disease risk from small fluid samples, similar to a personal ELISA-like point-of-care test (POCT). It overcomes challenges in on-site biomarker detection, allowing both detection and quantification through a portable wireless spectrometer for healthcare internet of things (H-IoT). The platform\'s effectiveness and adaptability are confirmed using IgG/IgM antibodies from SARS-CoV-2 infected patients and nuclear matrix protein (NMP22) from urothelial carcinoma (UC) patients as biomarkers. These tests demonstrated its accuracy and flexibility. This approach offers vast potential for diverse disease applications, provided that the relevant protein biomarkers in bodily fluids are identified.
    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