Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.083
Filter
1.
J Med Virol ; 96(5): e29657, 2024 May.
Article in English | MEDLINE | ID: mdl-38727035

ABSTRACT

The H1N1pdm09 virus has been a persistent threat to public health since the 2009 pandemic. Particularly, since the relaxation of COVID-19 pandemic mitigation measures, the influenza virus and SARS-CoV-2 have been concurrently prevalent worldwide. To determine the antigenic evolution pattern of H1N1pdm09 and develop preventive countermeasures, we collected influenza sequence data and immunological data to establish a new antigenic evolution analysis framework. A machine learning model (XGBoost, accuracy = 0.86, area under the receiver operating characteristic curve = 0.89) was constructed using epitopes, physicochemical properties, receptor binding sites, and glycosylation sites as features to predict the antigenic similarity relationships between influenza strains. An antigenic correlation network was constructed, and the Markov clustering algorithm was used to identify antigenic clusters. Subsequently, the antigenic evolution pattern of H1N1pdm09 was analyzed at the global and regional scales across three continents. We found that H1N1pdm09 evolved into around five antigenic clusters between 2009 and 2023 and that their antigenic evolution trajectories were characterized by cocirculation of multiple clusters, low-level persistence of former dominant clusters, and local heterogeneity of cluster circulations. Furthermore, compared with the seasonal H1N1 virus, the potential cluster-transition determining sites of H1N1pdm09 were restricted to epitopes Sa and Sb. This study demonstrated the effectiveness of machine learning methods for characterizing antigenic evolution of viruses, developed a specific model to rapidly identify H1N1pdm09 antigenic variants, and elucidated their evolutionary patterns. Our findings may provide valuable support for the implementation of effective surveillance strategies and targeted prevention efforts to mitigate the impact of H1N1pdm09.


Subject(s)
Antigens, Viral , Influenza A Virus, H1N1 Subtype , Influenza, Human , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Humans , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Influenza, Human/virology , Influenza, Human/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Machine Learning , Evolution, Molecular , Epitopes/genetics , Epitopes/immunology , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19/virology , COVID-19/immunology , Pandemics/prevention & control , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , SARS-CoV-2/genetics , SARS-CoV-2/immunology
2.
Nat Commun ; 15(1): 3833, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714654

ABSTRACT

Antigenic characterization of circulating influenza A virus (IAV) isolates is routinely assessed by using the hemagglutination inhibition (HI) assays for surveillance purposes. It is also used to determine the need for annual influenza vaccine updates as well as for pandemic preparedness. Performing antigenic characterization of IAV on a global scale is confronted with high costs, animal availability, and other practical challenges. Here we present a machine learning model that accurately predicts (normalized) outputs of HI assays involving circulating human IAV H3N2 viruses, using their hemagglutinin subunit 1 (HA1) sequences and associated metadata. Each season, the model learns an updated nonlinear mapping of genetic to antigenic changes using data from past seasons only. The model accurately distinguishes antigenic variants from non-variants and adaptively characterizes seasonal dynamics of HA1 sites having the strongest influence on antigenic change. Antigenic predictions produced by the model can aid influenza surveillance, public health management, and vaccine strain selection activities.


Subject(s)
Antigens, Viral , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H3N2 Subtype , Influenza, Human , Machine Learning , Seasons , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Humans , Influenza, Human/immunology , Influenza, Human/virology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Antigens, Viral/immunology , Antigens, Viral/genetics , Hemagglutination Inhibition Tests , Antigenic Variation/genetics , Influenza Vaccines/immunology
3.
J Med Virol ; 96(5): e29681, 2024 May.
Article in English | MEDLINE | ID: mdl-38773815

ABSTRACT

Rotavirus gastroenteritis is accountable for an estimated 128 500 deaths among children younger than 5 years worldwide, and the majority occur in low-income countries. Although the clinical trials of rotavirus vaccines in Bangladesh revealed a significant reduction of severe rotavirus disease by around 50%, the vaccines are not yet included in the routine immunization program. The present study was designed to provide data on rotavirus diarrhea with clinical profiles and genotypes before (2017-2019) and during the COVID-19 pandemic period (2020-2021). Fecal samples were collected from 2% of the diarrheal patients at icddr,b Dhaka hospital of all ages between January 2017 and December 2021 and were tested for VP6 rotavirus antigen using ELISA. The clinical manifestations such as fever, duration of diarrhea and hospitalization, number of stools, and dehydration and so on were collected from the surveillance database (n = 3127). Of the positive samples, 10% were randomly selected for genotyping using Sanger sequencing method. A total of 12 705 fecal samples were screened for rotavirus A antigen by enzyme immunoassay. Overall, 3369 (27%) were rotavirus antigen-positive, of whom children <2 years had the highest prevalence (88.6%). The risk of rotavirus A infection was 4.2 times higher in winter than in summer. Overall, G3P[8] was the most prominent genotype (45.3%), followed by G1P[8] (32.1%), G9P[8] (6.8%), and G2P[4] (6.1%). The other unusual combinations, such as G1P[4], G1P[6], G2P[6], G3P[4], G3P[6], and G9P[6], were also present. Genetic analysis on Bangladeshi strains revealed that the selection pressure (dN/dS) was estimated as <1. The number of hospital visits showed a 37% drop during the COVID-19 pandemic relative to the years before the pandemic. Conversely, there was a notable increase in the rate of rotavirus positivity during the pandemic (34%, p < 0.00) compared to the period before COVID-19 (23%). Among the various clinical symptoms, only the occurrence of watery stool significantly increased during the pandemic. The G2P[4] strain showed a sudden rise (19%) in 2020, which then declined in 2021. In the same year, G1P[8] was more prevalent than G3P[8] (40% vs. 38%, respectively). The remaining genotypes were negligible and did not exhibit much fluctuation. This study reveals that the rotavirus burden remained high during the COVID-19 prepandemic and pandemic in Bangladesh. Considering the lack of antigenic variations between the circulating and vaccine-targeted strains, integrating the vaccine into the national immunization program could reduce the prevalence of the disease, the number of hospitalizations, and the severity of cases.


Subject(s)
COVID-19 , Feces , Genotype , Rotavirus Infections , Rotavirus , Humans , Bangladesh/epidemiology , Rotavirus/genetics , Rotavirus/isolation & purification , Rotavirus/classification , Rotavirus Infections/epidemiology , Rotavirus Infections/virology , Child, Preschool , Infant , COVID-19/epidemiology , COVID-19/virology , COVID-19/prevention & control , Feces/virology , Female , Male , Child , Diarrhea/virology , Diarrhea/epidemiology , Adolescent , Adult , Antigens, Viral/genetics , Infant, Newborn , Gastroenteritis/epidemiology , Gastroenteritis/virology , Young Adult , Prevalence , SARS-CoV-2/genetics , SARS-CoV-2/classification , Middle Aged , Seasons
4.
Elife ; 122024 May 28.
Article in English | MEDLINE | ID: mdl-38805550

ABSTRACT

Human H3N2 influenza viruses are subject to rapid antigenic evolution which translates into frequent updates of the composition of seasonal influenza vaccines. Despite these updates, the effectiveness of influenza vaccines against H3N2-associated disease is suboptimal. Seasonal influenza vaccines primarily induce hemagglutinin-specific antibody responses. However, antibodies directed against influenza neuraminidase (NA) also contribute to protection. Here, we analysed the antigenic diversity of a panel of N2 NAs derived from human H3N2 viruses that circulated between 2009 and 2017. The antigenic breadth of these NAs was determined based on the NA inhibition (NAI) of a broad panel of ferret and mouse immune sera that were raised by infection and recombinant N2 NA immunisation. This assessment allowed us to distinguish at least four antigenic groups in the N2 NAs derived from human H3N2 viruses that circulated between 2009 and 2017. Computational analysis further revealed that the amino acid residues in N2 NA that have a major impact on susceptibility to NAI by immune sera are in proximity of the catalytic site. Finally, a machine learning method was developed that allowed to accurately predict the impact of mutations that are present in our N2 NA panel on NAI. These findings have important implications for the renewed interest to develop improved influenza vaccines based on the inclusion of a protective NA antigen formulation.


Two proteins, the hemagglutinin and the neuraminidase, protrude from the surface of the influenza virus. Their detection by the immune system allows the host organism to mount defences against the viral threat. The virus evolves in response to this pressure, which manifests as changes in the appearance of its hemagglutinin and neuraminidase. This process, known as antigenic drift, leads to the proteins evading detection. It is also why flu vaccines require frequent updates, as they rely on 'training' the immune system to recognise the most important strains in circulation ­ primarily by exposing it to appropriate versions of hemagglutinin. While the antigenic drift of hemagglutinin has been extensively studied, much less is known about how the neuraminidase accumulates mutations, and how these affect the immune response. To investigate this question, Catani et al. selected 43 genetically distant neuraminidases from human viral samples isolated between 2009 and 2017. Statistical analyses were applied to define their relatedness, revealing that a group of closely related neuraminidases predominated from 2009 to 2015, before they were being taken over by a second group. A third group, which was identified in viruses isolated in 2013, was remarkably close to the neuraminidase of strains that circulated in the late 1990s. The fourth and final group of neuraminidases was derived from influenza viruses that normally circulate in pigs but can also occasionally infect humans. Next, Catani et al. examined the immune response that these 43 neuraminidases could elicit in mice, as well as in ferrets ­ the animal most traditionally used in influenza research. This allowed them to pinpoint which changes in the neuraminidase sequences were important to escape recognition by the host. Data obtained from the two model species were comparable, suggesting that these experiments could be conducted on mice going forward, which are easier to work with than ferrets. Finally, Catani et al. used machine learning to build a computational model that could predict how strongly the immune system would respond to a specific neuraminidase variant. These findings could help guide the development of new vaccines that include neuraminidases tailored to best prime and train the immune system against a larger variety of strains. This may aid the development of 'supra-seasonal' vaccines that protect against a broad range of influenza viruses, reducing the need for yearly updates.


Subject(s)
Antigens, Viral , Ferrets , Influenza A Virus, H3N2 Subtype , Influenza, Human , Neuraminidase , Neuraminidase/immunology , Neuraminidase/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/enzymology , Humans , Animals , Antigens, Viral/immunology , Antigens, Viral/genetics , Mice , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza, Human/virology , Antibodies, Viral/immunology , Influenza Vaccines/immunology , Antigenic Variation , Viral Proteins/immunology , Viral Proteins/genetics , Viral Proteins/chemistry , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology
5.
Arch Virol ; 169(6): 131, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38819530

ABSTRACT

Noroviruses (NoVs) are the chief cause of acute viral gastroenteritis worldwide. By employing the major capsid protein VP1 of a GII.6 NoV strain as an immunogen, we generated two monoclonal antibodies (mAbs) with wide-spectrum binding activities against NoV genogroup II (GII) VP1 proteins. One mAb (10G7) could bind to native and denatured GII-specific VP1 proteins. The other mAb (10F2) could bind to all tested native GII VP1 proteins, but not to denatured GII.3, GII.4, GII.7, or GII.17 VP1 proteins. Using GII.6/GII.4 fusion proteins, the mAb 10F2 binding region was confirmed to be located in the C-terminal P1 domain. An enzyme-linked immunosorbent assay based on peptides covering the P domain did not detect any binding. Using a panel of VP1 proteins with swapped regions, deletions, and mutations, the mAb 10F2 binding region was determined to be located between residues 496 and 513. However, the residue(s) responsible for its varied binding affinity for different denatured GII VP1 proteins remain to be identified. In summary, two NoV GII-specific cross-reactive mAbs were generated, and their binding regions were determined. Our results might facilitate the detection and immunogenic study of NoVs.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Capsid Proteins , Epitopes , Norovirus , Norovirus/genetics , Norovirus/immunology , Antibodies, Monoclonal/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Capsid Proteins/chemistry , Epitopes/immunology , Epitopes/genetics , Antibodies, Viral/immunology , Animals , Antigens, Viral/immunology , Antigens, Viral/genetics , Mice , Humans , Caliciviridae Infections/virology , Caliciviridae Infections/immunology , Mice, Inbred BALB C , Epitope Mapping , Cross Reactions
6.
J Nanobiotechnology ; 22(1): 295, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38807131

ABSTRACT

The signal sequence played a crucial role in the efficacy of mRNA vaccines against virus pandemic by influencing antigen translation. However, limited research had been conducted to compare and analyze the specific mechanisms involved. In this study, a novel approach was introduced by substituting the signal sequence of the mRNA antigen to enhance its immune response. Computational simulations demonstrated that various signal peptides differed in their binding capacities with the signal recognition particle (SRP) 54 M subunit, which positively correlated with antigen translation efficiency. Our data revealed that the signal sequences of tPA and IL-6-modified receptor binding domain (RBD) mRNA vaccines sequentially led to higher antigen expression and elicited more robust humoral and cellular immune protection against the SARS-CoV-2 compared to the original signal sequence. By highlighting the importance of the signal sequence, this research provided a foundational and safe approach for ongoing modifications in signal sequence-antigen design, aiming to optimize the efficacy of mRNA vaccines.


Subject(s)
Protein Sorting Signals , SARS-CoV-2 , mRNA Vaccines , Animals , Mice , SARS-CoV-2/immunology , COVID-19/prevention & control , COVID-19/immunology , Mice, Inbred BALB C , RNA, Messenger/genetics , COVID-19 Vaccines/immunology , Female , Humans , Antigens, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/chemistry , Antibodies, Viral/immunology , Immunity, Humoral , Vaccines, Synthetic/immunology , Immunity, Cellular
7.
Viruses ; 16(5)2024 04 24.
Article in English | MEDLINE | ID: mdl-38793544

ABSTRACT

The continuing mutability of the SARS-CoV-2 virus can result in failures of diagnostic assays. To address this, we describe a generalizable bioinformatics-to-biology pipeline developed for the calibration and quality assurance of inactivated SARS-CoV-2 variant panels provided to Radical Acceleration of Diagnostics programs (RADx)-radical program awardees. A heuristic genetic analysis based on variant-defining mutations demonstrated the lowest genetic variance in the Nucleocapsid protein (Np)-C-terminal domain (CTD) across all SARS-CoV-2 variants. We then employed the Shannon entropy method on (Np) sequences collected from the major variants, verifying the CTD with lower entropy (less prone to mutations) than other Np regions. Polyclonal and monoclonal antibodies were raised against this target CTD antigen and used to develop an Enzyme-linked immunoassay (ELISA) test for SARS-CoV-2. Blinded Viral Quality Assurance (VQA) panels comprised of UV-inactivated SARS-CoV-2 variants (XBB.1.5, BF.7, BA.1, B.1.617.2, and WA1) and distractor respiratory viruses (CoV 229E, CoV OC43, RSV A2, RSV B, IAV H1N1, and IBV) were assembled by the RADx-rad Diagnostics core and tested using the ELISA described here. The assay tested positive for all variants with high sensitivity (limit of detection: 1.72-8.78 ng/mL) and negative for the distractor virus panel. Epitope mapping for the monoclonal antibodies identified a 20 amino acid antigenic peptide on the Np-CTD that an in-silico program also predicted for the highest antigenicity. This work provides a template for a bioinformatics pipeline to select genetic regions with a low propensity for mutation (low Shannon entropy) to develop robust 'pan-variant' antigen-based assays for viruses prone to high mutational rates.


Subject(s)
Antigens, Viral , COVID-19 , Coronavirus Nucleocapsid Proteins , Phosphoproteins , SARS-CoV-2 , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Humans , Coronavirus Nucleocapsid Proteins/immunology , Coronavirus Nucleocapsid Proteins/genetics , COVID-19/diagnosis , COVID-19/immunology , COVID-19/virology , Antigens, Viral/immunology , Antigens, Viral/genetics , Phosphoproteins/immunology , Phosphoproteins/genetics , Enzyme-Linked Immunosorbent Assay/methods , Enzyme-Linked Immunosorbent Assay/standards , COVID-19 Serological Testing/methods , COVID-19 Serological Testing/standards , Antibodies, Viral/immunology , Antibodies, Monoclonal/immunology , Computational Biology/methods , Mutation , Animals
8.
Emerg Microbes Infect ; 13(1): 2343910, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38618740

ABSTRACT

Japanese encephalitis (JE), caused by the Japanese encephalitis virus (JEV), is a highly threatening disease with no specific treatment. Fortunately, the development of vaccines has enabled effective defense against JE. However, re-emerging genotype V (GV) JEV poses a challenge as current vaccines are genotype III (GIII)-based and provide suboptimal protection. Given the isolation of GV JEVs from Malaysia, China, and the Republic of Korea, there is a concern about the potential for a broader outbreak. Under the hypothesis that a GV-based vaccine is necessary for effective defense against GV JEV, we developed a pentameric recombinant antigen using cholera toxin B as a scaffold and mucosal adjuvant, which was conjugated with the E protein domain III of GV by genetic fusion. This GV-based vaccine antigen induced a more effective immune response in mice against GV JEV isolates compared to GIII-based antigen and efficiently protected animals from lethal challenges. Furthermore, a bivalent vaccine approach, inoculating simultaneously with GIII- and GV-based antigens, showed protective efficacy against both GIII and GV JEVs. This strategy presents a promising avenue for comprehensive protection in regions facing the threat of diverse JEV genotypes, including both prevalent GIII and GI as well as emerging GV strains.


Subject(s)
Encephalitis Virus, Japanese , Encephalitis, Japanese , Genotype , Japanese Encephalitis Vaccines , Encephalitis Virus, Japanese/genetics , Encephalitis Virus, Japanese/immunology , Encephalitis Virus, Japanese/classification , Animals , Encephalitis, Japanese/prevention & control , Encephalitis, Japanese/immunology , Encephalitis, Japanese/virology , Japanese Encephalitis Vaccines/immunology , Japanese Encephalitis Vaccines/administration & dosage , Japanese Encephalitis Vaccines/genetics , Mice , Antibodies, Viral/immunology , Antibodies, Viral/blood , Humans , Mice, Inbred BALB C , Female , Antigens, Viral/immunology , Antigens, Viral/genetics , Vaccine Efficacy , Cholera Toxin/genetics , Cholera Toxin/immunology
9.
Emerg Infect Dis ; 30(5): 1026-1029, 2024 May.
Article in English | MEDLINE | ID: mdl-38666659

ABSTRACT

Norovirus is a major cause of acute gastroenteritis; GII.4 is the predominant strain in humans. Recently, 2 new GII.4 variants, Hong Kong 2019 and San Francisco 2017, were reported. Characterization using GII.4 monoclonal antibodies and serum demonstrated different antigenic profiles for the new variants compared with historical variants.


Subject(s)
Antigens, Viral , Caliciviridae Infections , Gastroenteritis , Norovirus , Humans , Norovirus/genetics , Norovirus/immunology , Norovirus/classification , Hong Kong/epidemiology , Caliciviridae Infections/virology , Caliciviridae Infections/epidemiology , Caliciviridae Infections/immunology , Gastroenteritis/virology , Gastroenteritis/epidemiology , Antigens, Viral/immunology , Antigens, Viral/genetics , San Francisco/epidemiology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Genotype , Phylogeny , Antibodies, Monoclonal/immunology
10.
Cell Host Microbe ; 32(5): 755-767.e4, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38653242

ABSTRACT

Kaposi sarcoma (KS) is the most common cancer in persons living with HIV. It is caused by KS-associated herpesvirus (KSHV). There exists no animal model for KS. Pronuclear injection of the 170,000-bp viral genome induces early-onset, aggressive angiosarcoma in transgenic mice. The tumors are histopathologically indistinguishable from human KS. As in human KS, all tumor cells express the viral latency-associated nuclear antigen (LANA). The tumors transcribe most viral genes, whereas endothelial cells in other organs only transcribe the viral latent genes. The tumor cells are of endothelial lineage and exhibit the same molecular pattern of pathway activation as KS, namely phosphatidylinositol 3-kinase (PI3K)/Akt/mTOR, interleukin-10 (IL-10), and vascular endothelial growth factor (VEGF). The KSHV-induced tumors are more aggressive than Ha-ras-induced angiosarcomas. Overall survival is increased by prophylactic ganciclovir. Thus, whole-virus KSHV-transgenic mice represent an accurate model for KS and open the door for the genetic dissection of KS pathogenesis and evaluation of therapies, including vaccines.


Subject(s)
Disease Models, Animal , Hemangiosarcoma , Herpesvirus 8, Human , Mice, Transgenic , Sarcoma, Kaposi , Animals , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/pathogenicity , Mice , Hemangiosarcoma/virology , Hemangiosarcoma/genetics , Hemangiosarcoma/pathology , Sarcoma, Kaposi/virology , Sarcoma, Kaposi/pathology , Genome, Viral , Humans , Antigens, Viral/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Ganciclovir/therapeutic use , Ganciclovir/pharmacology , Interleukin-10/genetics
11.
Emerg Microbes Infect ; 13(1): 2343909, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38616729

ABSTRACT

The recent emergence of a SARS-CoV-2 saltation variant, BA.2.87.1, which features 65 spike mutations relative to BA.2, has attracted worldwide attention. In this study, we elucidate the antigenic characteristics and immune evasion capability of BA.2.87.1. Our findings reveal that BA.2.87.1 is more susceptible to XBB-induced humoral immunity compared to JN.1. Notably, BA.2.87.1 lacks critical escaping mutations in the receptor binding domain (RBD) thus allowing various classes of neutralizing antibodies (NAbs) that were escaped by XBB or BA.2.86 subvariants to neutralize BA.2.87.1, although the deletions in the N-terminal domain (NTD), specifically 15-23del and 136-146del, compensate for the resistance to humoral immunity. Interestingly, several neutralizing antibody drugs have been found to restore their efficacy against BA.2.87.1, including SA58, REGN-10933 and COV2-2196. Hence, our results suggest that BA.2.87.1 may not become widespread until it acquires multiple RBD mutations to achieve sufficient immune evasion comparable to that of JN.1.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 , Immune Evasion , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Antibodies, Neutralizing/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , COVID-19/immunology , COVID-19/virology , Antibodies, Viral/immunology , Humans , Mutation , Animals , Antigens, Viral/immunology , Antigens, Viral/genetics , Immunity, Humoral
12.
Viruses ; 16(4)2024 04 05.
Article in English | MEDLINE | ID: mdl-38675907

ABSTRACT

Rotavirus A (RVA) is the leading cause of diarrhea requiring hospitalization in children and causes over 100,000 annual deaths in Sub-Saharan Africa. In order to generate next-generation vaccines against African RVA genotypes, a reverse genetics system based on a simian rotavirus strain was utilized here to exchange the antigenic capsid proteins VP4, VP7 and VP6 with those of African human rotavirus field strains. One VP4/VP7/VP6 (genotypes G9-P[6]-I2) triple-reassortant was successfully rescued, but it replicated poorly in the first cell culture passages. However, the viral titer was enhanced upon further passaging. Whole genome sequencing of the passaged virus revealed a single point mutation (A797G), resulting in an amino acid exchange (E263G) in VP4. After introducing this mutation into the VP4-encoding plasmid, a VP4 mono-reassortant as well as the VP4/VP7/VP6 triple-reassortant replicated to high titers already in the first cell culture passage. However, the introduction of the same mutation into the VP4 of other human RVA strains did not improve the rescue of those reassortants, indicating strain specificity. The results show that specific point mutations in VP4 can substantially improve the rescue and replication of recombinant RVA reassortants in cell culture, which may be useful for the development of novel vaccine strains.


Subject(s)
Capsid Proteins , Reassortant Viruses , Rotavirus , Virus Replication , Rotavirus/genetics , Capsid Proteins/genetics , Humans , Reassortant Viruses/genetics , Animals , Mutation , Cell Line , Reverse Genetics/methods , Genotype , Point Mutation , Rotavirus Infections/virology , Genome, Viral , Antigens, Viral/genetics , Antigens, Viral/immunology
13.
Virol J ; 21(1): 94, 2024 04 24.
Article in English | MEDLINE | ID: mdl-38659036

ABSTRACT

BACKGROUND: The causative agents of diarrhea, rotavirus B (RVB) and rotavirus C (RVC) are common in adults and patients of all age groups, respectively. Due to the Rotavirus A (RVA) vaccination program, a significant decrease in the number of gastroenteritis cases has been observed globally. The replacement of RVA infections with RVB, RVC, or other related serogroups is suspected due to the possibility of reducing natural selective constraints due to RVA infections. The data available on RVB and RVC incidence are scant due to the lack of cheap and rapid commercial diagnostic assays and the focus on RVA infections. The present study aimed to develop real-time RT‒PCR assays using the data from all genomic RNA segments of human RVB and RVC strains available in the Gene Bank. RESULTS: Among the 11 gene segments, NSP3 and NSP5 of RVB and the VP6 gene of RVC were found to be suitable for real-time RT‒PCR (qRT‒PCR) assays. Fecal specimens collected from diarrheal patients were tested simultaneously for the presence of RVB (n = 192) and RVC (n = 188) using the respective conventional RT‒PCR and newly developed qRT‒PCR assays. All RVB- and RVC-positive specimens were reactive in their respective qRT‒PCR assays and had Ct values ranging between 23.69 and 41.97 and 11.49 and 36.05, respectively. All known positive and negative specimens for other viral agents were nonreactive, and comparative analysis showed 100% concordance with conventional RT‒PCR assays. CONCLUSIONS: The suitability of the NSP5 gene of RVB and the VP6 gene of RVC was verified via qRT‒PCR assays, which showed 100% sensitivity and specificity. The rapid qRT‒PCR assays developed will be useful diagnostic tools, especially during diarrheal outbreaks for testing non-RVA rotaviral agents and reducing the unnecessary use of antibiotics.


Subject(s)
Diarrhea , Feces , Real-Time Polymerase Chain Reaction , Rotavirus Infections , Rotavirus , Rotavirus/genetics , Rotavirus/isolation & purification , Humans , Rotavirus Infections/virology , Rotavirus Infections/diagnosis , Real-Time Polymerase Chain Reaction/methods , Feces/virology , Diarrhea/virology , Diarrhea/diagnosis , Sensitivity and Specificity , Reverse Transcriptase Polymerase Chain Reaction/methods , Viral Nonstructural Proteins/genetics , Antigens, Viral/genetics , RNA, Viral/genetics , Capsid Proteins/genetics , Genome, Viral/genetics , Gastroenteritis/virology , Gastroenteritis/diagnosis
14.
15.
Mamm Genome ; 35(2): 296-307, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38600211

ABSTRACT

Varicella-zoster virus (VZV), a common pathogen with humans as the sole host, causes primary infection and undergoes a latent period in sensory ganglia. The recurrence of VZV is often accompanied by severe neuralgia in skin tissue, which has a serious impact on the life of patients. During the acute infection of VZV, there are few related studies on the pathophysiological mechanism of skin tissue. In this study, transcriptome sequencing data from the acute response period within 2 days of VZV antigen stimulation of the skin were used to explore a model of the trajectory of skin tissue changes during VZV infection. It was found that early VZV antigen stimulation caused activation of mainly natural immune-related signaling pathways, while in the late phase activation of mainly active immune-related signaling pathways. JAK-STAT, NFκB, and TNFα signaling pathways are gradually activated with the progression of infection, while Hypoxia is progressively inhibited. In addition, we found that dendritic cell-mediated immune responses play a dominant role in the lesion damage caused by VZV antigen stimulation of the skin. This study provides a theoretical basis for the study of the molecular mechanisms of skin lesions during acute VZV infection.


Subject(s)
Herpesvirus 3, Human , Signal Transduction , Skin , Varicella Zoster Virus Infection , Herpesvirus 3, Human/genetics , Skin/pathology , Skin/virology , Skin/immunology , Animals , Varicella Zoster Virus Infection/virology , Varicella Zoster Virus Infection/immunology , Varicella Zoster Virus Infection/genetics , Varicella Zoster Virus Infection/pathology , Humans , Mice , Dendritic Cells/immunology , Herpes Zoster/virology , Herpes Zoster/pathology , Herpes Zoster/genetics , Herpes Zoster/immunology , Transcriptome , Disease Models, Animal , Antigens, Viral/immunology , Antigens, Viral/genetics , NF-kappa B/metabolism , NF-kappa B/genetics
16.
Biosci Rep ; 44(5)2024 May 29.
Article in English | MEDLINE | ID: mdl-38592735

ABSTRACT

The rotavirus capsid protein VP6 forms the middle of three protein layers and is responsible for many critical steps in the viral life cycle. VP6 as a structural protein can be used in various applications including as a subunit vaccine component. The head domain of VP6 (VP6H) contains key sequences that allow the protein to trimerize and that represent epitopes that are recognized by human antibodies in the viral particle. The domain is rich in ß-sheet secondary structures. Here, VP6H was solubilised from bacterial inclusion bodies and purified using a single affinity chromatography step. Spectral (far-UV circular dichroism and intrinsic tryptophan fluorescence) analysis revealed that the purified domain had native-like secondary and tertiary structures. The domain could maintain structure up to 44°C during thermal denaturation following which structural changes result in an intermediate forming and finally irreversible aggregation and denaturation. The chemical denaturation with urea and guanidinium hydrochloride produces intermediates that represent a loss in the cooperativity. The VP6H domain is stable and can fold to produce its native structure in the absence of the VP6 base domain but cannot be defined as an independent folding unit.


Subject(s)
Antigens, Viral , Capsid Proteins , Rotavirus , Capsid Proteins/chemistry , Capsid Proteins/genetics , Antigens, Viral/chemistry , Antigens, Viral/genetics , Rotavirus/chemistry , Protein Denaturation , Protein Domains , Circular Dichroism , Protein Folding , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
17.
Microb Pathog ; 190: 106630, 2024 May.
Article in English | MEDLINE | ID: mdl-38556102

ABSTRACT

Porcine circovirus type 2 (PCV2) is a globally prevalent infectious pathogen affecting swine, with its capsid protein (Cap) being the sole structural protein critical for vaccine development. Prior research has demonstrated that PCV2 Cap proteins produced in Escherichia coli (E. coli) can form virus-like particles (VLPs) in vitro, and nuclear localization signal peptides (NLS) play a pivotal role in stabilizing PCV2 VLPs. Recently, PCV2d has emerged as an important strain within the PCV2 epidemic. In this study, we systematically optimized the PCV2d Cap protein and successfully produced intact PCV2d VLPs containing NLS using E. coli. The recombinant PCV2d Cap protein was purified through affinity chromatography, yielding 7.5 mg of recombinant protein per 100 ml of bacterial culture. We augmented the conventional buffer system with various substances such as arginine, ß-mercaptoethanol, glycerol, polyethylene glycol, and glutathione to promote VLP assembly. The recombinant PCV2d Cap self-assembled into VLPs approximately 20 nm in diameter, featuring uniform distribution and exceptional stability in the optimized buffer. We developed the vaccine and immunized pigs and mice, evaluating the immunogenicity of the PCV2d VLPs vaccine by measuring PCV2-IgG, IL-4, TNF-α, and IFN-γ levels, comparing them to commercial vaccines utilizing truncated PCV2 Cap antigens. The HE staining and immunohistochemical tests confirmed that the PCV2 VLPs vaccine offered robust protection. The results revealed that animals vaccinated with the PCV2d VLPs vaccine exhibited high levels of PCV2 antibodies, with TNF-α and IFN-γ levels rapidly increasing at 14 days post-immunization, which were higher than those observed in commercially available vaccines, particularly in the mouse trial. This could be due to the fact that full-length Cap proteins can assemble into more stable PCV2d VLPs in the assembling buffer. In conclusion, our produced PCV2d VLPs vaccine elicited stronger immune responses in pigs and mice compared to commercial vaccines. The PCV2d VLPs from this study serve as an excellent candidate vaccine antigen, providing insights for PCV2d vaccine research.


Subject(s)
Antibodies, Viral , Capsid Proteins , Circovirus , Escherichia coli , Recombinant Proteins , Vaccines, Virus-Like Particle , Animals , Circovirus/immunology , Circovirus/genetics , Swine , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/genetics , Capsid Proteins/immunology , Capsid Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Mice , Antibodies, Viral/immunology , Antibodies, Viral/blood , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Circoviridae Infections/prevention & control , Circoviridae Infections/immunology , Swine Diseases/prevention & control , Viral Vaccines/immunology , Viral Vaccines/genetics , Vaccine Development , Antigens, Viral/immunology , Antigens, Viral/genetics , Immunoglobulin G/blood , Cost-Benefit Analysis , Female , Interferon-gamma/metabolism , Immunogenicity, Vaccine
18.
J Appl Microbiol ; 135(3)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38471668

ABSTRACT

AIMS: Enteroviruses are significant human pathogens associated with a range of mild to severe diseases. This study aims to understand the diversity and genetic characterization of enteroviruses circulated in southwest China's border cities by using environmental surveillance. METHODS AND RESULTS: A total of 96 sewage samples were collected in three border cities and a port located in Yunnan Province, China from July 2020 to June 2022. After cell culture and VP1 sequencing, a total of 590 enterovirus isolates were identified, belonging to 21 types. All PV strains were Sabin-like with ≤6 nucleotide mutations in the VP1 coding region. Echovirus 6, echovirus 21 (a rare serotype in previous studies), and coxsackievirus B5 were the predominant serotypes, which accounted for 21.19%, 18.31%, and 13.39% of the total isolates, respectively. The prevalence of the common serotypes varied across different border cities and periods. Phylogenetic analysis revealed the presence of multiple evolutionary lineages for E21, E6, and E30, some of which formed distinct branches. CONCLUSIONS: High diversity of enteroviruses and distinct lineages of predominant serotypes circulated in southwest China's border cities.


Subject(s)
Enterovirus Infections , Enterovirus , Humans , Cities , Phylogeny , China/epidemiology , Enterovirus Infections/epidemiology , Enterovirus B, Human/genetics , Antigens, Viral/genetics , Environmental Monitoring/methods
19.
Viruses ; 16(3)2024 03 20.
Article in English | MEDLINE | ID: mdl-38543842

ABSTRACT

Coxsackievirus A6 (CV-A6) has emerged as the predominant causative agent of hand, foot, and mouth disease (HFMD) in young children. Since the declaration of coronavirus disease 2019 (COVID-19) as a global pandemic, the incidence of infectious diseases, including HFMD, has decreased markedly. When social mitigation was relaxed during the COVID-19 pandemic in 2022, the re-emergence of HFMD was observed in Gwangju, South Korea, and seasonal characteristics of the disease appeared to have changed. To investigate the molecular characteristics of enterovirus (EV) associated with HFMD during 2022, 277 specimens were collected. Children aged younger than 5 years accounted for the majority of affected individuals. EV detection and genotyping were performed using real-time RT-PCR and nested RT-PCR followed by sequence analysis. The EV detection rate was found to be 82.3%, and the main genotype identified was CV-A6. Sixteen CV-A6 samples were selected for whole genome sequencing. According to phylogenetic analysis, all CV-A6 strains from this study belonged to the sub-genotype D3 clade based on VP1 sequences. Analysis of 3D polymerase phylogeny showed that only the recombinant RF-A group was identified. In conclusion, circulating EV types should be continuously monitored to understand pathogen emergence and evolution during the post-pandemic era.


Subject(s)
Enterovirus , Hand, Foot and Mouth Disease , Child , Humans , Child, Preschool , Hand, Foot and Mouth Disease/epidemiology , Phylogeny , Pandemics , Enterovirus/genetics , Antigens, Viral/genetics , Real-Time Polymerase Chain Reaction , Genotype , China/epidemiology
20.
Cell Rep ; 43(3): 113888, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38416644

ABSTRACT

Higher-order genome structure influences the transcriptional regulation of cellular genes through the juxtaposition of regulatory elements, such as enhancers, close to promoters of target genes. While enhancer activation has emerged as an important facet of Kaposi sarcoma-associated herpesvirus (KSHV) biology, the mechanisms controlling enhancer-target gene expression remain obscure. Here, we discover that the KSHV genome tethering protein latency-associated nuclear antigen (LANA) potentiates enhancer-target gene expression in primary effusion lymphoma (PEL), a highly aggressive B cell lymphoma causally associated with KSHV. Genome-wide analyses demonstrate increased levels of enhancer RNA transcription as well as activating chromatin marks at LANA-bound enhancers. 3D genome conformation analyses identified genes critical for latency and tumorigenesis as targets of LANA-occupied enhancers, and LANA depletion results in their downregulation. These findings reveal a mechanism in enhancer-gene coordination and describe a role through which the main KSHV tethering protein regulates essential gene expression in PEL.


Subject(s)
Herpesvirus 8, Human , Sarcoma, Kaposi , Humans , Herpesvirus 8, Human/physiology , Genome-Wide Association Study , Antigens, Viral/genetics , Antigens, Viral/metabolism , Promoter Regions, Genetic/genetics , Gene Expression Regulation , Virus Latency
SELECTION OF CITATIONS
SEARCH DETAIL
...