Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Infect Dis ; 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38382087

RESUMO

BACKGROUND: Rapidly evolving RNA viruses, such as human norovirus, generate extraordinary sequence diversity, posing a significant challenge to vaccine design. This diversity coupled with short-lasting natural immunity leads to re-infection throughout one's lifetime. How re-exposure shapes humoral immunity to future norovirus strains remains incompletely understood. METHODS: We profiled the antibody responses following two community gastroenteritis outbreaks with GII.2 and GII.6 noroviruses in 1971. Using diverse VLPs, ELISA, and carbohydrate-blocking assays (surrogate for neutralization), we examined the antibody response at acute and convalescent timepoints following GII.6 infection. RESULTS: Convalescent sera displayed strong homologous blocking, demonstrating a 5-fold increase in GII.6 carbohydrate-blockade over acute samples, and broad blocking of diverse archival and modern GII.6 noroviruses. Convalescent sera displayed limited carbohydrate-blocking of heterotypic VLPs, despite high ELISA binding titers. Select individuals developed broad cross-genotype blockade, but this response was established before the second outbreak. Finally, we applied a novel competitive carbohydrate-blocking assay to demonstrate the epitope-specificity and discrete compartments of the neutralizing response. CONCLUSIONS: Our data show that infection generates narrow, focused immunity directed towards the infecting genotype. We did detect broad cross-blocking in specific individuals, but these responses could be attributed to diverse, genotype-specific antibodies pre-dating GII.6 infection.

3.
J Virol ; 97(2): e0171622, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36688654

RESUMO

Norovirus is a major human pathogen that can cause severe gastroenteritis in vulnerable populations. The extensive viral diversity presented by human noroviruses constitutes a major roadblock for the development of effective vaccines. In addition to the large number of genotypes, antigenically distinct variants of GII.4 noroviruses have chronologically emerged over the last 3 decades. The last variant to emerge, Sydney_2012, has been circulating at high incidence worldwide for over a decade. We analyzed 1449 capsid sequences from GII.4 Sydney_2012 viruses to determine genetic changes indicative of antigenic diversification. Phylogenetic analyses show that Sydney_2012 viruses scattered within the tree topology with no single cluster dominating during a given year or geographical location. Fourteen residues presented high variability, 7 of which mapped to 4 antigenic sites. Notably, ~52% of viruses presented mutations at 2 or more antigenic sites. Mutational patterns showed that residues 297 and 372, which map to antigenic site A, changed over time. Virus-like particles (VLPs) developed from wild-type Sydney_2012 viruses and engineered to display all mutations detected at antigenic sites were tested against polyclonal sera and monoclonal antibodies raised against Sydney_2012 and Farmington_Hills_2002 VLPs. Minimal changes in reactivity were detected with polyclonal sera and only 4 MAbs lost binding, with all mapping to antigenic site A. Notably, reversion of residues from Sydney_2012 reconstituted epitopes from ancestral GII.4 variants. Overall, this study demonstrates that, despite circulating for over a decade, Sydney_2012 viruses present minimal antigenic diversification and provides novel insights on the diversification of GII.4 noroviruses that could inform vaccine design. IMPORTANCE GII.4 noroviruses are the major cause of acute gastroenteritis in all age groups. This predominance has been attributed to the continued emergence of phylogenetically discrete variants that escape immune responses to previous infections. The last GII.4 variant to emerge, Sydney_2012, has been circulating at high incidence for over a decade, raising the question of whether this variant is undergoing antigenic diversification without presenting a major distinction at the phylogenetic level. Sequence analyses that include >1400 capsid sequences from GII.4 Sydney_2012 showed changes in 4 out of the 6 major antigenic sites. Notably, while changes were detected in one of the most immunodominant sites over time, these resulted in minimal changes in the antigenic profile of these viruses. This study provides new insights on the mechanism governing the antigenic diversification of GII.4 norovirus that could help in the development of cross-protective vaccines to human noroviruses.


Assuntos
Antígenos Virais , Infecções por Caliciviridae , Norovirus , Humanos , Anticorpos Monoclonais/metabolismo , Infecções por Caliciviridae/virologia , Proteínas do Capsídeo/genética , Epitopos/genética , Gastroenterite/virologia , Genótipo , Norovirus/classificação , Norovirus/genética , Filogenia , Evolução Molecular , Antígenos Virais/genética
4.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33836574

RESUMO

Noroviruses are the predominant cause of acute gastroenteritis, with a single genotype (GII.4) responsible for the majority of infections. This prevalence is characterized by the periodic emergence of new variants that present substitutions at antigenic sites of the major structural protein (VP1), facilitating escape from herd immunity. Notably, the contribution of intravariant mutations to changes in antigenic properties is unknown. We performed a comprehensive antigenic analysis on a virus-like particle panel representing major chronological GII.4 variants to investigate diversification at the inter- and intravariant level. Immunoassays, neutralization data, and cartography analyses showed antigenic similarities between phylogenetically related variants, with major switches to antigenic properties observed over the evolution of GII.4 variants. Genetic analysis indicated that multiple coevolving amino acid changes-primarily at antigenic sites-are associated with the antigenic diversification of GII.4 variants. These data highlight complexities of the genetic determinants and provide a framework for the antigenic characterization of emerging GII.4 noroviruses.


Assuntos
Variação Antigênica , Antígenos Virais/genética , Infecções por Caliciviridae/virologia , Proteínas do Capsídeo/genética , Gastroenterite/virologia , Norovirus/genética , Substituição de Aminoácidos , Anticorpos Antivirais/imunologia , Antígenos Virais/química , Antígenos Virais/imunologia , Sítios de Ligação de Anticorpos , Infecções por Caliciviridae/epidemiologia , Proteínas do Capsídeo/classificação , Gastroenterite/epidemiologia , Humanos , Norovirus/classificação , Pandemias
5.
PLoS Pathog ; 17(7): e1009744, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34255807

RESUMO

Norovirus is a major cause of acute gastroenteritis worldwide. Over 30 different genotypes, mostly from genogroup I (GI) and II (GII), have been shown to infect humans. Despite three decades of genome sequencing, our understanding of the role of genomic diversification across continents and time is incomplete. To close the spatiotemporal gap of genomic information of human noroviruses, we conducted a large-scale genome-wide analyses that included the nearly full-length sequencing of 281 archival viruses circulating since the 1970s in over 10 countries from four continents, with a major emphasis on norovirus genotypes that are currently underrepresented in public genome databases. We provided new genome information for 24 distinct genotypes, including the oldest genome information from 12 norovirus genotypes. Analyses of this new genomic information, together with those publicly available, showed that (i) noroviruses evolve at similar rates across genomic regions and genotypes; (ii) emerging viruses evolved from transiently-circulating intermediate viruses; (iii) diversifying selection on the VP1 protein was recorded in genotypes with multiple variants; (iv) non-structural proteins showed a similar branching on their phylogenetic trees; and (v) contrary to the current understanding, there are restrictions on the ability to recombine different genomic regions, which results in co-circulating populations of viruses evolving independently in human communities. This study provides a comprehensive genetic analysis of diverse norovirus genotypes and the role of non-structural proteins on viral diversification, shedding new light on the mechanisms of norovirus evolution and transmission.


Assuntos
Genoma Viral/genética , Norovirus/genética , Evolução Biológica , Evolução Molecular , Estudo de Associação Genômica Ampla , Humanos
6.
Rev Med Virol ; 32(5): e2354, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35481689

RESUMO

Human noroviruses are the leading global cause of viral gastroenteritis. Attempts at developing effective vaccines and treatments against norovirus disease have been stymied by the extreme genetic diversity and rapid geographic distribution of these viruses. The emergence and replacement of predominantly circulating norovirus genotypes has primarily been attributed to mutations on the VP1 capsid protein leading to genetic drift, and more recently to recombination events between the ORF1/ORF2 junction. However, large-scale research into the historical and geographic distribution of recombinant norovirus strains has been limited in the literature. We performed a comprehensive historical analysis on 30,810 human norovirus sequences submitted to public databases between the years 1995 and 2019. During this time, 37 capsid genotypes and 56 polymerase types were detected across 90 different countries, and 97 unique recombinant genomes were also identified. GII.4, both capsid and polymerase, was the predominately circulating type worldwide for the majority of this time span, save for a brief swell of GII.17 and GII.2 capsid genotypes and a near-total eclipse by GII.P16, GII.P21 and GII.P31 beginning in 2013. Interestingly, an analysis of 4067 recombinants found that 50.2% (N = 2039) of all recorded sequences belonged to three recently emerged recombinant strains: GII.2[P16], GII.4[P31], and GII.4[P16]. This analysis should provide an important historical foundation for future studies that evaluate the emergence and distribution of noroviruses, as well as the design of cross-protective vaccines.


Assuntos
Infecções por Caliciviridae , Gastroenterite , Norovirus , Infecções por Caliciviridae/epidemiologia , Proteínas do Capsídeo/genética , Surtos de Doenças , Gastroenterite/epidemiologia , Genótipo , Humanos , Norovirus/genética , Filogenia
7.
J Infect Dis ; 225(7): 1205-1214, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32179892

RESUMO

Human noroviruses are the most common viral agents of acute gastroenteritis. Recently, human intestinal enteroids were shown to be permissive for norovirus infection. We tested their suitability as a system to study norovirus neutralization. Hyperimmune sera raised against virus-like particles (VLPs) representing different genotypes showed highly specific neutralization activity against GII.4 and GII.6 noroviruses. Carbohydrate blocking assays and neutralization exhibited similar patterns in antibody responses. Notably, sera produced against chimeric VLPs that presented swapped structural shell and protruding (P) domains, from different genotypes showed that neutralization is primarily mediated by antibodies mapping to the P domain of the norovirus capsid protein. This study provides empirical information on the antigenic differences among genotypes as measured by neutralization, which could guide vaccine design.


Assuntos
Anticorpos Neutralizantes , Infecções por Caliciviridae , Norovirus , Humanos , Anticorpos Antivirais , Proteínas do Capsídeo/imunologia , Gastroenterite/virologia , Genótipo , Norovirus/genética , Soros Imunes/imunologia
8.
Arch Virol ; 167(12): 2723-2727, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36087133

RESUMO

The genetic characterization of archival specimens is important for evaluating the evolutionary processes of noroviruses. Complete viral genome sequences, GVIII.1[GII.P28] and GIX.1[GII.P15], were determined from two archival specimens collected in Tokyo, Japan, in 1986 and 1995. In addition, complete VP1 and partial RdRp sequences of four samples collected between 1975 and 1983 were determined. Two viruses were classified as GI.5[P5] and GI.9[P9]; however, the viruses from the other two samples could not be assigned to any known genotypes using norovirus typing tools and phylogenetic analysis, suggesting that they might be untypable genotypes. Further evolutionary analysis of these viruses is warranted.


Assuntos
Infecções por Caliciviridae , Norovirus , Vírus , Humanos , Norovirus/genética , Filogenia , Genoma Viral , Genótipo , Vírus/genética
9.
Emerg Infect Dis ; 26(1): 157-159, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31855537

RESUMO

We report multiple nontypeable genotype II noroviruses circulating in South America; nucleotides differed by >25% from those of other genotypes. These viruses have been circulating in the Americas for ≈20 years and show recombination with other genotypes. Clues to norovirus natural history can guide development of treatment and prevention plans.


Assuntos
Norovirus/genética , América/epidemiologia , Infecções por Caliciviridae/epidemiologia , Infecções por Caliciviridae/virologia , Genótipo , Humanos , Filogenia , Recombinação Genética/genética
10.
J Gen Virol ; 99(8): 1027-1035, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29916802

RESUMO

Noroviruses are extremely diverse, with ≥30 genotypes infecting humans. GII genotype 4 (GII.4) noroviruses, the most prevalent genotype, present a constant accumulation of mutations on the major capsid protein (VP1), resulting in the chronological emergence of new variants every 2-8 years. On the other hand, non-GII.4 noroviruses present a limited number of changes on the capsid protein over time. Despite limited diversification, non-GII.4 viruses can also be associated with large outbreaks. To gain insights into the evolutionary dynamics of non-GII.4 viruses, we performed variant-specific phylogenetic analyses on a comprehensive dataset of 13 genotypes. Although the genotypes with a single variant presented a linear (clock-like) evolution, maximum-likelihood analyses revealed a lack of clock-like signals for the genotypes with ≥3 variants: GI.3, GII.6 and GII.17. Notably, the evolutionary pattern of non-GII.4 viruses showed clock-like signals when each variant was analysed separately. A minimal impact on the long-term clock-like evolution of VP1 was detected due to the exchange (recombination) of the polymerase types. The linear evolution, without replacement among variants, is explained by minimal changes at the protein level due to the higher ratio of synonymous compared to non-synonymous substitutions in their evolution. Taken together, these data indicate that (i) the variants of non-GII.4 noroviruses evolve and persist in the population independently, probably due to strong evolutionary constraints on VP1, and (ii) variant-specific analyses with robust sequence databases that cover long periods of surveillance are needed to limit the potential for misinterpretation of the evolutionary dynamics of non-GII.4 noroviruses.


Assuntos
Evolução Molecular , Variação Genética , Norovirus/genética , Genótipo , Humanos , Filogenia
11.
Appl Environ Microbiol ; 83(9)2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28213546

RESUMO

Sewage samples have been investigated to study the norovirus concentrations in sewage or the genotypes of noroviruses circulating in human populations. However, the statistical relationship between the concentration of the virus and the number of infected individuals and the clinical importance of genotypes or strains detected in sewage are unclear. In this study, we carried out both environmental and clinical surveillance of noroviruses for 3 years, 2013 to 2016. We performed cross-correlation analysis of the concentrations of norovirus GI or GII in sewage samples collected weekly and the reported number of gastroenteritis cases. Norovirus genotypes in sewage were also analyzed by pyrosequencing and compared with those identified in stool samples. The cross-correlation analysis found the peak coefficient (R = 0.51) at a lag of zero, indicating that the variation in the GII concentration, expressed as the log10 number of copies per milliliter, was coincident with that in the gastroenteritis cases. A total of 15 norovirus genotypes and up to 8 genotypes per sample were detected in sewage, which included all of the 13 genotypes identified in the stool samples except 2. GII.4 was most frequently detected in both sample types, followed by GII.17. Phylogenetic analysis revealed that a strain belonging to the GII.17 Kawasaki 2014 lineage had been introduced into the study area in the 2012-2013 season. An increase in GI.3 cases was observed in the 2015-2016 season, and sewage monitoring identified the presence of GI.3 in the previous season (2014-2015). Our results demonstrated that monitoring of noroviruses in sewage is useful for sensitive detection of epidemic variants in human populations.IMPORTANCE We obtained statistical evidence of the relationship between the variation in the norovirus GII concentration in sewage and that of gastroenteritis cases during the 3-year study period. Sewage sample analysis by a pyrosequencing approach enabled us to understand the temporal variation in the norovirus genotypes circulating in human populations. We found that a strain closely related to the GII.17 Kawasaki 2014 lineage had been introduced into the study area at least 1 year before its appearance and identification in clinical cases. A similar pattern was observed for GI.3; cases were reported in the 2015-2016 season, and closely related strains were found in sewage in the previous season. Our observation indicates that monitoring of noroviruses in sewage is useful for the rapid detection of an epidemic and is also sensitive enough to study the molecular epidemiology of noroviruses. Applying this approach to other enteric pathogens in sewage will enhance our understanding of their ecology.


Assuntos
Infecções por Caliciviridae/epidemiologia , Infecções por Caliciviridae/virologia , Monitoramento Ambiental , Genótipo , Norovirus/classificação , Norovirus/isolamento & purificação , Esgotos/virologia , Epidemias , Gastroenterite/epidemiologia , Gastroenterite/virologia , Humanos , Norovirus/genética , Carga Viral
12.
Emerg Infect Dis ; 22(5): 887-90, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27088593

RESUMO

During 2011-2013, a nationwide outbreak of chikungunya virus infection occurred in the Philippines. The Asian genotype was identified as the predominant genotype; sporadic cases of the East/Central/South African genotype were detected in Mindanao. Further monitoring is needed to define the transmission pattern of this virus in the Philippines.


Assuntos
Febre de Chikungunya/epidemiologia , Febre de Chikungunya/virologia , Vírus Chikungunya/classificação , Vírus Chikungunya/genética , Anticorpos Antivirais/imunologia , Febre de Chikungunya/diagnóstico , Febre de Chikungunya/história , Surtos de Doenças , Ensaio de Imunoadsorção Enzimática , Genes Virais , Genótipo , Geografia , História do Século XXI , Humanos , Imunoglobulina M/imunologia , Filipinas/epidemiologia , Filogenia , Filogeografia , Vigilância da População
13.
Virus Genes ; 50(1): 129-33, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25352228

RESUMO

Two novel G3P[4] rotavirus strains were detected from children with acute diarrhea in Sendai, Japan, identified as a G3-P[4]-I2-R2-C2-M2-A2-N2-T2-E2-H2 genotype constellation by whole-genome sequence analysis. The VP7 gene of the two strains displayed the highest nucleotide sequence identity (91 %) and showed a close genetic relationship (99 % bootstrap value) to an equine rotavirus reported in India. The other gene segments were related to human group A rotaviruses. This report suggests a possible reassortment event between human and equine rotaviruses.


Assuntos
RNA Viral/genética , Vírus Reordenados/genética , Vírus Reordenados/isolamento & purificação , Infecções por Rotavirus/virologia , Rotavirus/genética , Rotavirus/isolamento & purificação , Animais , Antígenos Virais/genética , Proteínas do Capsídeo/genética , Pré-Escolar , Análise por Conglomerados , Diarreia/epidemiologia , Diarreia/virologia , Feminino , Cavalos , Humanos , Lactente , Japão/epidemiologia , Masculino , Dados de Sequência Molecular , Filogenia , Infecções por Rotavirus/epidemiologia , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos
14.
BMC Public Health ; 14: 34, 2014 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-24423060

RESUMO

BACKGROUND: The Great East Japan Earthquake of magnitude 9.0 that struck on 11 March 2011 resulted in more than 18000 deaths or cases of missing persons. The large-scale tsunami that followed the earthquake devastated many coastal areas of the Tohoku region, including Miyagi Prefecture, and many residents of the tsunami-affected areas were compelled to reside in evacuation centres (ECs). In Japan, seasonal influenza epidemics usually occur between December and March. At the time of the Great East Japan Earthquake on 11 March 2011, influenza A (H3N2) was still circulating and there was a heightened concern regarding severe outbreaks due to influenza A (H3N2). METHODS: After local hospital staff and public health nurses detected influenza cases among the evacuees, an outbreak investigation was conducted in five ECs that had reported at least one influenza case from 23 March to 11 April 2011. Cases were confirmed by point-of-care tests and those residues were obtained and subjected to reverse transcription PCR and/or real time RT-PCR for sub-typing of influenza. RESULTS: There were 105 confirmed cases detected during the study period with a mean attack rate of 5.3% (range, 0.8%-11.1%). An epidemiological tree for two ECs demonstrated same-room and familial links that accounted for 88.5% of cases. The majority of cases occurred in those aged 15-64 years, who were likely to have engaged in search and rescue activities. No deaths were reported in this outbreak. Familial link accounted for on average 40.5% of influenza cases in two ECs and rooms where two or more cases were reported accounted for on average 85% in those ECs. A combination of preventative measures, including case cohorting, personal hygiene, wearing masks, and early detection and treatment, were implemented during the outbreak period. CONCLUSIONS: Influenza can cause outbreaks in a disaster setting when the disaster occurs during an epidemic influenza season. The transmission route is more likely to be associated with sharing room and space and with familial links. The importance of influenza surveillance and early treatments should be emphasized in EC settings for implementing preventive control measures.


Assuntos
Desastres , Surtos de Doenças , Terremotos , Vírus da Influenza A Subtipo H3N2/isolamento & purificação , Influenza Humana/epidemiologia , Adolescente , Adulto , Feminino , Humanos , Incidência , Japão/epidemiologia , Masculino , Pessoa de Meia-Idade , Trabalho de Resgate , Tsunamis
15.
Virus Genes ; 2013 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-24078044

RESUMO

Analyzing the evolutionary pattern of the influenza A(H1N1)pdm09 strain in different regions is important for understanding its diversification. We therefore conducted this study to elucidate the genetic variability and molecular evolution of the influenza A(H1N1)pdm09 strains that circulated during the 2009-2010 and 2010-2011 influenza seasons in Sendai, Japan. Nasopharyngeal swab specimens were collected from patients with influenza-like illnesses who visited outpatient clinics in Sendai City, Japan, from September 2009 to April 2011. A total of 75 isolates were selected from September 2009 to April 2011 to analyze the genetic changes in the entire hemagglutinin 1 (HA1) segment of the HA gene and the neuraminidase (NA) gene based on sequence analysis. Bayesian coalescent Markov chain Monte Carlo analyses of HA1 and NA gene sequences were performed for further analysis. High sequence identities were observed for HA1 and NA in influenza A(H1N1)pdm09, displaying 99.06 and 99.33 % nucleotide identities, respectively, with the A(H1N1)pdm09 vaccine strain A/California/07/2009. The substitution rates of nucleotides for HA1 in the 2009-2010 and 2010-2011 were 1.5 × 10-3 and 1.6 × 10-3 substitutions per site per year, respectively. Phylogenetic tree analysis demonstrated that Sendai isolates were clustered into global clade 7, which is characterized by an S203T mutation in the HA1 gene. Moreover, two distinct circulation clusters were present in the 2010-2011 season. Mutations were present in antigenic or receptor-binding domains of the HA1 segment, including A141V, S143G, S183P, S185T, and S203T. The Bayesian skyline plot model illustrated a steady rate for the maintenance of genetic diversity, followed by a slight increase in the later part of the 2010-2011 season. Selection analysis revealed that the HA1 (position 197) and NA (position 46) sites were under positive selection; however, no known mutation conferring resistance to NA inhibitors such as H275Y was observed. The effect on control of the influenza A(H1N1)pdm09 virus, including vaccine strain selection, requires continuous monitoring of the strain by genetic surveillance.

16.
mBio ; : e0217723, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37905910

RESUMO

Noroviruses are a major cause of acute gastroenteritis worldwide and can establish chronic infection in immunocompromised individuals. To investigate the mechanisms of norovirus evolution during chronic infection, we selected seven representative patients from a National Institutes of Health study cohort who sustained norovirus infection for periods ranging from 73 to 1,492 days. Six patients shed viruses belonging to a single genotype (GII.2[PNA], GII.4 New Orleans[P4], GII.4 Den Haag[P4], GII.3[P21], GII.6[P7], or GII.14[P7]) over the period examined, while one patient sequentially shed two genotypes (GII.6[P7] followed by GII.4 Sydney[P31]). Norovirus genomes from consecutive stool samples were sequenced at high resolution (>3,300 reads/nucleotide position) using the Illumina platform and subjected to bioinformatics analysis. Norovirus sequences could be resolved into one or more discrete clonal RNA genomes that persisted within these patients over time. Phylogenetic analyses inferred that clonal populations originated from a single founder virus and not by reinfection with community strains. Estimated evolutionary rates of clonal populations during persistent infection were similar to those of noroviruses from acute infection in the global database, suggesting that inherently higher RNA-dependent polymerase error rates were not associated with the ability to persist. The high-resolution analysis of norovirus diversity and evolution at the population level described here should allow a better understanding of adaptive mutations sustained during chronic infection. IMPORTANCE Noroviruses are an important cause of chronic diarrhea in patients with compromised immune systems. Presently, there are no effective therapies to clear the virus, which can persist for years in the intestinal tract. The goal of our study was to develop a better understanding of the norovirus strains that are associated with these long-term infections. With the remarkable diversity of norovirus strains detected in the immunocompromised patient cohort we studied, it appears that most, if not all, noroviruses circulating in nature may have the capacity to establish a chronic infection when a person is unable to mount an effective immune response. Our work is the most comprehensive genetic data set generated to date in which near full-length genomes from noroviruses associated with chronic infection were analyzed by high-resolution next-generation sequencing. Analysis of this data set led to our discovery that certain patients in our cohort were shedding noroviruses that could be subdivided into distinct haplotypes or populations of viruses that were co-evolving independently. The ability to track haplotypes of noroviruses during chronic infection will allow us to fine-tune our understanding of how the virus adapts and maintains itself in the human host, and how selective pressures such as antiviral drugs can affect these distinct populations.

17.
Plant Physiol ; 155(3): 1226-36, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21228101

RESUMO

Jasmonates are ubiquitously occurring plant growth regulators with high structural diversity that mediate numerous developmental processes and stress responses. We have recently identified 12-O-ß-D-glucopyranosyljasmonic acid as the bioactive metabolite, leaf-closing factor (LCF), which induced nyctinastic leaf closure of Samanea saman. We demonstrate that leaf closure of isolated Samanea pinnae is induced upon stereospecific recognition of (-)-LCF, but not by its enantiomer, (+)-ent-LCF, and that the nonglucosylated derivative, (-)-12-hydroxyjasmonic acid also displays weak activity. Similarly, rapid and cell type-specific shrinkage of extensor motor cell protoplasts was selectively initiated upon treatment with (-)-LCF, whereas flexor motor cell protoplasts did not respond. In these bioassays related to leaf movement, all other jasmonates tested were inactive, including jasmonic acid (JA) and the potent derivates JA-isoleucine and coronatine. By contrast, (-)-LCF and (-)-12-hydroxyjasmonic acid were completely inactive with respect to activation of typical JA responses, such as induction of JA-responsive genes LOX2 and OPCL1 in Arabidopsis (Arabidopsis thaliana) or accumulation of plant volatile organic compounds in S. saman and lima bean (Phaseolus lunatus), generally considered to be mediated by JA-isoleucine in a COI1-dependent fashion. Furthermore, application of selective inhibitors indicated that leaf movement in S. saman is mediated by rapid potassium fluxes initiated by opening of potassium-permeable channels. Collectively, our data point to the existence of at least two separate JA signaling pathways in S. saman and that 12-O-ß-D-glucopyranosyljasmonic acid exerts its leaf-closing activity through a mechanism independent of the COI1-JAZ module.


Assuntos
Ciclopentanos/farmacologia , Fabaceae/efeitos dos fármacos , Fabaceae/fisiologia , Glucosídeos/farmacologia , Oxilipinas/farmacologia , Folhas de Planta/efeitos dos fármacos , Folhas de Planta/fisiologia , Proteínas de Plantas/metabolismo , Ciclopentanos/química , Fabaceae/citologia , Fabaceae/genética , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Genes de Plantas/genética , Glucosídeos/química , Moduladores de Transporte de Membrana/farmacologia , Movimento/efeitos dos fármacos , Oxilipinas/química , Folhas de Planta/citologia , Proteínas de Plantas/genética , Regiões Promotoras Genéticas/genética , Protoplastos/citologia , Protoplastos/efeitos dos fármacos , Protoplastos/metabolismo , Compostos Orgânicos Voláteis/análise
18.
Cell Rep ; 39(2): 110689, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35417705

RESUMO

A paradigm of RNA viruses is their ability to mutate and escape from herd immunity. Because antibody responses are a major effector for viral immunity, antigenic sites are usually under strong diversifying pressure. Here, we use norovirus as a model to study mechanisms of antigenic diversification of non-enveloped, fast-evolving RNA viruses. We comprehensively characterize all variable antigenic sites involved in virus neutralization and find that single neutralizing monoclonal antibodies (mAbs) map to multiple antigenic sites of GII.4 norovirus. Interactions of multiple epitopes on the viral capsid surface provide a broad mAb-binding repertoire with a remarkable difference in the mAb-binding profiles and immunodominance hierarchy for two distantly related GII.4 variants. Time-ordered mutant viruses confirm a progressive change of antibody immunodominance along with point mutations during the process of norovirus evolution. Thus, in addition to point mutations, switches in immunodominance that redirect immune responses could facilitate immune escape in RNA viruses.


Assuntos
Infecções por Caliciviridae , Norovirus , Anticorpos Neutralizantes , Anticorpos Antivirais , Formação de Anticorpos , Infecções por Caliciviridae/genética , Proteínas do Capsídeo/química , Humanos , Norovirus/genética
19.
Front Immunol ; 13: 1040836, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36389818

RESUMO

Human noroviruses are the major viral cause of acute gastroenteritis around the world. Although norovirus symptoms are in most cases mild and self-limited, severe and prolonged symptoms can occur in the elderly and in immunocompromised individuals. Thus, there is a great need for the development of specific therapeutics that can help mitigate infection. In this study, we sought to characterize a panel of human monoclonal antibodies (mAbs; NORO-123, -115, -273A, -263, -315B, and -250B) that showed carbohydrate blocking activity against the current pandemic variant, GII.4 Sydney 2012. All antibodies tested showed potent neutralization against GII.4 Sydney virus in human intestinal enteroid culture. While all mAbs recognized only GII.4 viruses, they exhibited differential binding patterns against a panel of virus-like particles (VLPs) representing major and minor GII.4 variants spanning twenty-five years. Using mutant VLPs, we mapped five of the mAbs to variable antigenic sites A (NORO-123, -263, -315B, and -250B) or C (NORO-115) on the major capsid protein. Those mapping to the antigenic site A showed blocking activity against multiple variants dating back to 1987, with one mAb (NORO-123) showing reactivity to all variants tested. NORO-115, which maps to antigenic site C, showed reactivity against multiple variants due to the low susceptibility for mutations presented by naturally-occurring variants at the proposed binding site. Notably, we show that cross-blocking and neutralizing antibodies can be elicited against variable antigenic sites. These data provide new insights into norovirus immunity and suggest potential for the development of cross-protective vaccines and therapeutics.


Assuntos
Antígenos de Grupos Sanguíneos , Infecções por Caliciviridae , Norovirus , Humanos , Idoso , Norovirus/genética , Proteínas do Capsídeo/genética , Anticorpos Neutralizantes , Anticorpos Antivirais , Anticorpos Monoclonais
20.
Gut Microbes ; 13(1): 1-13, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33783322

RESUMO

Human noroviruses are the most common viral cause of acute gastroenteritis worldwide. Currently, there are no approved vaccines or specific therapeutics to treat the disease. Some obstacles delaying the development of a norovirus vaccine are: (i) the extreme diversity presented by noroviruses; (ii) our incomplete understanding of immunity to noroviruses; and (iii) the lack of a robust cell culture system or animal model for human noroviruses. Recent advances in in vitro cultivation of norovirus, novel approaches applied to viral genomics and immunity, and completion of vaccine trials and birth cohort studies have provided new information toward a better understanding of norovirus immunity. Here, we will discuss the complex relationship between norovirus diversity and correlates of protection for human noroviruses, and how this information could be used to guide the development of cross-protective vaccines.


Assuntos
Biodiversidade , Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/virologia , Imunidade , Norovirus/genética , Norovirus/imunologia , Vacinas , Animais , Coorte de Nascimento , Infecções por Caliciviridae/prevenção & controle , Predisposição Genética para Doença , Interações entre Hospedeiro e Microrganismos , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA