Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 542
Filtrar
1.
Food Res Int ; 194: 114877, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39232515

RESUMEN

Human norovirus (HuNoV), the leading cause of foodborne acute gastroenteritis, poses a serious threat to public health. Traditional disinfection methods lead to destructions of food properties and functions, and/or environmental contaminations. Green and efficient approaches are urgently needed to disinfect HuNoV. Plasma-activated water (PAW) containing amounts of reactive species is an emerging nonthermal and eco-friendly disinfectant towards the pathogenic microorganisms. However, the disinfection efficacy and mechanism of PAW on HuNoV has not yet been studied. Murine norovirus 1 (MNV-1) is one of the most commonly used HuNoV surrogates to evaluate the efficacy of disinfectants. In the current study, the inactivation efficacy of MNV-1 by PAW was investigated. The results demonstrated that PAW significantly inactivated MNV-1, reducing the viral titer from approximately 6 log10 TCID50/mL to non-detectable level. The decreased pH, increased oxidation-reduction potential (ORP) and conductivity of PAW were observed compared with that of deionized water. Compositional analysis revealed that hydrogen peroxide (H2O2), nitrate (NO3-) and hydroxyl radical (OH) were the functional reactive species in MNV-1 inactivation. L-histidine could scavenge most of the inactivation effect in a concentration-dependent manner. Moreover, PAW could induce damage to viral proteins. Part of MNV-1 particles was destroyed, while others were structurally intact without infectiousness. After 45 days of storage at 4 °C, PAW generated with 80 % O2 and 100 % O2 could still reduce over 4 log10 TCID50/mL of the viral titer. In addition, PAW prepared using hard water induced approximately 6 log10 TCID50/mL reduction of MNV-1. PAW treatment of MNV-1-inoculated blueberries reduced the viral titer from 3.79 log10 TCID50/mL to non-detectable level. Together, findings of the current study uncovered the crucial reactive species in PAW inactivate MNV-1 and provided a potential disinfection strategy to combat HuNoV in foods, water, and environment.


Asunto(s)
Desinfectantes , Desinfección , Peróxido de Hidrógeno , Norovirus , Inactivación de Virus , Agua , Norovirus/efectos de los fármacos , Norovirus/fisiología , Inactivación de Virus/efectos de los fármacos , Animales , Ratones , Agua/química , Desinfectantes/farmacología , Desinfección/métodos , Gases em Plasma/farmacología , Radical Hidroxilo/metabolismo , Nitratos/farmacología , Concentración de Iones de Hidrógeno
2.
PLoS Pathog ; 20(9): e1012480, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39226332

RESUMEN

Norovirus infection is characterised by a rapid onset of disease and the development of debilitating symptoms including projectile vomiting and diffuse diarrhoea. Vaccines and antivirals are sorely lacking and developments in these areas are hampered by the lack of an adequate cell culture system to investigate human norovirus replication and pathogenesis. Herein, we describe how the model norovirus, Mouse norovirus (MNV), produces a viral protein, NS3, with the functional capacity to attenuate host protein translation which invokes the activation of cell death via apoptosis. We show that this function of NS3 is conserved between human and mouse viruses and map the protein domain attributable to this function. Our study highlights a critical viral protein that mediates crucial activities during replication, potentially identifying NS3 as a worthy target for antiviral drug development.


Asunto(s)
Infecciones por Caliciviridae , Macrófagos , Norovirus , Norovirus/fisiología , Animales , Ratones , Infecciones por Caliciviridae/virología , Macrófagos/virología , Macrófagos/metabolismo , Humanos , Biosíntesis de Proteínas , Replicación Viral/fisiología , Muerte Celular/fisiología , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Apoptosis
3.
PLoS Pathog ; 20(7): e1011909, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38976719

RESUMEN

Viruses are obligate intracellular parasites that rely on host cell metabolism for successful replication. Thus, viruses rewire host cell pathways involved in central carbon metabolism to increase the availability of building blocks for successful propagation. However, the underlying mechanisms of virus-induced alterations to host metabolism are largely unknown. Noroviruses (NoVs) are highly prevalent pathogens that cause sporadic and epidemic viral gastroenteritis. In the present study, we uncovered several strain-specific and shared host cell metabolic requirements of three murine norovirus (MNV) strains, MNV-1, CR3, and CR6. While all three strains required glycolysis, glutaminolysis, and the pentose phosphate pathway for optimal infection of macrophages, only MNV-1 relied on host oxidative phosphorylation. Furthermore, the first metabolic flux analysis of NoV-infected cells revealed that both glycolysis and glutaminolysis are upregulated during MNV-1 infection of macrophages. Glutamine deprivation affected the viral lifecycle at the stage of genome replication, resulting in decreased non-structural and structural protein synthesis, viral assembly, and egress. Mechanistic studies further showed that MNV infection and overexpression of the non-structural protein NS1/2 increased the enzymatic activity of the rate-limiting enzyme glutaminase. In conclusion, the inaugural investigation of NoV-induced alterations to host glutaminolysis identified NS1/2 as the first viral molecule for RNA viruses that regulates glutaminolysis either directly or indirectly. This increases our fundamental understanding of virus-induced metabolic alterations and may lead to improvements in the cultivation of human NoVs.


Asunto(s)
Infecciones por Caliciviridae , Glutamina , Norovirus , Proteínas no Estructurales Virales , Replicación Viral , Norovirus/fisiología , Replicación Viral/fisiología , Ratones , Animales , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Glutamina/metabolismo , Infecciones por Caliciviridae/virología , Infecciones por Caliciviridae/metabolismo , Macrófagos/virología , Macrófagos/metabolismo , Humanos , Glutaminasa/metabolismo , Glucólisis/fisiología , Células RAW 264.7
4.
Food Microbiol ; 123: 104591, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39038896

RESUMEN

Human noroviruses (HuNoVs) are the leading etiological agent causing the worldwide outbreaks of acute epidemic non-bacterial gastroenteritis. Histo-blood group antigens (HBGAs) are commonly acknowledged as cellular receptors or co-receptors for HuNoVs. However, certain genotypes of HuNoVs cannot bind with any HBGAs, suggesting potential additional co-factors and attachment receptors have not been identified yet. In addition, food items, such as oysters and lettuce, play an important role in the transmission of HuNoVs. In the past decade, a couple of attachment factors other than HBGAs have been identified and analyzed from foods and microbiomes. Attachment factors exhibit potential as inhibitors of viral binding to receptors on host cells. Therefore, it is imperative to further characterize the attachment factors for HuNoVs present in foods to effectively control the spread of HuNoVs within the food chain. This review summarizes the potential attachment factors/receptors of HuNoVs in humans, foods, and microbiome.


Asunto(s)
Infecciones por Caliciviridae , Gastroenteritis , Norovirus , Acoplamiento Viral , Norovirus/genética , Norovirus/fisiología , Humanos , Gastroenteritis/virología , Gastroenteritis/microbiología , Infecciones por Caliciviridae/virología , Receptores Virales/metabolismo , Receptores Virales/genética , Animales , Antígenos de Grupos Sanguíneos/metabolismo , Microbiología de Alimentos
5.
J Virol ; 98(7): e0202023, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38884472

RESUMEN

Human noroviruses (HuNoVs) are a diverse group of RNA viruses that cause endemic and pandemic acute viral gastroenteritis. Previously, we reported that many HuNoV strains require bile or bile acid (BA) to infect human jejunal intestinal enteroid cultures. BA was not essential for the replication of a pandemic-causing GII.4 HuNoV strain. We found the hydrophobic BA glycochenodeoxycholic acid (GCDCA) promotes the replication of the BA-dependent strain GII.3 in jejunal enteroids. Furthermore, we found that inhibition of the G-protein-coupled BA receptor, sphingosine-1-phosphate receptor 2 (S1PR2), by JTE-013, reduced GII.3 infection dose-dependently and inhibited GII.3 cellular uptake in enteroids. Herein, we sought to determine whether S1PR2 is required for other BA-dependent HuNoV strains, the BA-independent GII.4, and whether S1PR2 is required for BA-dependent HuNoV infection in HIEs from other small intestinal segments. We found a second S1PR2 inhibitor, GLPG2938, reduces GII.3 infection dose-dependently, and an S1PR2 agonist (CYM-5520) enhances GII.3 replication in the absence of GCDCA. GII.3 replication also is abrogated in the presence of JTE-013 and CYM-5520. JTE-013 inhibition of S1PR2 in jejunal HIEs reduces GI.1, GII.3, and GII.17 (BA-dependent) but not GII.4 Sydney (BA-independent) infection, providing additional evidence of strain-specific differences in HuNoV infection. Finally, GII.3 infection of duodenal, jejunal, and ileal lines derived from the same individual is reduced with S1PR2 inhibition, indicating a common mechanism of BA-dependent infection among multiple segments of the small intestine. Our results support a model where BA-dependent HuNoVs exploit BA effects on S1PR2 to infect the entire small intestine.IMPORTANCEHuman noroviruses (HuNoVs) are important viral human pathogens that cause both outbreaks and sporadic gastroenteritis. These viruses are diverse, and many strains are capable of infecting humans. Our previous studies have identified strain-specific requirements for hydrophobic bile acids (BAs) to infect intestinal epithelial cells. Moreover, we identified a BA receptor, sphingosine-1-phosphate receptor 2 (S1PR2), required for infection by a BA-dependent strain. To better understand how various HuNoV strains enter and infect the small intestine and the role of S1PR2 in HuNoV infection, we evaluated infection by additional HuNoV strains using an expanded repertoire of intestinal enteroid cell lines. We found that multiple BA-dependent strains, but not a BA-independent strain, all require S1PR2 for infection. In addition, BA-dependent infection requires S1PR2 in multiple segments of the small intestine. Together, these results indicate that S1PR2 has value as a potential therapeutic target for BA-dependent HuNoV infection.


Asunto(s)
Ácidos y Sales Biliares , Norovirus , Receptores de Esfingosina-1-Fosfato , Replicación Viral , Humanos , Norovirus/efectos de los fármacos , Norovirus/fisiología , Norovirus/genética , Receptores de Esfingosina-1-Fosfato/metabolismo , Receptores de Esfingosina-1-Fosfato/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Ácidos y Sales Biliares/farmacología , Ácidos y Sales Biliares/metabolismo , Infecciones por Caliciviridae/virología , Infecciones por Caliciviridae/metabolismo , Piridinas/farmacología , Gastroenteritis/virología , Yeyuno/virología , Yeyuno/metabolismo , Organoides/virología , Organoides/metabolismo , Pirazoles
6.
PLoS Pathog ; 20(5): e1011961, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38701091

RESUMEN

Noroviruses (NoVs) are a leading cause of viral gastroenteritis. Despite global clinical relevance, our understanding of how host factors, such as antiviral cytokines interferons (IFNs), modulate NoV population dynamics is limited. Murine NoV (MNoV) is a tractable in vivo model for the study of host regulation of NoV. A persistent strain of MNoV, CR6, establishes a reservoir in intestinal tuft cells for chronic viral shedding in stool. However, the influence of host innate immunity and permissive cell numbers on viral population dynamics is an open question. We generated a pool of 20 different barcoded viruses (CR6BC) by inserting 6-nucleotide barcodes at the 3' position of the NS4 gene and used this pool as our viral inoculum for in vivo infections of different mouse lines. We found that over the course of persistent CR6 infection, shed virus was predominantly colon-derived, and viral barcode richness decreased over time irrespective of host immune status, suggesting that persistent infection involves a series of reinfection events. In mice lacking the IFN-λ receptor, intestinal barcode richness was enhanced, correlating with increased viral intestinal replication. IL-4 treatment, which increases tuft cell numbers, also increased barcode richness, indicating the abundance of permissive tuft cells to be a bottleneck during CR6 infection. In mice lacking type I IFN signaling (Ifnar1-/-) or all IFN signaling (Stat1-/-), barcode diversity at extraintestinal sites was dramatically increased, implicating different IFNs as critical bottlenecks at specific tissue sites. Of interest, extraintestinal barcodes were overlapping but distinct from intestinal barcodes, indicating that disseminated virus represents a distinct viral population than that replicating in the intestine. Barcoded viruses are a valuable tool to explore the influence of host factors on viral diversity in the context of establishment and maintenance of infection as well as dissemination and have provided important insights into how NoV infection proceeds in immunocompetent and immunocompromised hosts.


Asunto(s)
Infecciones por Caliciviridae , Interferones , Norovirus , Animales , Norovirus/fisiología , Infecciones por Caliciviridae/virología , Infecciones por Caliciviridae/inmunología , Ratones , Interferones/metabolismo , Infección Persistente/virología , Infección Persistente/inmunología , Ratones Endogámicos C57BL , Mucosa Intestinal/virología , Mucosa Intestinal/inmunología , Gastroenteritis/virología , Replicación Viral , Ratones Noqueados , Inmunidad Innata , Esparcimiento de Virus
7.
J Virol ; 98(5): e0004724, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38651898

RESUMEN

RNA viruses lack proofreading in their RNA polymerases and therefore exist as genetically diverse populations. By exposing these diverse viral populations to selective pressures, viruses with mutations that confer fitness advantages can be enriched. To examine factors important for viral tropism and host restriction, we passaged murine norovirus (MNV) in a human cell line, HeLa cells, to select mutant viruses with increased fitness in non-murine cells. A major determinant of host range is expression of the MNV receptor CD300lf on mouse cells, but additional host factors may limit MNV replication in human cells. We found that viruses passaged six times in HeLa cells had enhanced replication compared with the parental virus. The passaged viruses had several mutations throughout the viral genome, which were primarily located in the viral non-structural coding regions. Although viral attachment was not altered for the passaged viruses, their replication was higher than the parental virus when the entry was bypassed, suggesting that the mutant viruses overcame a post-entry block in human cells. Three mutations in the viral NS1 protein were sufficient for enhanced post-entry replication in human cells. We found that the human cell-adapted MNV variants had reduced fitness in murine BV2 cells and infected mice, with reduced viral titers. These results suggest a fitness tradeoff, where increased fitness in a non-native host cell reduces fitness in a natural host environment. Overall, this work suggests that MNV tropism is determined by the presence of not only the viral receptor but also post-entry factors. IMPORTANCE: Viruses infect specific species and cell types, which is dictated by the expression of host factors required for viral entry as well as downstream replication steps. Murine norovirus (MNV) infects mouse cells, but not human cells. However, human cells expressing the murine CD300lf receptor support MNV replication, suggesting that receptor expression is a major determinant of MNV tropism. To determine whether other factors influence MNV tropism, we selected for variants with enhanced replication in human cells. We identified mutations that enhance MNV replication in human cells and demonstrated that these mutations enhance infection at a post-entry replication step. Therefore, MNV infection of human cells is restricted at both entry and post-entry stages. These results shed new light on factors that influence viral tropism and host range.


Asunto(s)
Norovirus , Tropismo Viral , Internalización del Virus , Animales , Humanos , Ratones , Infecciones por Caliciviridae/virología , Genoma Viral , Células HeLa , Especificidad del Huésped , Mutación , Norovirus/genética , Norovirus/fisiología , Receptores Virales/metabolismo , Receptores Virales/genética , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Acoplamiento Viral , Replicación Viral
8.
Food Environ Virol ; 16(2): 241-252, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38570420

RESUMEN

As a natural nonflavonoid polyphenol compound, resveratrol is the main functional component of Reynoutria japonica and has anti-inflammatory, antioxidant, antiviral, and other physiological activities. In this study, the effect of resveratrol on the viability of RAW264.7 cells was examined, and murine norovirus (MNV-1) was used as a surrogate for human norovirus to evaluate the inhibitory effect of resveratrol. The concentrations of resveratrol resulting in 50% cytotoxicity (CC50) for RAW264.7 cells were 21.32 and 24.97 µg/mL after 24 and 48 h of incubation, respectively, and resveratrol at a concentration lower than the half-effective inhibitory concentration (EC50) could not damage cell DNA. The EC50 of resveratrol on MNV-1 in infected RAW264.7 cells was determined to equal 5.496 µg/mL. After RAW264.7 cells, virus, and a fresh mixture of virus and RAW264.7 cells were treated with resveratrol solution for 1 h (denoted cell pre-treatment, virus pre-treatment, and mixture coprocessing), the RAW264.7 cells obtained after cell pre-treatment exhibited lower virus infection, and MNV-1 obtained after virus pre-treatment and mixture coprocessing showed a decreased infectious capacity. The inhibition ratio of resveratrol on MNV-1 did not significantly differ between the treatments at 4 and 25 °C or among the various pH values except for the lower acidic condition (pH 2). TEM revealed significant changes in the morphology of MNV-1 after treatment with resveratrol, and molecular docking indicated that resveratrol strongly binds to the viral capsid protein of MNV-1. In addition, resveratrol regulated the expression of cytokine that protects against MNV-1 infection. Therefore, at a lower concentration, resveratrol, a natural component from Reynoutria japonica, exerts an inhibitory effect on MNV-1 growth and could be used as a safe additive in food products to improve the nutritional status and control norovirus.


Asunto(s)
Antivirales , Norovirus , Resveratrol , Resveratrol/farmacología , Resveratrol/química , Norovirus/efectos de los fármacos , Norovirus/crecimiento & desarrollo , Norovirus/fisiología , Ratones , Animales , Células RAW 264.7 , Antivirales/farmacología , Antivirales/química , Humanos , Extractos Vegetales/farmacología , Extractos Vegetales/química , Estilbenos/farmacología , Estilbenos/química , Infecciones por Caliciviridae/virología , Infecciones por Caliciviridae/veterinaria , Infecciones por Caliciviridae/tratamiento farmacológico , Macrófagos/virología , Macrófagos/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos
9.
Sci Immunol ; 9(93): eadi7038, 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38517952

RESUMEN

The persistent murine norovirus strain MNVCR6 is a model for human norovirus and enteric viral persistence. MNVCR6 causes chronic infection by directly infecting intestinal tuft cells, rare chemosensory epithelial cells. Although MNVCR6 induces functional MNV-specific CD8+ T cells, these lymphocytes fail to clear infection. To examine how tuft cells promote immune escape, we interrogated tuft cell interactions with CD8+ T cells by adoptively transferring JEDI (just EGFP death inducing) CD8+ T cells into Gfi1b-GFP tuft cell reporter mice. Unexpectedly, some intestinal tuft cells partially resisted JEDI CD8+ T cell-mediated killing-unlike Lgr5+ intestinal stem cells and extraintestinal tuft cells-despite seemingly normal antigen presentation. When targeting intestinal tuft cells, JEDI CD8+ T cells predominantly adopted a T resident memory phenotype with decreased effector and cytotoxic capacity, enabling tuft cell survival. JEDI CD8+ T cells neither cleared nor prevented MNVCR6 infection in the colon, the site of viral persistence, despite targeting a virus-independent antigen. Ultimately, we show that intestinal tuft cells are relatively resistant to CD8+ T cells independent of norovirus infection, representing an immune-privileged niche that can be leveraged by enteric microbes.


Asunto(s)
Linfocitos T CD8-positivos , Norovirus , Ratones , Humanos , Animales , Células en Penacho , Norovirus/fisiología , Privilegio Inmunológico , Intestinos
10.
Food Environ Virol ; 16(2): 171-179, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38457095

RESUMEN

Norovirus is the leading cause of viral gastroenteritis globally. While person-to-person transmission is most commonly reported route of infection, human norovirus is frequently associated with foodborne transmission, including through consumption of contaminated bivalve molluscan shellfish. Reverse transcription (RT)-qPCR is most commonly used method for detecting human norovirus detection in foods, but does not inform on its infectivity, posing challenges for assessing intervention strategies aimed at risk elimination. In this study, RT-qPCR was used in conjunction with a derivative of the photoreactive DNA binding dye propidium monoazide (PMAxx™) (PMAxx-RT-qPCR) to evaluate the viral capsid integrity of norovirus genogroup I and II (GI and GII) in shellfish following high pressure processing (HPP). Norovirus GI.3 and GII.4 bioaccumulated oysters were subjected to HPP at pressures of 300 and 450 MPa at 15 °C, and 300, 450 and 600 MPa at 20 °C. Samples were analysed using both RT-qPCR and PMAxx-RT-qPCR. For each sample, norovirus concentration (genome copies/g digestive tissue) determined by RT-qPCR was divided by the PMAxx-RT-qPCR concentration, giving the relative non-intact (RNI) ratio. The RNI ratio values relate to the amount of non-intact (non-infectious) viruses compared to fully intact (possible infectious) viruses. Our findings revealed an increasing RNI ratio value, indicating decreasing virus integrity, with increasing pressure and decreasing pressure. At 300 MPa, for norovirus GI, the median [95% confidence interval, CI] RNI ratio values were 2.6 [1.9, 3.0] at 15 °C compared to 1.1 [0.9, 1.8] at 20 °C. At 450 MPa, the RNI ratio values were 5.5 [2.9, 7.0] at 15 °C compared to 1.3 [1.0, 1.6] at 20 °C. At 600 MPa, the RNI ratio value was 5.1 [2.9, 13.4] at 20 °C. For norovirus GII, RT-qPCR and PMAxx-RT-qPCR detections were significantly reduced at 450 and 600 MPa at both 15 °C and 20 °C, with the median [95% CI] RNI ratio value at 300 MPa being 1.1 [0.8, 1.6]. Following HPP treatment, the use of PMAxx-RT-qPCR enables the selective detection of intact and potential infectious norovirus, enhancing our understanding of the inactivation profiles and supporting the development of more effective risk assessment strategies.


Asunto(s)
Manipulación de Alimentos , Norovirus , Ostreidae , Reacción en Cadena en Tiempo Real de la Polimerasa , Mariscos , Inactivación de Virus , Norovirus/genética , Norovirus/aislamiento & purificación , Norovirus/fisiología , Norovirus/clasificación , Norovirus/crecimiento & desarrollo , Animales , Ostreidae/virología , Mariscos/virología , Manipulación de Alimentos/métodos , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Humanos , Contaminación de Alimentos/análisis , Presión Hidrostática , Propidio/química , Propidio/análogos & derivados , Azidas/química , Infecciones por Caliciviridae/virología
11.
J Virol ; 98(4): e0166323, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38470106

RESUMEN

Human norovirus (HuNoV) is a major cause of acute gastroenteritis and foodborne diseases, affecting all age groups. Despite its clinical needs, no approved antiviral therapies are available. Since the discovery of HuNoV in 1972, studies on anti-norovirals, mechanism of HuNoV infection, viral inactivation, etc., have been hampered by the lack of a robust laboratory-based cultivation system for HuNoV. A recent breakthrough in the development of HuNoV cultivation systems has opened opportunities for researchers to investigate HuNoV biology in the context of de novo HuNoV infections. A tissue stem cell-derived human intestinal organoid/enteroid (HIO) culture system is one of those that supports HuNoV replication reproducibly and, to our knowledge, is most widely distributed to laboratories worldwide to study HuNoV and develop therapeutic strategies. This review summarizes recently developed HuNoV cultivation systems, including HIO, and their use in antiviral studies.


Asunto(s)
Norovirus , Humanos , Antivirales/farmacología , Infecciones por Caliciviridae/tratamiento farmacológico , Infecciones por Caliciviridae/virología , Gastroenteritis/tratamiento farmacológico , Gastroenteritis/virología , Intestinos/virología , Norovirus/efectos de los fármacos , Norovirus/fisiología , Animales , Organoides/efectos de los fármacos , Organoides/virología , Cultivo de Virus
12.
J Virol ; 98(3): e0185123, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38353537

RESUMEN

Recently, we identified the coxsackie and adenovirus receptor (CAR) as the entry receptor for rhesus enteric calicivirus (ReCV) isolate FT285 and demonstrated that co-expression of the CAR and the type B histo-blood group antigen (HBGA) is required to convert the resistant CHO cell line susceptible to infection. To address whether the CAR is also the functional entry receptor for other ReCV isolates and the requirement for specific HBGAs or other glycans, here we used a panel of recombinant CHO cell lines expressing the CAR and the type A, B, or H HBGAs alone or in combination. Infection studies with three diverse ReCV strains, the prototype GI.1 Tulane virus (TV), GI.2 ReCV-FT285, and GI.3 ReCV-FT7, identified that cell surface expression of the CAR is an absolute requirement for all three strains to promote susceptibility to infection, while the requirement for HBGAs varies among the strains. In addition to the CAR, ReCV-FT285 and TV require type A or B HBGAs for infection. In the absence of HBGAs, TV, but not Re-CV FT285, can also utilize sialic acids, while ReCV-FT7 infection is HBGA-independent and relies on CAR and sialic acid expression. In summary, we demonstrated strain-specific diversity of susceptibility requirements for ReCV infections and that CAR, type A and B HBGA, and sialic acid expression control susceptibility to infection with the three ReCV isolates studied. Our study also indicates that the correlation between in vitro HBGA binding and HBGAs required for infection is relatively high, but not absolute. This has direct implications for human noroviruses.IMPORTANCEHuman noroviruses (HuNoVs) are important enteric pathogens. The lack of a robust HuNoV cell culture system is a bottleneck for HuNoV cell culture-based studies. Often, cell culture-adapted caliciviruses that rapidly replicate in conventional cell lines and recapitulate biological features of HuNoVs are utilized as surrogates. Particularly, rhesus enteric caliciviruses (ReCVs) display remarkable similarities, including the primate host, clinical manifestation of gastroenteritis, genetic/antigenic diversity, and reliance on histo-blood group antigens (HBGAs) for attachment. While the HuNoV entry receptor(s) is unknown, the coxsackie and adenovirus receptor (CAR) has recently been identified as the ReCV entry receptor. Here, we identified the CAR, the type A and B HBGAs, and sialic acids as critical cell surface molecules controlling susceptibility to ReCV infections. The CAR is required for all ReCV isolates studied. However, the requirement for the different carbohydrate molecules varies among different ReCV strains. Our findings have direct implications for HuNoVs.


Asunto(s)
Infecciones por Caliciviridae , Caliciviridae , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Animales , Cricetinae , Humanos , Antígenos de Grupos Sanguíneos/metabolismo , Caliciviridae/fisiología , Infecciones por Caliciviridae/virología , Células CHO , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/metabolismo , Intestino Delgado/virología , Ácido N-Acetilneuramínico/metabolismo , Norovirus/fisiología
13.
Int J Food Microbiol ; 413: 110601, 2024 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-38301540

RESUMEN

Vegetables are globally associated with a considerable number of foodborne outbreaks caused by viral infections, specifically human norovirus. In fresh produce industry, washing represents a critical step for food safety as process wash water (PWW) needs to be maintained at appropriate microbial quality to prevent water-mediated cross-contamination. This study aimed to explore the disinfection efficacy of chlorine (free chlorine, FC), chlorine dioxide (ClO2) and peracetic acid (PAA) in PWW against infectious human norovirus and Tulane virus (TV). First, we tested the extent of TV inactivation in baby leaf, bell pepper, and vegetables mix PWW and monitored the viral decay by cell culture. Then, inactivation kinetics were defined for infectious human norovirus exposed to FC, ClO2 and PAA in baby leaves PWW using the human intestinal enteroids (HIE) system. Finally, kinetic inactivation models were fitted to TV reduction and decay of sanitizers to aid the implementation of disinfection strategies. Results showed that >8 log10 human norovirus and 3.9 log10 TV were inactivated by 20 ppm FC within 1 min; and by 3 ppm ClO2 in 1 min (TV) or 5 min (norovirus). PAA treatment at 80 ppm reduced ca. 2 log10 TV but not completely inactivated the virus even after 20 min exposure, while 5 min treatment prevented norovirus replication in HIE. TV inactivation in PWWs was described using an exponential decay model. Taking these data together, we demonstrated the value of applying the HIE model to validate current operational limits for the most commonly used sanitizers. The inactivation kinetics for human norovirus and TV, along with the predictive model described in this study expand the current knowledge to implement post-harvest produce safety procedures in industry settings.


Asunto(s)
Desinfectantes , Norovirus , Humanos , Desinfección/métodos , Verduras , Cloro/farmacología , Ácido Peracético/farmacología , Norovirus/fisiología , Agua , Inactivación de Virus , Desinfectantes/farmacología
14.
Int J Food Microbiol ; 413: 110582, 2024 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-38290272

RESUMEN

Heat treatment of food represents an important measure to prevent pathogen transmission. Thus far, evaluation of heat treatment processes is mainly based on data from bacteria. However, foodborne viruses have gained increasing attention during the last decades. Here, the published literature on heat stability and inactivation of human norovirus (NoV), hepatitis A virus (HAV) and hepatitis E virus (HEV) was reviewed. Data for surrogate viruses were not included. As stability assessment for foodborne viruses is often hampered by missing infectivity assays, an overview of applied methods is also presented. For NoV, molecular capsid integrity assays were mainly applied, but data from initial studies utilizing novel intestinal enteroid or zebrafish larvae assays are available now. However, these methods are still limited in applicability and sensitivity. For HAV, sufficient cell culture-based inactivation data are available, but almost exclusively for one single strain, thus limiting interpretation of the data for the wide range of field strains. For HEV, data are now available from studies using pig inoculation or cell culture. The results of the reviewed studies generally indicate that NoV, HAV and HEV possess a high heat stability. Heating at 70-72 °C for 2 min significantly reduces infectious titers, but often does not result in a >4 log10 decrease. However, heat stability greatly varied dependent on virus strain, matrix and heating regime. In addition, the applied method largely influenced the result, e.g. capsid integrity assays tend to result in higher measured stabilities than cell culture approaches. It can be concluded that the investigated foodborne viruses show a high heat stability, but can be inactivated by application of appropriate heating protocols. For HAV, suggestions for safe time/temperature combinations for specific foods can be derived from the published studies, with the limitation that they are mostly based on one strain only. Although significant improvement of infectivity assays for NoV and HEV have been made during the last years, further method development regarding sensitivity, robustness and broader applicability is important to generate more reliable heat inactivation data for these foodborne viruses in future.


Asunto(s)
Virus de la Hepatitis A , Virus de la Hepatitis E , Norovirus , Animales , Humanos , Porcinos , Calor , Pez Cebra , Virus de la Hepatitis A/fisiología , Temperatura , Virus de la Hepatitis E/fisiología , Norovirus/fisiología , Inactivación de Virus
15.
Virology ; 589: 109921, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37939648

RESUMEN

Human norovirus is the leading cause of acute gastroenteritis worldwide, however despite the significance of this pathogen, we have a limited understanding of how noroviruses cause disease, and modulate the innate immune response. Programmed cell death (PCD) is an important part of the innate response to invading pathogens, but little is known about how specific PCD pathways contribute to norovirus replication. Here, we reveal that murine norovirus (MNV) virus-induced PCD in macrophages correlates with the release of infectious virus. We subsequently show, genetically and chemically, that MNV-induced cell death and viral replication occurs independent of the activity of inflammatory mediators. Further analysis revealed that MNV infection promotes the cleavage of apoptotic caspase-3 and PARP. Correspondingly, pan-caspase inhibition, or BAX and BAK deficiency, perturbed viral replication rates and delayed virus release and cell death. These results provide new insights into how MNV harnesses cell death to increase viral burden.


Asunto(s)
Infecciones por Caliciviridae , Norovirus , Ratones , Humanos , Animales , Macrófagos , Apoptosis , Inmunidad Innata , Norovirus/fisiología , Replicación Viral
16.
Virology ; 585: 232-239, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37406580

RESUMEN

Viruses can be involved in respiratory disorders in horses, with limited therapeutic options. Citrate-complexed silver nanoparticles (C-AgNP) have shown bactericidal properties after in vitro nebulization. The aim of the present study was to assess the virucidal activity of C-AgNP after in vitro instillation or nebulization on equine herpesvirus-1 (EHV-1) and murine norovirus (MNV), the latter used as surrogate for small non-enveloped viruses. Both viruses were instilled or nebulized with C-AgNP of increasing concentrations, and titres were determined via TCID50 method. We demonstrated efficient inactivation of enveloped EHV-1 following instillation and nebulization of C-AgNP (infectivity losses of ≥ three orders of magnitude). While tenacious MNV was inactivated via 2000 ppm C-AgNP instillation, nebulized C-AgNP did not lead to reduction in MNV titres. Nebulization of C-AgNP may represent a novel virucidal therapeutic approach in horses. Further investigations are needed to assess its safety and effective concentrations for in vivo use.


Asunto(s)
Herpesvirus Équido 1 , Nanopartículas del Metal , Norovirus , Animales , Caballos , Ratones , Ácido Cítrico , Plata/farmacología , Norovirus/fisiología
17.
Nature ; 616(7955): 152-158, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36991121

RESUMEN

Non-enveloped viruses require cell lysis to release new virions from infected cells, suggesting that these viruses require mechanisms to induce cell death. Noroviruses are one such group of viruses, but there is no known mechanism that causes norovirus infection-triggered cell death and lysis1-3. Here we identify a molecular mechanism of norovirus-induced cell death. We found that the norovirus-encoded NTPase NS3 contains an N-terminal four-helix bundle domain homologous to the membrane-disruption domain of the pseudokinase mixed lineage kinase domain-like (MLKL). NS3 has a mitochondrial localization signal and thus induces cell death by targeting mitochondria. Full-length NS3 and an N-terminal fragment of the protein bound the mitochondrial membrane lipid cardiolipin, permeabilized the mitochondrial membrane and induced mitochondrial dysfunction. Both the N-terminal region and the mitochondrial localization motif of NS3 were essential for cell death, viral egress from cells and viral replication in mice. These findings suggest that noroviruses have acquired a host MLKL-like pore-forming domain to facilitate viral egress by inducing mitochondrial dysfunction.


Asunto(s)
Muerte Celular , Norovirus , Nucleósido-Trifosfatasa , Proteínas Quinasas , Proteínas Virales , Animales , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Norovirus/enzimología , Norovirus/crecimiento & desarrollo , Norovirus/patogenicidad , Norovirus/fisiología , Proteínas Quinasas/química , Replicación Viral , Proteínas Virales/química , Proteínas Virales/metabolismo , Nucleósido-Trifosfatasa/química , Nucleósido-Trifosfatasa/metabolismo , Señales de Clasificación de Proteína , Cardiolipinas/metabolismo , Membranas Mitocondriales/química , Membranas Mitocondriales/metabolismo
18.
Food Environ Virol ; 15(2): 167-175, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36920726

RESUMEN

Aqueous extracts of Quillaja saponaria Molina are US FDA approved as food additives in beverages with known antiviral activity. Due to lack of commercially available vaccines against human noroviruses (HNoVs), alternate methods to prevent their spread and the subsequent emergence of variant strains are being researched. Furthermore, HNoVs are not yet culturable at high enough titers to determine inactivation, therefore surrogates continue to be used. This research analyzed the effect of aqueous Quillaja saponaria extracts (QE) against HNoV surrogates, Tulane virus (TV), murine norovirus (MNV-1), and feline calicivirus (FCV-F9) at room temperature (RT) and 37 °C. Viruses (~ 5 log PFU/mL) were individually treated with 1:1 or 1:5 (v/v) diluted QE (pH ~ 3.75), malic acid control (pH 3.0) or phosphate-buffered saline (pH 7.2, as control) at 37 °C or RT for up to 6 h. Individual treatments were replicated three times using duplicate plaque assays for each treatment. FCV-F9 at ~ 5 log PFU/mL was not detectable after 15 min by 1:1 QE at 37 °C and RT. At RT, 1:5 QE lowered FCV-F9 titers by 2.05, 2.14 and 2.74 log PFU/mL after 0.5 h, 1 h and 2 h, respectively. MNV-1 showed marginal reduction of < 1 log PFU/mL after 15 min with 1:1 or 1:5 QE at 37 °C without any significant reduction at RT, while TV titers decreased by 2.2 log PFU/mL after 30 min and were undetectable after 3 h at 37 °C. Longer incubation with higher QE concentrations may be required for improved antiviral activity against MNV-1 and TV.


Asunto(s)
Calicivirus Felino , Enfermedades Transmitidas por los Alimentos , Norovirus , Gatos , Humanos , Animales , Ratones , Antivirales/farmacología , Quillaja , Norovirus/fisiología
19.
Nat Commun ; 14(1): 1148, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36854760

RESUMEN

Globally, most cases of gastroenteritis are caused by pandemic GII.4 human norovirus (HuNoV) strains with no approved therapies or vaccines available. The cellular pathways that these strains exploit for cell entry and internalization are unknown. Here, using nontransformed human jejunal enteroids (HIEs) that recapitulate the physiology of the gastrointestinal tract, we show that infectious GII.4 virions and virus-like particles are endocytosed using a unique combination of endosomal acidification-dependent clathrin-independent carriers (CLIC), acid sphingomyelinase (ASM)-mediated lysosomal exocytosis, and membrane wound repair pathways. We found that besides the known interaction of the viral capsid Protruding (P) domain with host glycans, the Shell (S) domain interacts with both galectin-3 (gal-3) and apoptosis-linked gene 2-interacting protein X (ALIX), to orchestrate GII.4 cell entry. Recognition of the viral and cellular determinants regulating HuNoV entry provides insight into the infection process of a non-enveloped virus highlighting unique pathways and targets for developing effective therapeutics.


Asunto(s)
Membrana Celular , Norovirus , Internalización del Virus , Humanos , Clatrina , Norovirus/fisiología , Transducción de Señal , Membrana Celular/virología
20.
Sci Total Environ ; 874: 162380, 2023 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-36841407

RESUMEN

Safeguarding the seafood industry is important given its contribution to supporting our growing global population. However, shellfish are filter feeders that bioaccumulate microbial contaminants in their tissue from wastewater discharged into the same coastal growing environments leading to significant human disease outbreaks unless appropriately mitigated. Removal or inactivation of enteric viruses is very challenging particularly as human norovirus (hNoV) binds to specific histo-blood ligands in live oyster tissue that are consumed raw or lightly cooked. The regulatory framework that sets out use of clean seawater and UV disinfection is appropriate for bacterial decontamination at the post-harvest land-based depuration (cleaning) stage. However, additional non-thermal technologies are required to eliminate hNoV in live shellfish (particularly oysters) where published genomic studies report that low-pressure UV has limited effectiveness in inactivating hNoV. The use of the standard genomic detection method (ISO 15, 216-1:2017) is not appropriate for assessing the loss of infectious hNoV in treated live shellfish. The use of surrogate viral infectivity methods appear to offer some insight into the loss of hNoV infectiousness in live shellfish during decontamination. This paper reviews the use of existing and potentially other combinational treatment approaches to enhance the removal or inactivation of enteric viruses in live shellfish. The use of alternative and complementary novel diagnostic approaches to discern viable hNoV are discussed. The effectiveness and virological safety of new affordable hNoV intervention(s) require testing and validating at commercial shellfish production in conjunction with laboratory-based research. Appropriate risk management planning should encompass key stakeholders including local government and the wastewater industry. Gaining a mechanistic understanding of the relationship between hNoV response at molecular and structural levels in individually treated oysters as a unit will inform predictive modeling and appropriate treatment technologies. Global warming of coastal growing environments may introduce additional contaminant challenges (such as invasive species); thus, underscoring need to develop real-time ecosystem monitoring of growing environments to alert shellfish producers to appropriately mitigate these threats.


Asunto(s)
Norovirus , Ostreidae , Humanos , Animales , Norovirus/fisiología , Aguas Residuales , Descontaminación , Ecosistema , Mariscos/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA