Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
J Virol ; 98(5): e0035024, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38591900

ABSTRACT

Feline calicivirus (FCV) is one of the few members of the Caliciviridae family that grows well in cell lines and, therefore, serves as a surrogate to study the biology of other viruses in the family. Conley et al. (14) demonstrated that upon the receptor engagement to the capsid, FCV VP2 forms a portal-like assembly, which might provide a channel for RNA release. However, the process of calicivirus RNA release is not yet fully understood. Our findings suggest that the separation of the FCV capsid from its genome RNA (gRNA) occurs rapidly in the early endosomes of infected cells. Using a liposome model decorated with the FCV cell receptor fJAM-A, we demonstrate that FCV releases its gRNA into the liposomes by penetrating membranes under low pH conditions. Furthermore, we found that VP2, which is rich in hydrophobic residues at its N-terminus, functions as the pore-forming protein. When we substituted the VP2 N-terminal hydrophobic residues, the gRNA release efficacy of the FCV mutants decreased. In conclusion, our results suggest that in the acidic environment of early endosomes, FCV VP2 functions as the pore-forming protein to mediate gRNA release into the cytoplasm of infected cells. This provides insight into the mechanism of calicivirus genome release.IMPORTANCEResearch on the biology and pathogenicity of certain caliciviruses, such as Norovirus and Sapovirus, is hindered by the lack of easy-to-use cell culture system. Feline calicivirus (FCV), which grows effectively in cell lines, is used as a substitute. At present, there is limited understanding of the genome release mechanism in caliciviruses. Our findings suggest that FCV uses VP2 to pierce the endosome membrane for genome release and provide new insights into the calicivirus gRNA release mechanism.


Subject(s)
Calicivirus, Feline , Capsid Proteins , Endosomes , Genome, Viral , RNA, Viral , Calicivirus, Feline/genetics , Calicivirus, Feline/metabolism , Calicivirus, Feline/physiology , Cats , Endosomes/virology , Endosomes/metabolism , Animals , RNA, Viral/metabolism , RNA, Viral/genetics , Cell Line , Capsid Proteins/metabolism , Capsid Proteins/genetics , Caliciviridae Infections/virology , Caliciviridae Infections/metabolism , Virus Release , Capsid/metabolism , Liposomes/metabolism
2.
J Virol ; 97(12): e0171923, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38032199

ABSTRACT

IMPORTANCE: All viruses initiate infection by utilizing receptors to attach to target host cells. These virus-receptor interactions can therefore dictate viral replication and pathogenesis. Understanding the nature of virus-receptor interactions could also be important for the development of novel therapies. Noroviruses are non-enveloped icosahedral viruses of medical importance. They are a common cause of acute gastroenteritis with no approved vaccine or therapy and are a tractable model for studying fundamental virus biology. In this study, we utilized the murine norovirus model system to show that variation in a single amino acid of the major capsid protein alone can affect viral infectivity through improved attachment to suspension cells. Modulating plasma membrane mobility reduced infectivity, suggesting an importance of membrane mobility for receptor recruitment and/or receptor conformation. Furthermore, different substitutions at this site altered viral tissue distribution in a murine model, illustrating how in-host capsid evolution could influence viral infectivity and/or immune evasion.


Subject(s)
Caliciviridae Infections , Capsid Proteins , Norovirus , Animals , Mice , Amino Acid Substitution , Caliciviridae Infections/metabolism , Capsid/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Immune Evasion , Norovirus/metabolism , Viral Core Proteins/metabolism
3.
Cell Rep ; 41(6): 111593, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36351394

ABSTRACT

Murine norovirus (MNoV) is a model for human norovirus and for interrogating mechanisms of viral tropism and persistence. We previously demonstrated that the persistent strain MNoVCR6 infects tuft cells, which are dispensable for the non-persistent strain MNoVCW3. We now show that diverse MNoV strains require tuft cells for chronic enteric infection. We also demonstrate that interferon-λ (IFN-λ) acts directly on tuft cells to cure chronic MNoVCR6 infection and that type I and III IFNs signal together via STAT1 in tuft cells to restrict MNoVCW3 tropism. We then develop an enteroid model and find that MNoVCR6 and MNoVCW3 similarly infect tuft cells with equal IFN susceptibility, suggesting that IFN derived from non-epithelial cells signals on tuft cells in trans to restrict MNoVCW3 tropism. Thus, tuft cell tropism enables MNoV persistence and is determined by tuft cell-intrinsic factors (viral receptor expression) and -extrinsic factors (immunomodulatory signaling by non-epithelial cells).


Subject(s)
Caliciviridae Infections , Norovirus , Mice , Humans , Animals , Norovirus/physiology , Caliciviridae Infections/metabolism , Mice, Inbred C57BL , Viral Tropism , Tropism
4.
Biochem Biophys Res Commun ; 604: 8-13, 2022 05 14.
Article in English | MEDLINE | ID: mdl-35279444

ABSTRACT

Feline calicivirus (FCV) is an important and highly prevalent pathogen of cats that causes acute infectious respiratory disease. Here it is shown in vitro that FCV induces the production of cyclooxygenase-2 (COX-2) through the MEK1-ERK1/2 signaling pathway. Screening of FCV proteins revealed that FCV non-structural protein VPg enhanced COX-2 mRNA expression and protein production in CRFK cells in a concentration-dependent manner. Regions 24-54aa and 84-111aa in FCV VPg were essential for up-regulation. In vivo, COX-2 and IL-6 production caused by FCV infection of kittens was significantly suppressed by the MEK1 inhibitor AZD6244 (selumetinib) and lung inflammation and injury were practically eliminated, with body temperature being returned to normal. AZD6244 may therefore find application as an effective therapeutic agent for the treatment of FCV infection.


Subject(s)
Caliciviridae Infections , Calicivirus, Feline , Pneumonia , Animals , Benzimidazoles , Caliciviridae Infections/drug therapy , Caliciviridae Infections/metabolism , Caliciviridae Infections/veterinary , Cats , Cyclooxygenase 2/metabolism , Female , MAP Kinase Signaling System
5.
J Virol ; 96(3): e0192321, 2022 02 09.
Article in English | MEDLINE | ID: mdl-34787460

ABSTRACT

Akt (protein kinase B) is a key signaling protein in eukaryotic cells that controls many cellular processes, such as glucose metabolism and cell proliferation, for survival. As obligate intracellular pathogens, viruses modulate host cellular processes, including Akt signaling, for optimal replication. The mechanisms by which viruses modulate Akt and the resulting effects on the infectious cycle differ widely depending on the virus. In this study, we explored the effect of Akt serine 473 phosphorylation (p-Akt) during murine norovirus (MNV) infection. p-Akt increased during infection of murine macrophages with acute MNV-1 and persistent CR3 and CR6 strains. Inhibition of Akt with MK2206, an inhibitor of all three isoforms of Akt (Akt1/2/3), reduced infectious virus progeny of all three virus strains. This reduction was due to decreased viral genome replication (CR3), defective virus assembly (MNV-1), or altered cellular egress (CR3 and CR6) in a virus strain-dependent manner. Collectively, our data demonstrate that Akt activation increases in macrophages during the later stages of the MNV infectious cycle, which may enhance viral infection in unique ways for different virus strains. The data, for the first time, indicate a role for Akt signaling in viral assembly and highlight additional phenotypic differences between closely related MNV strains. IMPORTANCE Human noroviruses (HNoV) are a leading cause of viral gastroenteritis, resulting in high annual economic burden and morbidity, yet there are no small-animal models supporting productive HNoV infection or robust culture systems producing cell culture-derived virus stocks. As a result, research on drug discovery and vaccine development against norovirus infection has been challenging, and no targeted antivirals or vaccines against HNoV are approved. On the other hand, murine norovirus (MNV) replicates to high titers in cell culture and is a convenient and widespread model in norovirus research. Our data demonstrate the importance of Akt signaling during the late stage of the MNV life cycle. Notably, the effect of Akt signaling on genome replication, virus assembly, and cellular egress is virus strain specific, highlighting the diversity of biological phenotypes despite small genetic variability among norovirus strains. This study is the first to demonstrate a role for Akt in viral assembly.


Subject(s)
Caliciviridae Infections/metabolism , Caliciviridae Infections/virology , Macrophages/metabolism , Macrophages/virology , Norovirus/physiology , Proto-Oncogene Proteins c-akt/metabolism , Virus Replication , Animals , Caliciviridae Infections/immunology , Disease Susceptibility , Host-Pathogen Interactions , Macrophage Activation , Macrophages/immunology , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Species Specificity
6.
Viruses ; 13(10)2021 10 14.
Article in English | MEDLINE | ID: mdl-34696500

ABSTRACT

Recognition of cell-surface glycans is an important step in the attachment of several viruses to susceptible host cells. The molecular basis of glycan interactions and their functional consequences are well studied for human norovirus (HuNoV), an important gastrointestinal pathogen. Histo-blood group antigens (HBGAs), a family of fucosylated carbohydrate structures that are present on the cell surface, are utilized by HuNoVs to initially bind to cells. In this review, we describe the discovery of HBGAs as genetic susceptibility factors for HuNoV infection and review biochemical and structural studies investigating HuNoV binding to different HBGA glycans. Recently, human intestinal enteroids (HIEs) were developed as a laboratory cultivation system for HuNoV. We review how the use of this novel culture system has confirmed that fucosylated HBGAs are necessary and sufficient for infection by several HuNoV strains, describe mechanisms of antibody-mediated neutralization of infection that involve blocking of HuNoV binding to HBGAs, and discuss the potential for using the HIE model to answer unresolved questions on viral interactions with HBGAs and other glycans.


Subject(s)
Blood Group Antigens/metabolism , Caliciviridae Infections/metabolism , Polysaccharides/metabolism , Animals , Blood Group Antigens/chemistry , Blood Group Antigens/genetics , Caliciviridae Infections/epidemiology , Fucosyltransferases/genetics , Glycoconjugates , Host Microbial Interactions , Humans , Intestines , Models, Molecular , Norovirus/genetics , Polysaccharides/genetics , Protein Binding , Protein Conformation , Protein Domains , Virus Attachment , Galactoside 2-alpha-L-fucosyltransferase
7.
Gut Microbes ; 13(1): 1979876, 2021.
Article in English | MEDLINE | ID: mdl-34586017

ABSTRACT

The study tested the hypothesis that harboring high levels of histo-blood group antigen-expressing Enerobactero cloacae is a risk factor for norovirus diarrhea. The fecal E. cloacae abundance in diarrheic norovirus positive (DNP), non-diarrheic norovirus negative (NDNN), diarrhea norovirus negative (DNN), and non-diarrhea norovirus positive (NDNP) infants was determined by qPCR, and the risk of norovirus diarrhea was assessed by logistical regression. DNP infants contained significantly higher counts of E. cloacae than NDNN and DNN infants, p = .0294, and 0.0001, respectively. The risk of norovirus diarrhea was significantly high in infants with higher counts of E. cloacae than those with lower counts, p = .009. Harboring higher counts of E. cloacae is a risk factor for norovirus diarrhea.


Subject(s)
Blood Group Antigens/genetics , Caliciviridae Infections/virology , Diarrhea/virology , Enterobacter cloacae/growth & development , Enterobacter cloacae/genetics , Feces/microbiology , Norovirus/physiology , Blood Group Antigens/metabolism , Caliciviridae Infections/genetics , Caliciviridae Infections/metabolism , Caliciviridae Infections/microbiology , Diarrhea/genetics , Diarrhea/metabolism , Diarrhea/microbiology , Enterobacter cloacae/isolation & purification , Enterobacter cloacae/metabolism , Feces/chemistry , Gastrointestinal Microbiome , Humans , Infant , Male , Norovirus/genetics , South Africa
8.
J Virol ; 95(20): e0113421, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34346771

ABSTRACT

Murine norovirus (MNV) infection results in a late translation shutoff that is proposed to contribute to the attenuated and delayed innate immune response observed both in vitro and in vivo. Recently, we further demonstrated the activation of the α subunit of eukaryotic initiation factor 2 (eIF2α) kinase GCN2 during MNV infection, which has been previously linked to immunomodulation and resistance to inflammatory signaling during metabolic stress. While viral infection is usually associated with activation of double-stranded RNA (dsRNA) binding pattern recognition receptor PKR, we hypothesized that the establishment of a metabolic stress in infected cells is a proviral event, exploited by MNV to promote replication through weakening the activation of the innate immune response. In this study, we used multi-omics approaches to characterize cellular responses during MNV replication. We demonstrate the activation of pathways related to the integrated stress response, a known driver of anti-inflammatory phenotypes in macrophages. In particular, MNV infection causes an amino acid imbalance that is associated with GCN2 and ATF2 signaling. Importantly, this reprogramming lacks the features of a typical innate immune response, with the ATF/CHOP target GDF15 contributing to the lack of antiviral responses. We propose that MNV-induced metabolic stress supports the establishment of host tolerance to viral replication and propagation. IMPORTANCE During viral infection, host defenses are typically characterized by the secretion of proinflammatory autocrine and paracrine cytokines, potentiation of the interferon (IFN) response, and induction of the antiviral response via activation of JAK and Stat signaling. To avoid these and propagate, viruses have evolved strategies to evade or counteract host sensing. In this study, we demonstrate that murine norovirus controls the antiviral response by activating a metabolic stress response that activates the amino acid response and impairs inflammatory signaling. This highlights novel tools in the viral countermeasures arsenal and demonstrates the importance of the currently poorly understood metabolic reprogramming occurring during viral infections.


Subject(s)
Caliciviridae Infections/immunology , Macrophages/virology , Activating Transcription Factor 2/metabolism , Animals , Antiviral Agents , Caliciviridae Infections/metabolism , Cell Line , Eukaryotic Initiation Factor-2/metabolism , Immunity, Innate/immunology , Inflammation/immunology , Interferons , Macrophages/immunology , Mice , Norovirus/pathogenicity , Protein Serine-Threonine Kinases/metabolism , RAW 264.7 Cells , RNA, Double-Stranded/genetics , Signal Transduction/immunology , Viral Nonstructural Proteins/metabolism , Virus Replication/genetics
9.
PLoS One ; 16(4): e0248581, 2021.
Article in English | MEDLINE | ID: mdl-33831019

ABSTRACT

Human norovirus (NoV) is the leading cause of acute viral gastroenteritis and a major source of foodborne illness. Detection of NoV in food and environmental samples is typically performed using molecular techniques, including real-time reverse transcription polymerase chain reaction (RT-PCR) and less frequently, nested real-time PCR. In this study, we conducted a controlled comparison of two published NoV detection assays: a broadly reactive one-step real-time RT-PCR and a two-step nested real-time PCR assay. A 20% human fecal suspension containing a genogroup II human NoV was serially diluted, genome extracted, and subjected to amplification using the two assays compared via PCR Units. Additional amplicon confirmation was performed by dot blot hybridization using digoxigenin (DIG)-labeled oligonucleotide probes. Both assays displayed similar amplification standard curves/amplification efficiencies; however, the nested assay consistently detected one log10 lower virus. Dot blot hybridization improved the detection limit of the nested real-time PCR by one log10 NoV genome copies but impaired the detection limit of the one-step real-time RT-PCR by one log10 NoV genome copies. These results illustrate the complexities in designing and interpreting molecular techniques having a sufficient detection limit to detect low levels of viruses that might be anticipated in contaminated food and environmental samples.


Subject(s)
Caliciviridae Infections/genetics , Feces/virology , Norovirus/genetics , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Caliciviridae Infections/diagnosis , Caliciviridae Infections/metabolism , Female , Genome, Viral , Humans , Male , RNA, Viral/metabolism
10.
J Virol ; 95(3)2021 01 13.
Article in English | MEDLINE | ID: mdl-33115870

ABSTRACT

Human noroviruses are the most common nonbacterial cause of gastroenteritis outbreaks, with new variants and genotypes frequently emerging. The origin of these new viruses is unknown; however, animals have been proposed as a potential source, as human noroviruses have been detected in animal species. Here, we investigated the potential of animals to serve as a reservoir of human noroviruses by testing norovirus attachment to formalin-fixed intestinal tissues of a range of potential reservoir animals. We set up a novel method to study norovirus binding using fluorescein isothiocyanate (FITC)-labeled virus-like particles (VLPs). In humans, noroviruses interact with histo-blood group antigens (HBGAs), carbohydrates that are expressed, among others, on the epithelial lining of the gastrointestinal tract. In animals, this interaction is not well understood. To test if virus binding depends on HBGAs, we characterized the HBGA phenotype in animal tissues by immunohistochemistry. With the exception of the black-headed gull and the straw-colored fruitbat, we observed the attachment of several human norovirus genotypes to the intestinal epithelium of all tested animal species. However, we did not find an association between the expression of a specific HBGA phenotype and virus-like particle (VLP) attachment. We show that selected human noroviruses can attach to small-intestinal tissues across species, supporting the hypothesis that human noroviruses can reside in an animal reservoir. However, whether this attachment can subsequently lead to infection needs to be further assessed.IMPORTANCE Noroviruses are a major cause of acute gastroenteritis in humans. New norovirus variants and recombinants (re)emerge regularly in the human population. From animal experiments and surveillance studies, it has become clear that at least seven animal models are susceptible to infection with human strains and that domesticated and wild animals shed human noroviruses in their feces. As virus attachment is an important first step for infection, we used a novel method utilizing FITC-labeled VLPs to test for norovirus attachment to intestinal tissues of potential animal hosts. We further characterized these tissues with regard to their HBGA expression, a well-studied norovirus susceptibility factor in humans. We found attachment of several human strains to a variety of animal species independent of their HBGA phenotype. This supports the hypothesis that human strains could reside in an animal reservoir.


Subject(s)
Blood Group Antigens/metabolism , Caliciviridae Infections/virology , Disease Models, Animal , Gastroenteritis/virology , Intestinal Mucosa/virology , Norovirus/physiology , Virus Attachment , Amino Acid Sequence , Animals , Caliciviridae Infections/metabolism , Caliciviridae Infections/pathology , Feces/virology , Gastroenteritis/metabolism , Gastroenteritis/pathology , Humans , Intestinal Mucosa/metabolism , Sequence Homology
11.
J Virol ; 95(3)2021 01 13.
Article in English | MEDLINE | ID: mdl-33177207

ABSTRACT

Noroviruses are a leading cause of gastrointestinal infection in humans and mice. Understanding human norovirus (HuNoV) cell tropism has important implications for our understanding of viral pathogenesis. Murine norovirus (MNoV) is extensively used as a surrogate model for HuNoV. We previously identified CD300lf as the receptor for MNoV. Here, we generated a Cd300lf conditional knockout (CD300lfF/F ) mouse to elucidate the cell tropism of persistent and nonpersistent strains of murine norovirus. Using this mouse model, we demonstrated that CD300lf expression on intestinal epithelial cells (IECs), and on tuft cells in particular, is essential for transmission of the persistent MNoV strain CR6 (MNoVCR6) in vivo In contrast, the nonpersistent MNoV strain CW3 (MNoVCW3) does not require CD300lf expression on IECs for infection. However, deletion of CD300lf in myelomonocytic cells (LysM Cre+) partially reduces CW3 viral load in lymphoid and intestinal tissues. Disruption of CD300lf expression on B cells (CD19 Cre), neutrophils (Mrp8 Cre), and dendritic cells (CD11c Cre) did not affect MNoVCW3 viral RNA levels. Finally, we show that the transcription factor STAT1, which is critical for the innate immune response, partially restricts the cell tropism of MNoVCW3 to LysM+ cells. Taken together, these data demonstrate that CD300lf expression on tuft cells is essential for MNoVCR6; that myelomonocytic cells are a major, but not exclusive, target cell of MNoVCW3; and that STAT1 signaling restricts the cellular tropism of MNoVCW3 This study provides the first genetic system for studying the cell type-specific role of CD300lf in norovirus pathogenesis.IMPORTANCE Human noroviruses (HuNoVs) are a leading cause of gastroenteritis resulting in up to 200,000 deaths each year. The receptor and cell tropism of HuNoV in immunocompetent humans are unclear. We use murine norovirus (MNoV) as a model for HuNoV. We recently identified CD300lf as the sole physiologic receptor for MNoV. Here, we leverage this finding to generate a Cd300lf conditional knockout mouse to decipher the contributions of specific cell types to MNoV infection. We demonstrate that persistent MNoVCR6 requires CD300lf expression on tuft cells. In contrast, multiple CD300lf+ cell types, dominated by myelomonocytic cells, are sufficient for nonpersistent MNoVCW3 infection. CD300lf expression on epithelial cells, B cells, neutrophils, and dendritic cells is not critical for MNoVCW3 infection. Mortality associated with the MNoVCW3 strain in Stat1-/- mice does not require CD300lf expression on LysM+ cells, highlighting that both CD300lf receptor expression and innate immunity regulate MNoV cell tropism in vivo.


Subject(s)
Epithelial Cells/immunology , Host-Pathogen Interactions , Immunity, Innate/immunology , Intestines/immunology , Norovirus/physiology , Receptors, Immunologic/physiology , Viral Tropism , Animals , Caliciviridae Infections/immunology , Caliciviridae Infections/metabolism , Caliciviridae Infections/virology , Epithelial Cells/virology , Female , Intestines/virology , Male , Mice , Mice, Knockout
12.
Viruses ; 12(9)2020 08 31.
Article in English | MEDLINE | ID: mdl-32878241

ABSTRACT

Current knowledge on the role of microRNAs (miRNAs) in rabbit hemorrhagic disease virus (RHDV) infection and the pathogenesis of rabbit hemorrhagic disease (RHD) is still limited. RHDV replicates in the liver, causing hepatic necrosis and liver failure. MiRNAs are a class of short RNA molecules, and their expression profiles vary over the course of diseases, both in the tissue environment and in the bloodstream. This paper evaluates the expression of miRNAs in the liver tissue (ocu-miR-122-5p, ocu-miR-155-5p, and ocu-miR-16b-5p) and serum (ocu-miR-122-5p) of rabbits experimentally infected with RHDV. The expression levels of ocu-miR-122-5p, ocu-miR-155-5p, and ocu-miR-16b-5p in liver tissue were determined using reverse transcription quantitative real-time PCR (RT-qPCR), and the expression level of circulating ocu-miR-122-5p was established using droplet digital PCR (ddPCR). The expression levels of ocu-miR-155-5p and ocu-miR-16b-5p were significantly higher in the infected rabbits compared to the healthy rabbits (a fold-change of 5.8 and 2.5, respectively). The expression of ocu-miR-122-5p was not significantly different in the liver tissue from the infected rabbits compared to the healthy rabbits (p = 0.990), while the absolute expression level of the circulating ocu-miR-122-5p was significantly higher in the infected rabbits than in the healthy rabbits (p < 0.0001). Furthermore, a functional analysis showed that ocu-miR-155-5p, ocu-miR-16b-5p, and ocu-miR-122-5p can regulate the expression of genes involved in processes correlated with acute liver failure (ALF) in rabbits. Search tool for the retrieval of interacting genes/proteins (STRING) analysis showed that the potential target genes of the three selected miRNAs may interact with each other in different pathways. The results indicate the roles of these miRNAs in RHDV infection and over the course of RHD and may reflect hepatic inflammation and impairment/dysfunction in RHD.


Subject(s)
Caliciviridae Infections/genetics , Caliciviridae Infections/virology , Hemorrhagic Disease Virus, Rabbit , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Caliciviridae Infections/metabolism , Female , Gene Expression Regulation , Liver/metabolism , Liver Failure, Acute/genetics , Male , MicroRNAs/blood , Rabbits
13.
J Biol Chem ; 295(47): 15974-15987, 2020 11 20.
Article in English | MEDLINE | ID: mdl-32913124

ABSTRACT

The molecular mechanisms behind infection and propagation of human restricted pathogens such as human norovirus (HuNoV) have defied interrogation because they were previously unculturable. However, human intestinal enteroids (HIEs) have emerged to offer unique ex vivo models for targeted studies of intestinal biology, including inflammatory and infectious diseases. Carbohydrate-dependent histo-blood group antigens (HBGAs) are known to be critical for clinical infection. To explore whether HBGAs of glycosphingolipids contribute to HuNoV infection, we obtained HIE cultures established from stem cells isolated from jejunal biopsies of six individuals with different ABO, Lewis, and secretor genotypes. We analyzed their glycerolipid and sphingolipid compositions and quantified interaction kinetics and the affinity of HuNoV virus-like particles (VLPs) to lipid vesicles produced from the individual HIE-lipid extracts. All HIEs had a similar lipid and glycerolipid composition. Sphingolipids included HBGA-related type 1 chain glycosphingolipids (GSLs), with HBGA epitopes corresponding to the geno- and phenotypes of the different HIEs. As revealed by single-particle interaction studies of Sydney GII.4 VLPs with glycosphingolipid-containing HIE membranes, both binding kinetics and affinities explain the patterns of susceptibility toward GII.4 infection for individual HIEs. This is the first time norovirus VLPs have been shown to interact specifically with secretor gene-dependent GSLs embedded in lipid membranes of HIEs that propagate GII.4 HuNoV ex vivo, highlighting the potential of HIEs for advanced future studies of intestinal glycobiology and host-pathogen interactions.


Subject(s)
Blood Group Antigens/metabolism , Caliciviridae Infections/metabolism , Glycosphingolipids/metabolism , Intestinal Mucosa/metabolism , Norovirus/metabolism , Organoids/metabolism , Virus Attachment , Caliciviridae Infections/pathology , Humans , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Organoids/pathology , Organoids/virology
14.
Food Microbiol ; 92: 103594, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32950136

ABSTRACT

Human noroviruses (HuNoVs) are a main cause of acute gastroenteritis worldwide. They are frequently involved in foodborne and waterborne outbreaks. Environmental transmission of the virus depends on two main factors: the ability of viral particles to remain infectious and their adhesion capacity onto different surfaces. Until recently, adhesion of viral particles to food matrices was mainly investigated by considering non-specific interactions (e.g. electrostatic, hydrophobic) and there was only limited information about infectious HuNoVs because of the absence of a reliable in vitro HuNoV cultivation system. Many HuNoV strains have now been described as having specific binding interactions with human Histo-Blood Group Antigens (HBGAs) and non-HBGA ligands found in food and the environment. Relevant approaches to the in vitro replication of HuNoVs were also proposed recently. On the basis of the available literature data, this review discusses the opportunities to use this new knowledge to obtain a better understanding of HuNoV transmission to human populations and better evaluate the hazard posed by HuNoVs in foodstuffs and the environment.


Subject(s)
Blood Group Antigens/metabolism , Caliciviridae Infections/metabolism , Gastroenteritis/metabolism , Norovirus/metabolism , Animals , Blood Group Antigens/genetics , Caliciviridae Infections/therapy , Caliciviridae Infections/transmission , Caliciviridae Infections/virology , Gastroenteritis/genetics , Gastroenteritis/therapy , Gastroenteritis/virology , Humans , Norovirus/genetics , Norovirus/isolation & purification , Norovirus/physiology , Protein Binding , Viral Proteins/genetics , Viral Proteins/metabolism
15.
PLoS One ; 15(8): e0237044, 2020.
Article in English | MEDLINE | ID: mdl-32745122

ABSTRACT

Human norovirus (HuNoV) is a leading cause of acute gastroenteritis. Outbreaks normally occur via the fecal-oral route. HuNoV infection is thought to occur by viral particle transmission, but increasing evidence suggests a function for exosomes in HuNoV infection. HuNoV is contained within stool-derived exosomes, and exosome-associated HuNoV has been shown to replicate in human intestinal enteroids. In this study, we examine exosome-associated HuNoV infection of Vero cells and show that exosomes containing HuNoV may attach, infect, and be passaged in Vero cells. These findings support earlier findings and have implications for developing HuNoV disease intervention strategies.


Subject(s)
Caliciviridae Infections/metabolism , Caliciviridae Infections/transmission , Exosomes/metabolism , Animals , Caliciviridae Infections/genetics , Child , Child, Preschool , Chlorocebus aethiops , Enterocolitis/virology , Exosomes/genetics , Feces/virology , Female , Gastroenteritis/virology , Humans , Male , Norovirus/pathogenicity , Vero Cells , Virion
16.
Front Immunol ; 11: 961, 2020.
Article in English | MEDLINE | ID: mdl-32612600

ABSTRACT

Human noroviruses (HuNoVs) are the cause of more than 95% of epidemic non-bacterial gastroenteritis worldwide, with some lethal cases. These viral agents affect people of all ages. However, young children and older adults are the highest-risk groups, being affected with the greatest rate of hospitalizations and morbidity cases. HuNoV structural proteins, especially VP1, have been studied extensively. In contrast, the functions of the non-structural proteins of the virus have been undescribed in depth. Studies on HuNoV non-structural proteins have mostly been made by expressing them individually in in vitro cultures, providing insights of their functions and the role that they play in HuNoV replication and pathogenesis. This review examines exhaustively the functions of both HuNoV structural and non-structural proteins and their possible role within the viral replicative cycle and the pathogenesis of the virus. It also highlights recent findings regarding the host's innate and adaptive immune responses against HuNoV, which are of great relevance for diagnostics and vaccine development so as to prevent infections caused by these fastidious viruses.


Subject(s)
Adaptive Immunity , Caliciviridae Infections/virology , Immunity, Innate , Norovirus/pathogenicity , Viral Proteins/metabolism , Virus Replication , Animals , Caliciviridae Infections/immunology , Caliciviridae Infections/metabolism , Host-Pathogen Interactions , Humans , Norovirus/growth & development , Norovirus/immunology , Norovirus/metabolism , Protein Conformation , Structure-Activity Relationship , Viral Proteins/chemistry , Viral Proteins/immunology , Virulence
17.
PLoS One ; 15(4): e0230975, 2020.
Article in English | MEDLINE | ID: mdl-32287278

ABSTRACT

Feline infectious peritonitis (FIP) is a systemic, fatal, viral-induced, immune-mediated disease of cats caused by feline infectious peritonitis virus (FIPV). Mefloquine, a human anti-malarial agent, has been shown to inhibit FIPV in vitro. As a first step to evaluate its efficacy and safety profile as a potential FIP treatment for cats, mefloquine underwent incubation in feline, canine and common brush-tailed possum microsomes and phase I metabolism cofactors to determine its rate of phase I depletion. Tramadol was used as a phase I positive control as it undergoes this reaction in both dogs and cats. Using the substrate depletion method, the in vitro intrinsic clearance (mean ± S.D.) of mefloquine by pooled feline and common brush-tailed possum microsomes was 4.5 ± 0.35 and 18.25 ± 3.18 µL/min/mg protein, respectively. However, phase I intrinsic clearance was too slow to determine with canine microsomes. Liquid chromatography-mass spectrometry (LC-MS) identified carboxymefloquine in samples generated by feline microsomes as well as negative controls, suggesting some mefloquine instability. Mefloquine also underwent incubation with feline, canine and common brush-tailed possum microsomes and phase II glucuronidative metabolism cofactors. O-desmethyltramadol (ODMT or M1) was used as a positive control as it undergoes a phase II glucuronidation reaction in these species. The rates of phase II mefloquine depletion by microsomes by all three species were too slow to estimate. Therefore mefloquine likely undergoes phase I hepatic metabolism catalysed by feline and common brush-tailed possum microsomes but not phase II glucuronidative metabolism in all three species and mefloquine is not likely to have delayed elimination in cats with clinically normal, hepatic function.


Subject(s)
Antimalarials/metabolism , Mefloquine/metabolism , Microsomes, Liver/metabolism , Trichosurus/metabolism , Animals , Antimalarials/pharmacokinetics , Antiviral Agents/metabolism , Antiviral Agents/pharmacokinetics , Caliciviridae Infections/drug therapy , Caliciviridae Infections/metabolism , Caliciviridae Infections/veterinary , Calicivirus, Feline , Cats , Coronavirus, Feline , Dogs , Drug Repositioning/veterinary , Feline Infectious Peritonitis/drug therapy , Feline Infectious Peritonitis/metabolism , Feline Infectious Peritonitis/virology , In Vitro Techniques , Mefloquine/pharmacokinetics , Metabolic Clearance Rate , Species Specificity
18.
J Virol ; 94(10)2020 05 04.
Article in English | MEDLINE | ID: mdl-32161178

ABSTRACT

The rabbit hemorrhagic disease virus (RHDV), which belongs to the family Caliciviridae and the genus Lagovirus, causes lethal fulminant hepatitis in rabbits. RHDV decreases the activity of antioxidant enzymes regulated by Nrf2 in the liver. Antioxidants are important for the maintenance of cellular integrity and cytoprotection. However, the mechanism underlying the regulation of the Nrf2-antioxidant response element (ARE) signaling pathway by RHDV remains unclear. Using isobaric tags for relative and absolute quantification (iTRAQ) technology, the current study demonstrated that RHDV inhibits the induction of ARE-regulated genes and increases the expression of the p50 subunit of the NF-κB transcription factor. We showed that RHDV replication causes a remarkable increase in reactive oxygen species (ROS), which is simultaneously accompanied by a significant decrease in Nrf2. It was found that nuclear translocation of Keap1 plays a key role in the nuclear export of Nrf2, leading to the inhibition of Nrf2 transcriptional activity. The p50 protein partners with Keap1 to form the Keap1-p50/p65 complex, which is involved in the nuclear translocation of Keap1. Moreover, upregulation of Nrf2 protein levels in liver cell nuclei by tert-butylhydroquinone (tBHQ) delayed rabbit deaths due to RHDV infection. Considered together, our findings suggest that RHDV inhibits the Nrf2-dependent antioxidant response via nuclear translocation of Keap1-NF-κB complex and nuclear export of Nrf2 and provide new insight into the importance of oxidative stress during RHDV infection.IMPORTANCE Recent studies have reported that rabbit hemorrhagic disease virus (RHDV) infection reduced Nrf2-related antioxidant function. However, the regulatory mechanisms underlying this process remain unclear. The current study showed that the NF-κB p50 subunit partners with Keap1 to form the Keap1-NF-κB complex, which plays a key role in the inhibition of Nrf2 transcriptional activity. More importantly, upregulated Nrf2 activity delayed the death of RHDV-infected rabbits, strongly indicating the importance of oxidative damage during RHDV infection. These findings may provide novel insights into the pathogenesis of RHDV.


Subject(s)
Antioxidants/metabolism , Caliciviridae Infections/metabolism , Hemorrhagic Disease Virus, Rabbit/immunology , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Animals , Antioxidant Response Elements , Antioxidants/pharmacology , Caliciviridae Infections/immunology , Caliciviridae Infections/pathology , Cell Nucleus/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , HEK293 Cells , Hemorrhagic Disease Virus, Rabbit/pathogenicity , Humans , Hydroquinones , Kelch-Like ECH-Associated Protein 1/genetics , Liver/injuries , Liver/metabolism , Liver/pathology , NF-E2-Related Factor 2/genetics , Oxidative Stress , Proteomics , Rabbits , Signal Transduction/drug effects , Transcription Factor RelA , Virus Replication
19.
J Agric Food Chem ; 68(5): 1207-1212, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-31755264

ABSTRACT

Human noroviruses (HuNoVs) are among the main pathogens causing acute nonbacterial gastroenteritis. Histo-blood group antigens (HBGAs) are widely accepted receptors for HuNoV specific binding. HBGA-like substances in produce are also considered as the critical ligands for capture of HuNoVs. However, the composition of viral ligands from food substrates remains unknown. In this study, an oligosaccharide (H2N2F2) was captured and isolated from romaine lettuce extract by a bacterial surface display system. Using electrospray ionization mass spectrometry and tandem mass spectrometry, it was shown that H2N2F2 was most likely to be a chimera of type A, H, and Lewis a HBGAs. The composition was consistent with our ELISA results using a panel of monoclonal antibodies against HBGAs. Our results revealed a possible interaction mechanism between HuNoVs and romaine lettuce. Better understanding of the interaction of HuNoVs with easily contaminated produce will ultimately aid in the control of and reduction in disease outbreaks.


Subject(s)
Antigens, Plant/metabolism , Blood Group Antigens/metabolism , Lactuca/virology , Norovirus/physiology , Receptors, Virus/metabolism , Virus Attachment , Antigens, Plant/chemistry , Antigens, Plant/genetics , Blood Group Antigens/chemistry , Blood Group Antigens/genetics , Caliciviridae Infections/genetics , Caliciviridae Infections/metabolism , Caliciviridae Infections/virology , Humans , Lactuca/chemistry , Lactuca/genetics , Lactuca/metabolism , Mass Spectrometry , Norovirus/genetics , Oligosaccharides/chemistry , Oligosaccharides/genetics , Oligosaccharides/metabolism , Protein Binding , Receptors, Virus/chemistry , Receptors, Virus/genetics
20.
Viruses ; 11(11)2019 10 30.
Article in English | MEDLINE | ID: mdl-31671627

ABSTRACT

It is known that levels of the anti-apoptotic protein survivin are reduced during Murine norovirus MNV-1 and Feline calicivirus (FCV) infection as part of the apoptosis establishment required for virus release and propagation in the host. Recently, our group has reported that overexpression of survivin causes a reduction of FCV protein synthesis and viral progeny production, suggesting that survivin may affect early steps of the replicative cycle. Using immunofluorescence assays, we observed that overexpression of survivin, resulted in the reduction of FCV infection not only in transfected but also in the neighboring nontransfected CrFK cells, thus suggesting autocrine and paracrine protective effects. Cells treated with the supernatants collected from CrFK cells overexpressing survivin showed a reduction in FCV but not MNV-1 protein production and viral yield, suggesting that FCV binding and/or entry were specifically altered. The reduced ability of FCV to bind to the surface of the cells overexpressing survivin, or treated with the supernatants collected from these cells, correlate with the reduction in the cell surface of the FCV receptor, the feline junctional adhesion molecule (fJAM) 1, while no effect was observed in the cells transfected with the pAm-Cyan vector or in cells treated with the corresponding supernatants. Moreover, the overexpression of survivin affects neither Vaccinia virus (VACV) production in CrFK cells nor MNV-1 virus production in RAW 267.4 cells, indicating that the effect is specific for FCV. All of these results taken together indicate that cells that overexpress survivin, or cell treatment with the conditioned medium from these cells, results in the reduction of the fJAM-1 molecule and, therefore, a specific reduction in FCV entry and infection.


Subject(s)
Caliciviridae Infections/virology , Calicivirus, Feline/physiology , Survivin/metabolism , Animals , Caliciviridae Infections/genetics , Caliciviridae Infections/metabolism , Calicivirus, Feline/metabolism , Cats , Cell Line , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Gene Expression , Host-Pathogen Interactions , Junctional Adhesion Molecules/metabolism , Receptors, Virus/metabolism , Species Specificity , Survivin/genetics , Viral Proteins/biosynthesis , Virus Internalization/drug effects , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...