Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Food Sci ; 86(9): 4110-4118, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33929042

ABSTRACT

Aichi virus (AiV) that results in gastroenteritis worldwide, is spread through contaminated shellfish and water. The resistance/tolerance of AiV to common inactivation processes along with the absence of commercially available vaccines makes it necessary to study its thermal inactivation kinetics. This research evaluated the heat inactivation of AiV in cell-culture media using 2-ml sterile glass vials by the linear and Weibull models. Heat treatments of AiV titers of 7 log plaque forming units (PFU)/ml were conducted thrice in a water-bath at 50, 54, and 58 °C for up to 90 min. Plaque assays for each dilution in duplicate were used to determine infectious virus titers. Linear model D-values for AiV at 50 ± 1 °C (± = standard error) (come-up time = 68 s), 54 ± 0.7 °C (130 s), and 58 ± 0.6°C (251 s) were 43.3 ± 4.23 (R2 = 0.40, RMSE = 0.56), 5.69 ± 0.28 (R2 = 0.80, RMSE = 0.43), and 1.20 ± 0.63 min (R2 = 0.69, RMSE = 0.39), respectively, and the linear model z-value was 5.14 ± 0.39°C (R2 = 0.99, RMSE = 0.08). For the same temperatures, the Weibull model td = 1 values were 20.98 ± 8.8 (R2 = 0.62, RMSE = 0.46, α (scale parameter) = 2.30, ß (shape parameter) = 0.38), 3.84 ± 0.69 (R2 = 0.85, RMSE = 0.38, α = 1.08, ß = 0.66), and 0.87 ± 0.10 min (R2 = 0.80, RMSE = 0.32, α = 0.22, ß = 0.61), respectively and the z-value (using Td = 1 ) was 5.79 ± 0.22 °C (R2 = 1.0, RMSE = 0.03). A better fit was obtained with the Weibull model for log reductions versus time with higher R2 and lower RMSE values. Application of AiV inactivation parameters can help reduce the risk of AiV outbreaks.


Subject(s)
Food Microbiology , Hot Temperature , Kobuvirus , Virus Inactivation , Food Microbiology/methods , Kinetics , Kobuvirus/physiology , Shellfish/virology , Time Factors
2.
Vet Microbiol ; 235: 319-326, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31383319

ABSTRACT

Porcine kobuviruses are widely distributed in swine, but the clinical significance of these viruses remains unclear, since they have been associated with both diarrheic and healthy pigs. In addition, there is a paucity of data on Kobuvirus prevalence in Canadian pig herds. In this study, a total of 181 diarrheic and healthy piglets were monitored and sampled on four occasions, intended to represent the different stages of production. The piglets were sampled at the nursing farms (birth to weaning stage), at the nursery farms (post-weaning stage), and at finishing farms (at the beginning and the end of the fattening stage). Fecal and environmental samples were collected during each life stage. Following viral extraction, Kobuvirus detection by RT-PCR was conducted, and positive samples were sequenced. During the late-nursing stage (6-21 days old), piglets with diarrhea shed more Kobuvirus than healthy individuals. Piglets shed more Kobuvirus during the post-weaning stage (nursery farms) than during any of the other life stages. This was evidenced in individual samples as well as in environmental samples. Over 97% of the sampled piglets shed Kobuvirus at least once in their lifetime. All piglets shedding a Kobuvirus strain or mix of strains at the nursing stage did not appear to shed another porcine kobuvirus strain at a later life stage. Overall, our findings throw light on Kobuvirus shedding dynamics and their potential role in neonatal diarrhea at the nursing stage, which appears to be the point of entry for kobuviruses into swine production systems.


Subject(s)
Diarrhea/veterinary , Kobuvirus/physiology , Picornaviridae Infections/veterinary , Swine Diseases/virology , Virus Shedding , Age Factors , Animals , Diarrhea/virology , Farms , Feces/virology , Kobuvirus/isolation & purification , Phylogeny , Picornaviridae Infections/virology , RNA, Viral , Sequence Analysis, DNA , Swine , Weaning
3.
mSphere ; 4(2)2019 04 03.
Article in English | MEDLINE | ID: mdl-30944213

ABSTRACT

Several viruses encounter various bacterial species within the host and in the environment. Despite these close encounters, the effects of bacteria on picornaviruses are not completely understood. Previous work determined that poliovirus (PV), an enteric virus, has enhanced virion stability when exposed to bacteria or bacterial surface polysaccharides such as lipopolysaccharide. Virion stabilization by bacteria may be important for interhost transmission, since a mutant PV with reduced bacterial binding had a fecal-oral transmission defect in mice. Therefore, we investigated whether bacteria broadly enhance stability of picornaviruses from three different genera: Enterovirus (PV and coxsackievirus B3 [CVB3]), Kobuvirus (Aichi virus), and Cardiovirus (mengovirus). Furthermore, to delineate strain-specific effects, we examined two strains of CVB3 and a PV mutant with enhanced thermal stability. We determined that specific bacterial strains enhance thermal stability of PV and CVB3, while mengovirus and Aichi virus are stable at high temperatures in the absence of bacteria. Additionally, we determined that bacteria or lipopolysaccharide can stabilize PV, CVB3, Aichi virus, and mengovirus during exposure to bleach. These effects are likely mediated through direct interactions with bacteria, since viruses bound to bacteria in a pulldown assay. Overall, this work reveals shared and distinct effects of bacteria on a panel of picornaviruses.IMPORTANCE Recent studies have shown that bacteria promote infection and stabilization of poliovirus particles, but the breadth of these effects on other members of the Picornaviridae family is unknown. Here, we compared the effects of bacteria on four distinct members of the Picornaviridae family. We found that bacteria reduced inactivation of all of the viruses during bleach treatment, but not all viral strains were stabilized by bacteria during heat treatment. Overall, our data provide insight into how bacteria play differential roles in picornavirus stability.


Subject(s)
Bacteria/virology , Hot Temperature , Microbial Interactions , Picornaviridae/physiology , Cardiovirus/genetics , Cardiovirus/physiology , Enterovirus/genetics , Enterovirus/physiology , Kobuvirus/genetics , Kobuvirus/physiology , Mutation , Picornaviridae/genetics , Poliovirus/genetics , Poliovirus/physiology , Sodium Hypochlorite , Virus Inactivation/drug effects
4.
J Virol ; 92(8)2018 04 15.
Article in English | MEDLINE | ID: mdl-29367253

ABSTRACT

Positive-strand RNA viruses, including picornaviruses, utilize cellular machinery for genome replication. Previously, we reported that each of the 2B, 2BC, 2C, 3A, and 3AB proteins of Aichi virus (AiV), a picornavirus, forms a complex with the Golgi apparatus protein ACBD3 and phosphatidylinositol 4-kinase IIIß (PI4KB) at viral RNA replication sites (replication organelles [ROs]), enhancing PI4KB-dependent phosphatidylinositol 4-phosphate (PI4P) production. Here, we demonstrate AiV hijacking of the cellular cholesterol transport system involving oxysterol-binding protein (OSBP), a PI4P-binding cholesterol transfer protein. AiV RNA replication was inhibited by silencing cellular proteins known to be components of this pathway, OSBP, the ER membrane proteins VAPA and VAPB (VAP-A/B), the PI4P-phosphatase SAC1, and PI-transfer protein ß. OSBP, VAP-A/B, and SAC1 were present at RNA replication sites. We also found various previously unknown interactions among the AiV proteins (2B, 2BC, 2C, 3A, and 3AB), ACBD3, OSBP, VAP-A/B, and SAC1, and the interactions were suggested to be involved in recruiting the component proteins to AiV ROs. Importantly, the OSBP-2B interaction enabled PI4P-independent recruitment of OSBP to AiV ROs, indicating preferential recruitment of OSBP among PI4P-binding proteins. Protein-protein interaction-based OSBP recruitment has not been reported for other picornaviruses. Cholesterol was accumulated at AiV ROs, and inhibition of OSBP-mediated cholesterol transfer impaired cholesterol accumulation and AiV RNA replication. Electron microscopy showed that AiV-induced vesicle-like structures were close to ER membranes. Altogether, we conclude that AiV directly recruits the cholesterol transport machinery through protein-protein interactions, resulting in formation of membrane contact sites between the ER and AiV ROs and cholesterol supply to the ROs.IMPORTANCE Positive-strand RNA viruses utilize host pathways to modulate the lipid composition of viral RNA replication sites for replication. Previously, we demonstrated that Aichi virus (AiV), a picornavirus, forms a complex comprising certain proteins of AiV, the Golgi apparatus protein ACBD3, and the lipid kinase PI4KB to synthesize PI4P lipid at the sites for AiV RNA replication. Here, we confirmed cholesterol accumulation at the AiV RNA replication sites, which are established by hijacking the host cholesterol transfer machinery mediated by a PI4P-binding cholesterol transfer protein, OSBP. We showed that the component proteins of the machinery, OSBP, VAP, SAC1, and PITPNB, are all essential host factors for AiV replication. Importantly, the machinery is directly recruited to the RNA replication sites through previously unknown interactions of VAP/OSBP/SAC1 with the AiV proteins and with ACBD3. Consequently, we propose a specific strategy employed by AiV to efficiently accumulate cholesterol at the RNA replication sites via protein-protein interactions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cholesterol/metabolism , Kobuvirus/physiology , Membrane Proteins/metabolism , Models, Biological , Picornaviridae Infections/metabolism , RNA, Viral/metabolism , Receptors, Steroid/metabolism , Vesicular Transport Proteins/metabolism , Viral Proteins/metabolism , Virus Replication/physiology , Adaptor Proteins, Signal Transducing/genetics , Cholesterol/genetics , Humans , Membrane Proteins/genetics , Picornaviridae Infections/genetics , Picornaviridae Infections/pathology , RNA, Viral/genetics , Receptors, Steroid/genetics , Vesicular Transport Proteins/genetics , Viral Proteins/genetics
5.
J Appl Microbiol ; 122(2): 516-521, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27891729

ABSTRACT

AIMS: The aim of this study was to detect and quantify Aichi virus (AiV) in shellfish from three estuaries in Galicia, the main producer of molluscs in Europe. METHODS AND RESULTS: A total of 249 shellfish samples were analysed using a reverse transcription-quantitative PCR procedure. AiV was detected in 15 of 249 (6·02%) samples. Ría de Ares-Betanzos showed the highest prevalence (11·1%), followed by Ría do Burgo (3·7%) and Ría de Vigo, (2·56%). AiV quantifications ranged from nonquantifiable (under the limit of quantification of the method) to 6·9 × 103 RNAc per g DT, with a mean value of 1·9 × 102 RNAc per g DT. CONCLUSION: Results obtained indicated that the prevalence of this enteric virus in the studied area is considerably lower than those of other enteric viruses, such as Norovirus, Sapovirus, HAV or HEV. SIGNIFICANCE AND IMPACT OF THE STUDY: This is the first study that detects the presence of AiV in shellfish from authorized harvesting areas in Spain. Further studies with clinical samples are needed to determine the potential risk of AiV for human health in Galicia.


Subject(s)
Gastroenteritis/virology , Kobuvirus/physiology , Mollusca/virology , Shellfish/virology , Animals , Gastroenteritis/epidemiology , Humans , Prevalence , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spain/epidemiology
6.
Nat Microbiol ; 1(11): 16150, 2016 Sep 05.
Article in English | MEDLINE | ID: mdl-27595320

ABSTRACT

Aichi virus (AiV), an unusual and poorly characterized picornavirus, classified in the genus Kobuvirus, can cause severe gastroenteritis and deaths in children below the age of five years, especially in developing countries1,2. The seroprevalence of AiV is approximately 60% in children under the age of ten years and reaches 90% later in life3,4. There is no available vaccine or effective antiviral treatment. Here, we describe the structure of AiV at 3.7 Å. This first high-resolution structure for a kobuvirus is intermediate between those of the enteroviruses and cardioviruses, with a shallow, narrow depression bounded by the prominent VP0 CD loops (linking the C and D strands of the ß-barrel), replacing the depression known as the canyon, frequently the site of receptor attachment in enteroviruses. VP0 is not cleaved to form VP2 and VP4, so the 'VP2' ß-barrel structure is complemented with a unique extended structure on the inside of the capsid. On the outer surface, a polyproline helix structure, not seen previously in picornaviruses is present at the C terminus of VP1, a position where integrin binding motifs are found in some other picornaviruses. A peptide corresponding to this polyproline motif somewhat attenuates virus infectivity, presumably blocking host-cell attachment. This may guide cellular receptor identification.


Subject(s)
Kobuvirus/chemistry , Kobuvirus/ultrastructure , Receptors, Virus/metabolism , Viral Proteins/chemistry , Virus Attachment , Antigens, Viral/chemistry , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Child , Child, Preschool , Cryoelectron Microscopy , Genome, Viral , Humans , Kobuvirus/genetics , Kobuvirus/physiology , Protein Binding , Protein Conformation
7.
Acta Virol ; 60(1): 55-61, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26982468

ABSTRACT

Piglet diarrhea epidemics result in major economic losses for the swine industry. Four viruses are closely linked to porcine diarrhea: porcine kobuvirus (PKV), porcine epidemic diarrhea virus (PEDV), porcine transmissible gastroenteritis virus (TGEV), and porcine rotavirus (PRoV). We have conducted an epidemiology study to determine the frequency of infection and co-infection with these viruses in China, and characterized the genetic variation of the isolated PEDV and PKV strains. Stool and intestinal samples (n = 314) were collected from piglets with diarrhea in China from years 2012 to 2014. RT-PCR was used to detect PKV, PEDV, TGEV, and PRoV. Phylogenetic relationships between reference strains and the isolated PEDV and PKV strains were determined based on the M and 3D gene sequence. The rates of infection with PKV, PEDV, TGEV and PRoV were 29.9%, 24.2%, 1.91%, and 0.31%, respectively. Co-infections with PKV and the other three viruses were very common. Co-infection of PKV and PEDV was detected in 15.0% (47/314) of the samples. Phylogenetic analysis of the PKV 3D gene indicated that there were some phylogenetic differences in the PKV strains across regions within China. However, according to the PEDV M gene, strains clustered into three groups and the primary group was distinct from the vaccine strain CV777. This study provides insights in to the prevalence of diarrhea viruses and their prevention and control in China.


Subject(s)
Coinfection/veterinary , Diarrhea/veterinary , Kobuvirus/physiology , Porcine epidemic diarrhea virus/physiology , Swine Diseases/virology , Animals , China/epidemiology , Coinfection/epidemiology , Coinfection/virology , Diarrhea/epidemiology , Diarrhea/virology , Feces/virology , Kobuvirus/genetics , Kobuvirus/isolation & purification , Molecular Sequence Data , Phylogeny , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/isolation & purification , Swine , Swine Diseases/epidemiology
8.
Food Environ Virol ; 8(2): 112-9, 2016 06.
Article in English | MEDLINE | ID: mdl-26892338

ABSTRACT

Aqueous Hibiscus sabdariffa extracts possess antimicrobial properties with limited information available on their antiviral effects. Aichi virus (AiV) is an emerging foodborne pathogen that causes gastroenteritis. Vaccines are currently unavailable to prevent their disease transmission. The objective of this study was to determine the antiviral effects of aqueous H. sabdariffa extracts against AiV. AiV at ~5 log PFU/ml was incubated with undiluted (200 mg/ml), 1:1 (100 mg/ml) or 1:5 (40 mg/ml) diluted aqueous hibiscus extract (pH 3.6), phosphate-buffered saline (pH 7.2 as control), or malic acid (pH 3.0, acid control) at 37 °C over 24 h. Treatments were stopped by serially diluting in cell-culture media containing fetal bovine serum and titers were determined using plaque assays on confluent Vero cells. Each treatment was replicated thrice and assayed in duplicate. AiV did not show any significant reduction with 1:1 (100 mg/ml) or 1:5 (40 mg/ml) diluted aqueous hibiscus extracts or malic acid after 0.5, 1, or 2 h at 37 °C. However, AiV titers were reduced to non-detectable levels after 24 h with all the three tested concentrations, while malic acid showed only 0.93 log PFU/ml reduction after 24 h. AiV was reduced by 0.5 and 0.9 log PFU/ml with undiluted extracts (200 mg/ml) after 2 and 6 h, respectively. AiV treated with 1:1 (100 mg/ml) and 1:5 (40 mg/ml) diluted extracts showed a minimal ~0.3 log PFU/ml reduction after 6 h. These extracts show promise to reduce AiV titers mainly through alteration of virus structure, though higher concentrations may have improved effects.


Subject(s)
Antiviral Agents/pharmacology , Hibiscus/chemistry , Kobuvirus/drug effects , Plant Extracts/pharmacology , Animals , Antiviral Agents/isolation & purification , Chlorocebus aethiops , Flowers/chemistry , Kobuvirus/growth & development , Kobuvirus/physiology , Plant Extracts/isolation & purification , Vero Cells
9.
Appl Environ Microbiol ; 80(18): 5743-51, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25015883

ABSTRACT

Human norovirus is the leading cause of epidemic and sporadic acute gastroenteritis. Since no cell culture method for human norovirus exists, cultivable surrogate viruses (CSV), including feline calicivirus (FCV), murine norovirus (MNV), porcine enteric calicivirus (PEC), and Tulane virus (TuV), have been used to study responses to inactivation and disinfection methods. We compared the levels of reduction in infectivities of CSV and Aichi virus (AiV) after exposure to extreme pHs, 56°C heating, alcohols, chlorine on surfaces, and high hydrostatic pressure (HHP), using the same matrix and identical test parameters for all viruses, as well as the reduction of human norovirus RNA levels under these conditions. At pH 2, FCV was inactivated by 6 log10 units, whereas MNV, TuV, and AiV were resistant. All CSV were completely inactivated at 56°C within 20 min. MNV was inactivated 5 log10 units by alcohols, in contrast to 2 and 3 log10 units for FCV and PEC, respectively. TuV and AiV were relatively insensitive to alcohols. FCV was reduced 5 log10 units by 1,000 ppm chlorine, in contrast to 1 log10 unit for the other CSV. All CSV except FCV, when dried on stainless steel surfaces, were insensitive to 200 ppm chlorine. HHP completely inactivated FCV, MNV, and PEC at ≥300 MPa, and TuV at 600 MPa, while AiV was completely resistant to HHP up to 800 MPa. By reverse transcription-quantitative PCR (RT-qPCR), genogroup I (GI) noroviruses were more sensitive than GII noroviruses to alcohols, chlorine, and HHP. Although inactivation profiles were variable for each treatment, TuV and MNV were the most resistant CSV overall and therefore are the best candidates for studying the public health outcomes of norovirus infections.


Subject(s)
Caliciviridae/drug effects , Caliciviridae/radiation effects , Disinfection/methods , Kobuvirus/drug effects , Kobuvirus/radiation effects , Virus Inactivation/drug effects , Virus Inactivation/radiation effects , Caliciviridae/physiology , Disinfectants/pharmacology , Humans , Hydrogen-Ion Concentration , Hydrostatic Pressure , Kobuvirus/physiology , Temperature
10.
J Virol ; 88(12): 6586-98, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24672044

ABSTRACT

UNLABELLED: Phosphatidylinositol 4-kinase IIIß (PI4KB) is a host factor required for the replication of certain picornavirus genomes. We previously showed that nonstructural proteins 2B, 2BC, 2C, 3A, and 3AB of Aichi virus (AiV), a picornavirus, interact with the Golgi protein, acyl-coenzyme A binding domain containing 3 (ACBD3), which interacts with PI4KB. These five viral proteins, ACBD3, PI4KB, and the PI4KB product phosphatidylinositol 4-phosphate (PI4P) colocalize to the AiV RNA replication sites (J. Sasaki et al., EMBO J. 31:754-766, 2012). We here examined the roles of these viral and cellular molecules in the formation of AiV replication complexes. Immunofluorescence microscopy revealed that treatment of AiV polyprotein-expressing cells with a small interfering RNA targeting ACBD3 abolished colocalization of the viral 2B, 2C, and 3A proteins with PI4KB. A PI4KB-specific inhibitor also prevented their colocalization. Virus RNA replication increased the level of cellular PI4P without affecting that of PI4KB, and individual expression of 2B, 2BC, 2C, 3A, or 3AB stimulated PI4P generation. These results suggest that the viral protein/ACBD3/PI4KB complex plays an important role in forming the functional replication complex by enhancing PI4P synthesis. Of the viral proteins, 3A and 3AB were shown to stimulate the in vitro kinase activity of PI4KB through forming a 3A or 3AB/ACBD3/PI4KB complex, whereas the ACBD3-mediated PI4KB activation by 2B and 2C remains to be demonstrated. IMPORTANCE: The phosphatidylinositol 4-kinase PI4KB is a host factor required for the replication of certain picornavirus genomes. Aichi virus, a picornavirus belonging to the genus Kobuvirus, forms a complex comprising one of the viral nonstructural proteins 2B, 2BC, 2C, 3A, and 3AB, the Golgi protein ACBD3, and PI4KB to synthesize PI4P at the sites for viral RNA replication. However, the roles of this protein complex in forming the replication complex are unknown. This study showed that virus RNA replication and individual viral proteins enhance the level of cellular PI4P, and suggested that the viral protein/ACBD3/PI4KB complex plays an important role in forming a functional replication complex. Thus, the present study provides a new example of modulation of cellular lipid metabolism by viruses to support the replication of their genomes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Kobuvirus/physiology , Membrane Proteins/metabolism , Phosphatidylinositol Phosphates/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Picornaviridae Infections/enzymology , Viral Nonstructural Proteins/metabolism , Virus Replication , Adaptor Proteins, Signal Transducing/genetics , Humans , Kobuvirus/genetics , Membrane Proteins/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Picornaviridae Infections/genetics , Picornaviridae Infections/virology , Protein Binding , Protein Transport , Viral Nonstructural Proteins/genetics
11.
mBio ; 4(2): e00098-13, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23572552

ABSTRACT

UNLABELLED: Despite wide sequence divergence, multiple picornaviruses use the Golgi adaptor acyl coenzyme A (acyl-CoA) binding domain protein 3 (ACBD3/GCP60) to recruit phosphatidylinositol 4-kinase class III beta (PI4KIIIß/PI4KB), a factor required for viral replication. The molecular basis of this convergent interaction and the cellular function of ACBD3 are not fully understood. Using affinity purification-mass spectrometry, we identified the putative Rab33 GTPase-activating proteins TBC1D22A and TBC1D22B as ACBD3-interacting factors. Fine-scale mapping of binding determinants within ACBD3 revealed that the interaction domains for TBC1D22A/B and PI4KB are identical. Affinity purification confirmed that PI4KB and TBC1D22A/B interactions with ACBD3 are mutually exclusive, suggesting a possible regulatory mechanism for recruitment of PI4KB. The C-terminal Golgi dynamics (GOLD) domain of ACBD3 has been previously shown to bind the 3A replication protein from Aichi virus. We find that the 3A proteins from several additional picornaviruses, including hepatitis A virus, human parechovirus 1, and human klassevirus, demonstrate an interaction with ACBD3 by mammalian two-hybrid assay; however, we also find that the enterovirus and kobuvirus 3A interactions with ACBD3 are functionally distinct with respect to TBC1D22A/B and PI4KB recruitment. These data reinforce the notion that ACBD3 organizes numerous cellular functionalities and that RNA virus replication proteins likely modulate these interactions by more than one mechanism. IMPORTANCE: Multiple viruses use the same Golgi protein (ACBD3) to recruit the lipid kinase phosphatidylinositol 4-kinase class III beta (PI4KB) in order to replicate. We identify a new binding partner of ACBD3 in the evolutionarily conserved Rab GTPase-activating proteins (RabGAPs) TBC1D22A and -B. Interestingly, TBC1D22A directly competes with PI4KB for binding to the same location of ACBD3 by utilizing a similar binding domain. Different viruses are able to influence this interaction through distinct mechanisms to promote the association of PI4KB with ACBD3. This work informs our knowledge of both the physical interactions of the proteins that help maintain metazoan Golgi structure and how viruses subvert these evolutionarily conserved interactions for their own purposes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Enterovirus/physiology , GTPase-Activating Proteins/metabolism , Kobuvirus/physiology , Membrane Proteins/metabolism , Viral Proteins/metabolism , Chromatography, Affinity , Humans , Mass Spectrometry , Protein Binding , Protein Interaction Mapping , Two-Hybrid System Techniques
12.
Virus Res ; 163(2): 592-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22226945

ABSTRACT

Picornavirus genomes are translated into a single large polyprotein, which is processed by virus-encoded proteases into individual functional proteins. 3C of all picornaviruses is a protease, and the leader (L) and 2A proteins of some picornaviruses are also involved in polyprotein processing. Aichi virus (AiV), which is associated with acute gastroenteritis in humans, is a member of the genus Kobuvirus of the family Picornaviridae. The AiV L and 2A proteins have already been shown to exhibit no protease activity. In this study, we investigated AiV polyprotein processing by 3C and 3CD using a cell-free translation system. 3C and 3CD were capable of processing the polyprotein in trans; 3C, however, cleaved the VP1/2A site inefficiently, while 3CD cleaved this site almost completely. Mammalian two-hybrid and coimmunoprecipitation assays showed an interaction between 2A and 3CD. Using a 3CD mutant and various 2A mutants of substrate proteins, we showed a clear correlation between the 2A-3CD interaction and the VP1/2A cleavage by 3CD. Thus, this study suggests that tight interaction of 3CD with the 2A region of a precursor protein is required for efficient cleavage at the VP1/2A site.


Subject(s)
Kobuvirus/physiology , Polyproteins/metabolism , Protein Processing, Post-Translational , Viral Proteins/metabolism , Virus Replication , Immunoprecipitation , Protein Binding , Protein Interaction Mapping , Protein Multimerization , Two-Hybrid System Techniques
13.
Rev Med Virol ; 21(1): 32-41, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21294214

ABSTRACT

Kobuviruses are members of the large and growing family Picornaviridae. Until now, two official, Aichi virus and Bovine kobuvirus, and one candidate kobuvirus species, 'porcine kobuvirus', have been identified in human, cattle and swine, respectively. In addition, kobu-like viruses were detected very recently in the bat. Aichi virus could be one of the causative agents of gastroenteritis in humans, and kobuviruses probably also cause diarrhoea in cattle and swine. Although Aichi virus has been detected relatively infrequently (0-3%) in human diarrhoea, high seroprevalence, up to 80-95% at the age of 30-40, was found indicating the general nature of infection in different human populations. In the previous years, much new information has accumulated relating to kobuviruses and their host species. This review summarises the current knowledge on kobuviruses including taxonomy, biology and viral characteristics, and covers all aspects of infection including epidemiology, clinical picture, host species diversity, laboratory diagnosis and it gives a summary about possible future perspectives.


Subject(s)
Kobuvirus , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Animals , Cattle , Host Specificity , Humans , Kobuvirus/classification , Kobuvirus/genetics , Kobuvirus/physiology , Picornaviridae Infections/epidemiology , Picornaviridae Infections/transmission , Swine
14.
Environ Sci Technol ; 44(7): 2426-32, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20205469

ABSTRACT

Atomic force microscopy (AFM) was used to study the attachment of phiX174, MS2, and Aichi viruses on sands of different surface properties: oxide-removed (clean), goethite-coated, and aluminum oxide-coated. Interaction forces between viruses and sand surfaces were measured by contact mode AFM using tips coated with particles of each virus. Column experiments were conducted to quantify the macroscopic transport and retention of the viruses in sand. The average adhesion force measured with AFM was highest between aluminum oxide-coated sand and all three viruses, followed by goethite-coated sand, and was significantly lower on oxide-removed sand. Among the viruses, adhesion on goethite-coated and aluminum oxide-coated sands followed the order of MS2 > Aichi > phiX174, and on oxide-removed sand it was phiX174 > Aichi > MS2. Column breakthrough results revealed the same retention trend, which was completely consistent with AFM force measurements. Strong electrostatic attraction and, to a lesser extent, hydrophobic interactions are responsible for the much greater removal of all three viruses observed in the oxide-coated sands compared to the oxide-removed sand. Mass recovery data indicate that the removal of phiX174, MS2, and Aichi was largely reversible when eluted with 3% beef extract solution at pH 9.5. The Derjaguin-Landau-Verwey-Overbeek (DLVO) and extended DLVO theories provided correct qualitative predictions on the deposition trend observed in the experiments. This study, to the best of our knowledge, was the first to employ AFM to directly measure interaction forces between viruses and solid surfaces; and it was the first to evaluate the retention and transport behavior of Aichi virus, a human pathogen.


Subject(s)
Bacteriophage phi X 174/physiology , Kobuvirus/physiology , Levivirus/physiology , Microscopy, Atomic Force/methods , Silicon Dioxide/metabolism , Virus Attachment , Microscopy, Confocal , Movement
15.
Virus Res ; 147(1): 77-84, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19879907

ABSTRACT

Aichi virus (AiV), which is associated with acute gastroenteritis in humans, is a member of the genus Kobuvirus of the family Picornaviridae. Picornavirus genome replication occurs in replication complexes that include viral nonstructural proteins, host proteins and viral RNA. In poliovirus, all nonstructural proteins are found in the replication complexes, suggesting the ability of the viral nonstructural proteins to interact with each other. In this study, we examined the interactions between the AiV nonstructural proteins using a mammalian two-hybrid system. The results showed that all of the tested proteins could interact with more than one protein. We observed homodimerization of five proteins, bidirectional heterodimerization of six protein pairs, and unidirectional heterodimerization of eighteen protein pairs. Among the interactions detected in this study, the 2A-2BC, 2A-2BC, 2A-2C, 2BC-3CD, 2BC-3C, 2C-3C, 2C-3CD and 3AB-3C interactions have not been observed in the previous two-hybrid studies with other picornaviruses. The strongest interaction was observed between 2A and 3CD. AiV 2A has already been shown to be involved in genome replication. Domain mapping of the 2A and 3CD interaction in mammalian two-hybrid analysis revealed that the C-terminal quarter of 2A is not required for the interaction with 3CD.


Subject(s)
Kobuvirus/physiology , Protein Interaction Mapping , Viral Nonstructural Proteins/metabolism , Animals , Chlorocebus aethiops , Protein Binding , Protein Multimerization , Two-Hybrid System Techniques , Vero Cells
16.
Uirusu ; 57(1): 67-74, 2007 Jun.
Article in Japanese | MEDLINE | ID: mdl-18040156

ABSTRACT

Aichi virus is a member of the Family Picornaviridae. This virus was first isolated in 1989 from a stool specimen from a patient with oyster-associated gastroenteritis in Aichi, Japan. We analyzed the function of the 5' terminal region of the genome and the leader protein in virus replication. The results indicate that both the 5' terminal region of the genome and the leader protein are involved in viral RNA replication and encapsidation.


Subject(s)
Kobuvirus/genetics , Kobuvirus/physiology , Virus Replication/genetics , 5' Untranslated Regions/physiology , Genome, Viral/genetics , Genome, Viral/physiology , Humans , RNA, Viral/genetics , Viral Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...