Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 753683, 2021.
Article in English | MEDLINE | ID: mdl-34899705

ABSTRACT

Human parechovirus type 3 (PeV-A3) infection has been recognized as an emerging etiologic factor causing severe nerve disease or sepsis in infants and young children. But the neuropathogenic mechanisms of PeV-A3 remain unknown. To understand the pathogenesis of PeV-A3 infection in the neuronal system, PeV-A3-mediated cytopathic effects were analyzed in human glioblastoma cells and neuroblastoma cells. PeV-A3 induced interferons and inflammatory cytokine expression in these neuronal cells. The pronounced cytopathic effects accompanied with activation of death signaling pathways of apoptosis, autophagy, and pyroptosis were detected. A new experimental disease model of parechovirus encephalitis was established. In the disease model, intracranial inoculation with PeV-A3 in C57BL/6 neonatal mice showed body weight loss, hindlimb paralysis, and approximately 20% mortality. PeV-A3 infection in the hippocampus and cortex regions of the neonatal mouse brain was revealed. Mechanistic assay supported the in vitro results, indicating detection of PeV-A3 replication, inflammatory cytokine expression, and death signaling transduction in mouse brain tissues. These in vitro and in vivo studies revealed that the activation of death signaling and inflammation responses is involved in PeV-A3-mediated neurological disorders. The present results might account for some of the PeV-A3-associated clinical manifestations.


Subject(s)
Cytopathogenic Effect, Viral , Disease Models, Animal , Encephalitis, Viral/metabolism , Parechovirus/pathogenicity , Picornaviridae Infections/metabolism , Animals , Animals, Newborn , Apoptosis , Autophagy , Cell Line, Tumor , Cerebral Cortex/virology , Chlorocebus aethiops , Cytokines/biosynthesis , Cytokines/genetics , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Glioblastoma/pathology , Hippocampus/virology , Humans , Inflammation , Interferon Type I/biosynthesis , Interferon Type I/genetics , Interferon Type I/pharmacology , Interferons/biosynthesis , Interferons/genetics , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Neuroblastoma/pathology , Parechovirus/drug effects , Parechovirus/physiology , Picornaviridae Infections/pathology , Picornaviridae Infections/virology , Pyroptosis , Vero Cells , Virus Replication/drug effects , Interferon Lambda
2.
Int J Infect Dis ; 89: 175-178, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31626981

ABSTRACT

The majority of parechovirus A type 5 (PeV-A5) infections have been reported in patients with gastrointestinal syndromes. In contrast, a sepsis-like illness associated with PeV-A5 infection has been reported only anecdotally. Herein, we report the first case in Italy of a PeV-A5 neurological infection presenting in a neonate with a sepsis-like syndrome. The patient, a healthy male infant born at 41 weeks of gestation, was highly distressed and inconsolable, and had been crying persistently, with poor breastfeeding, since the previous day. From day 2 to day 4, the newborn was feverish with mild irritability; breastfeeding was preserved and regularly supported. His clinical condition progressively improved, with defervescence on day 4. He was discharged after 7 days, and neurological examination results indicated only mild impairment in visual fixation and vertical eye tracking and mild axial hypotonia. The Italian PeV-A5 strain was phylogenetically related to three strains detected in Denmark in 2012, as well as to one detected in Australia and one in Greece in 2015, with an average nucleotide identity of 97.9% (range 95.9-100.0%). Enterovirus/PeV infection in the newborn should be ruled out in cases of infants with unexplained fever and/or a sepsis-like syndrome and/or meningoencephalitis. An aetiological diagnosis is essential to avoid the unnecessary administration of antibiotics and to plan long-term follow-up until schooling.


Subject(s)
Infant, Newborn, Diseases/virology , Nervous System Diseases/virology , Parechovirus/isolation & purification , Picornaviridae Infections/virology , Humans , Infant , Infant, Newborn , Italy , Male , Nervous System Diseases/diagnosis , Parechovirus/classification , Parechovirus/genetics , Parechovirus/physiology , Phylogeny
3.
Pediatr Infect Dis J ; 38(1): 1-5, 2019 01.
Article in English | MEDLINE | ID: mdl-30204658

ABSTRACT

BACKGROUND: Human parechovirus particularly genotype 3 (HPeV3) is an emerging infection affecting predominantly young infants. The potential for neurologic sequelae in a vulnerable subset is increasingly apparent. A review of 2 epidemics of human parechovirus (HpeV) infection in 2013 and in 2015 in Queensland, Australia, was undertaken, with an emphasis on identifying adverse neurodevelopmental outcome. METHODS: All hospitalized cases with laboratory-confirmed HPeV infection between October 2013 June 2016 were identified. Clinical, demographic, laboratory and imaging data were collected and correlated with reported developmental outcome. RESULTS: Laboratory-confirmed HPeV infections were identified in 202 patients across 25 hospitals; 86.6% (n = 175) were younger than 3 months 16.3% (n = 33) received intensive care admission. Of 142 cerebrospinal fluid samples which were HPeV polymerase chain reaction positive, all 89 isolates successfully genotyped were HPeV3. Clinical information was available for 145 children; 53.1% (n = 77) had follow-up from a pediatrician, of whom 14% (n = 11) had neurodevelopmental sequelae, ranging from hypotonia and gross motor delay to spastic quadriplegic cerebral palsy and cortical visual impairment. Of 15 children with initially abnormal brain magnetic resonance imaging, 47% (n = 7) had neurodevelopmental concerns, the remainder had normal development at follow-up between 6 and 15 months of age. CONCLUSIONS: This is the largest cohort of HPeV3 cases with clinical data and pediatrician-assessed neurodevelopmental follow-up to date. Developmental concerns were identified in 11 children at early follow-up. Abnormal magnetic resonance imaging during acute infection did not specifically predict poor neurodevelopmental in short-term follow-up. Continued follow-up of infants and further imaging correlation is needed to explore predictors of long-term morbidity.


Subject(s)
Genotype , Neurodevelopmental Disorders/virology , Picornaviridae Infections/complications , Picornaviridae Infections/epidemiology , Australia/epidemiology , Cohort Studies , Female , Hospitalization/statistics & numerical data , Humans , Infant , Infant, Newborn , Male , Neurodevelopmental Disorders/epidemiology , Parechovirus/genetics , Parechovirus/physiology , Picornaviridae Infections/cerebrospinal fluid , Queensland/epidemiology , Sepsis/epidemiology , Sequence Analysis, DNA
4.
Antiviral Res ; 162: 51-53, 2019 02.
Article in English | MEDLINE | ID: mdl-30550798

ABSTRACT

Parechoviruses A (HPeV, Picornaviridae) are neglected human pathogens that cause sepsis-like illness and severe neurological complications in infants. There are no antivirals available for the treatment of HPeV infections. We here report on cell-based assays that allow for medium-throughput antiviral screening of compound libraries against HPeV. The nucleoside viral polymerase inhibitor 2'-C-methylcytidine was identified as being an in vitro replication inhibitor of HPeV1 and HPeV3 that can serve as a reference molecule for further antiviral studies.


Subject(s)
Antiviral Agents/pharmacology , Nucleosides/pharmacology , Parechovirus/drug effects , A549 Cells , Animals , Antiviral Agents/chemistry , Chlorocebus aethiops , HeLa Cells , High-Throughput Screening Assays , Humans , Mice , Parechovirus/physiology , Vero Cells , Virus Replication/drug effects
5.
Article in English | MEDLINE | ID: mdl-30211126

ABSTRACT

Human parechoviruses (HPeVs), a poorly studied genus within the Picornaviridae family, are classified into 19 genotypes of which HPeV1 and HPeV3 are the most often detected. HPeV1 VP1 C terminus contains an arginine-glycine-aspartic acid (RGD) motif and has been shown to depend on the host cell surface αV integrins (αV ITGs) and heparan sulfate (HS) for entry. HPeV3 lacks this motif and the receptors remain unknown. HPeVs can be detected in patient nasopharyngeal and stool samples, and infection is presumed to occur after respiratory or gastro-intestinal transmission. HPeV pathogenesis is poorly understood as there are no animal models and previous studies have been conducted in immortalized monolayer cell cultures which do not adequately represent the characteristics of human tissues. To bridge this gap, we determined the polarity of infection, replication kinetics, and cell tropism of HPeV1 and HPeV3 in the well-differentiated human airway epithelial (HAE) model. We found the HAE cultures to be permissive for HPeVs. Both HPeV genotypes infected the HAE preferentially from the basolateral surface while the progeny virus was shed toward the apical side. Confocal microscopy revealed the target cell type to be the p63+ basal cells for both viruses, αV ITG and HS blocking had no effect on the replication of either virus, and transcriptional profiling suggested that HPeV3 infection induced stronger immune activation than HPeV1. Genotype-specific host responses may contribute to the differences in pathogenesis and clinical outcomes associated with HPeV1 and HPeV3.


Subject(s)
Epithelial Cells/virology , Epithelium/virology , Parechovirus/physiology , Picornaviridae Infections/virology , Virus Internalization , Cell Polarity , Cells, Cultured , Humans , Microscopy, Confocal , Models, Theoretical , Viral Tropism , Virus Release , Virus Replication
6.
Sci Rep ; 8(1): 5820, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29643409

ABSTRACT

Human parechoviruses (HPeV) are picornaviruses with a highly-ordered RNA genome contained within icosahedrally-symmetric capsids. Ordered RNA structures have recently been shown to interact with capsid proteins VP1 and VP3 and facilitate virus assembly in HPeV1. Using an assay that combines reversible cross-linking, RNA affinity purification and peptide mass fingerprinting (RCAP), we mapped the RNA-interacting regions of the capsid proteins from the whole HPeV1 virion in solution. The intrinsically-disordered N-termini of capsid proteins VP1 and VP3, and unexpectedly, VP0, were identified to interact with RNA. Comparing these results to those obtained using recombinantly-expressed VP0 and VP1 confirmed the virion binding regions, and revealed unique RNA binding regions in the isolated VP0 not previously observed in the crystal structure of HPeV1. We used RNA fluorescence anisotropy to confirm the RNA-binding competency of each of the capsid proteins' N-termini. These findings suggests that dynamic interactions between the viral RNA and the capsid proteins modulate virus assembly, and suggest a novel role for VP0.


Subject(s)
Capsid Proteins/metabolism , Parechovirus/physiology , RNA, Viral/metabolism , Virion/metabolism , Virus Assembly , Capsid Proteins/chemistry , Cross-Linking Reagents/chemistry , HT29 Cells , Humans , Models, Molecular , RNA, Viral/chemistry
7.
Sci Rep ; 7(1): 12075, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28935894

ABSTRACT

Human parechovirus 3 (HPeV3), a member of the Picornavirus family, is frequently detected worldwide. However, the observed seropositivity rates for HPeV3 neutralizing antibodies (nAbs) vary from high in Japan to low in the Netherlands and Finland. To study if this can be explained by technical differences or antigenic diversity among HPeV3 strains included in the serological studies, we determined the neutralizing activity of Japanese and Dutch intravenous immunoglobulin batches (IVIG), a rabbit HPeV3 hyperimmune polyclonal serum, and a human HPeV3-specific monoclonal antibody (mAb) AT12-015, against the HPeV3 A308/99 prototype strain and clinical isolates from Japan, the Netherlands and Australia, collected between 1989 and 2015. The rabbit antiserum neutralized all HPeV3 isolates whereas the neutralization capacity of the IVIG batches varied, and the mAb exclusively neutralized the A308/99 strain. Mapping of the amino acid variation among a subset of the HPeV3 strains on an HPeV3 capsid structure revealed that the majority of the surface-exposed amino acid variation was located in the VP1. Furthermore, amino acid mutations in a mAb AT12-015-resistant HPeV3 A308/99 variant indicated the location for potential antigenic determinants. Virus aggregation and the observed antigenic diversity in HPeV3 can explain the varying levels of nAb seropositivity reported in previous studies.


Subject(s)
Antibodies, Neutralizing/immunology , Antigenic Variation/immunology , Capsid Proteins/immunology , Parechovirus/immunology , Picornaviridae Infections/immunology , Amino Acid Sequence , Animals , Antibodies, Neutralizing/genetics , Antigenic Variation/genetics , Capsid Proteins/genetics , Humans , Immune Sera/immunology , Japan , Mutation , Netherlands , Neutralization Tests , Parechovirus/classification , Parechovirus/physiology , Picornaviridae Infections/virology , Rabbits , Sequence Homology, Amino Acid , Species Specificity
8.
Nat Commun ; 7: 11387, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27435188

ABSTRACT

The poorly studied picornavirus, human parechovirus 3 (HPeV3) causes neonatal sepsis with no therapies available. Our 4.3-Å resolution structure of HPeV3 on its own and at 15 Å resolution in complex with human monoclonal antibody Fabs demonstrates the expected picornavirus capsid structure with three distinct features. First, 25% of the HPeV3 RNA genome in 60 sites is highly ordered as confirmed by asymmetric reconstruction, and interacts with conserved regions of the capsid proteins VP1 and VP3. Second, the VP0 N terminus stabilizes the capsid inner surface, in contrast to other picornaviruses where on expulsion as VP4, it forms an RNA translocation channel. Last, VP1's hydrophobic pocket, the binding site for the antipicornaviral drug, pleconaril, is blocked and thus inappropriate for antiviral development. Together, these results suggest a direction for development of neutralizing antibodies, antiviral drugs based on targeting the RNA-protein interactions and dissection of virus assembly on the basis of RNA nucleation.


Subject(s)
Capsid/metabolism , Neonatal Sepsis/virology , Parechovirus/physiology , Picornaviridae Infections/virology , Amino Acid Sequence , Capsid/chemistry , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Parechovirus/chemistry , Parechovirus/genetics , Protein Binding , Protein Conformation , Sequence Alignment , Virus Assembly
9.
PLoS One ; 11(4): e0154769, 2016.
Article in English | MEDLINE | ID: mdl-27128974

ABSTRACT

Human parechovirus 1 (HPeV-1) (family Picornaviridae) is a global cause of pediatric respiratory and CNS infections for which there is no treatment. Although biochemical and in vitro studies have suggested that HPeV-1 binds to αVß1, αVß3 and αVß6 integrin receptor(s), the actual cellular receptors required for infectious entry of HPeV-1 remain unknown. In this paper we analyzed the expression profiles of αVß1, αVß3, αVß6 and α5ß1 in susceptible cell lines (A549, HeLa and SW480) to identify which integrin receptors support HPeV-1 internalization and/or replication cycle. We demonstrate by antibody blocking assay, immunofluorescence microscopy and RT-qPCR that HPeV-1 internalizes and replicates in cell lines that express αVß1 integrin but not αVß3 or αVß6 integrins. To further study the role of ß1 integrin, we used a mouse cell line, GE11-KO, which is deficient in ß1 expression, and its derivate GE11-ß1 in which human integrin ß1 subunit is overexpressed. HPeV-1 (Harris strain) and three clinical HPeV-1 isolates did not internalize into GE11-KO whereas GE11-ß1 supported the internalization process. An integrin ß1-activating antibody, TS2/16, enhanced HPeV-1 infectivity, but infection occurred in the absence of visible receptor clustering. HPeV-1 also co-localized with ß1 integrin on the cell surface, and HPeV-1 and ß1 integrin co-endocytosed into the cells. In conclusion, our results demonstrate that in some cell lines the cellular entry of HPeV-1 is primarily mediated by the active form of αVß1 integrin without visible receptor clustering.


Subject(s)
Parechovirus/pathogenicity , Picornaviridae Infections/etiology , Receptors, Vitronectin/physiology , Virus Internalization , Animals , Antigens, Neoplasm/physiology , Cell Line , Cell Line, Tumor , HeLa Cells , Humans , Integrin alphaVbeta3/physiology , Integrins/physiology , Mice , Parechovirus/physiology , Picornaviridae Infections/physiopathology , Picornaviridae Infections/virology , Receptors, Virus/physiology
10.
Nat Commun ; 6: 8316, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26446437

ABSTRACT

Picornaviruses are responsible for a range of human and animal diseases, but how their RNA genome is packaged remains poorly understood. A particularly poorly studied group within this family are those that lack the internal coat protein, VP4. Here we report the atomic structure of one such virus, Ljungan virus, the type member of the genus Parechovirus B, which has been linked to diabetes and myocarditis in humans. The 3.78-Å resolution cryo-electron microscopy structure shows remarkable features, including an extended VP1 C terminus, forming a major protuberance on the outer surface of the virus, and a basic motif at the N terminus of VP3, binding to which orders some 12% of the viral genome. This apparently charge-driven RNA attachment suggests that this branch of the picornaviruses uses a different mechanism of genome encapsidation, perhaps explored early in the evolution of picornaviruses.


Subject(s)
Nucleic Acid Conformation , Parechovirus/physiology , RNA, Viral/physiology , Animals , Cell Line , Cryoelectron Microscopy , DNA, Complementary , RNA, Viral/chemistry
11.
Viruses ; 7(8): 4529-62, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26266417

ABSTRACT

The Enterovirus (EV) and Parechovirus genera of the picornavirus family include many important human pathogens, including poliovirus, rhinovirus, EV-A71, EV-D68, and human parechoviruses (HPeV). They cause a wide variety of diseases, ranging from a simple common cold to life-threatening diseases such as encephalitis and myocarditis. At the moment, no antiviral therapy is available against these viruses and it is not feasible to develop vaccines against all EVs and HPeVs due to the great number of serotypes. Therefore, a lot of effort is being invested in the development of antiviral drugs. Both viral proteins and host proteins essential for virus replication can be used as targets for virus inhibitors. As such, a good understanding of the complex process of virus replication is pivotal in the design of antiviral strategies goes hand in hand with a good understanding of the complex process of virus replication. In this review, we will give an overview of the current state of knowledge of EV and HPeV replication and how this can be inhibited by small-molecule inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Enterovirus/drug effects , Enterovirus/physiology , Parechovirus/drug effects , Parechovirus/physiology , Virus Replication/drug effects , Animals , Humans
12.
PLoS One ; 10(2): e0116158, 2015.
Article in English | MEDLINE | ID: mdl-25646764

ABSTRACT

Human parechoviruses (HPeVs), members of the family Picornaviridae, are associated with severe human clinical conditions such as gastrointestinal disease, encephalitis, meningitis, respiratory disease and neonatal sepsis. A new contemporary strain of HPeV1, KVP6 (accession no. KC769584), was isolated from a clinical specimen. Full-genome alignment revealed that HPeV1 KVP6 shares high genome homology with the German strain of HPeV1, 7555312 (accession no. FM178558) and could be classified in the clade 1B group. An intertypic recombination was shown within the P2-P3 genome regions of HPeV1. Cell-type tropism test showed that T84 cells (colon carcinoma cells), A549 cells (lung carcinoma cells) and DBTRG-5MG cells (glioblastoma cells) were susceptible to HPeV1 infection, which might be relevant clinically. A facilitated cytopathic effect and increased viral titers were reached after serial viral passages in Vero cells, with viral genome mutation found in later passages. HPeV1 is sensitive to elevated temperature because 39C incubation impaired virion production. HPeV1 induced innate immunity with phosphorylation of interferon (IFN) regulatory transcription factor 3 and production of type I IFN in A549 but not T84 cells. Furthermore, type I IFN inhibited HPeV1 production in A549 cells but not T84 cells; T84 cells may be less responsive to type I IFN stimulation. Moreover, HPeV1-infected cells showed downregulated type I IFN activation, which indicated a type I IFN evasion mechanism. The characterization of the complete genome and infection features of HPeV1 provide comprehensive information about this newly isolated HPeV1 for further diagnosis, prevention or treatment strategies.


Subject(s)
Antiviral Agents/pharmacology , Genome, Viral/genetics , Interferon Type I/pharmacology , Parechovirus/genetics , Parechovirus/physiology , Picornaviridae Infections , Animals , Antiviral Agents/metabolism , Cell Line , Genomics , Humans , Interferon Type I/metabolism , Kinetics , Molecular Sequence Data , Parechovirus/drug effects , Signal Transduction , Temperature , Viral Tropism/drug effects , Virus Replication/drug effects
13.
PLoS One ; 8(11): e78377, 2013.
Article in English | MEDLINE | ID: mdl-24265685

ABSTRACT

Human parechoviruses are known to cause asymptomatic to severe clinical illness predominantly respiratory and gastroenetric infections. Despite their global prevalence, epidemiological studies have not been performed in Pakistan. In this study, we retrospectively analyzed 110 fecal specimen and found 26 (24%) positive for viral RNA with HPeV-10 (n = 3, 23%), HPeV-13 (n = 4, 31%) and HPeV-15 (n = 6, 46%) genotypes. Clinical features of patients with different HPeV genotypes were compared. All HPeV positive children were aged ≤4 years (mean 13.92 months). The male-to-female ratio was 1: 1.17 (46.2 vs 53.8%) with significant association (p = .031) to HPeV infectivity. HPeV-10 and -13 were found during summer while HPeV-15 was only detected during late winter season. Disease symptoms were more severe in children infected with HPeV-10 and -13 as compared to HPeV-15. Fever and vomiting were observed in 100% cases of HPeV-10 and -13 while only 17% patients of HPeV-15 had these complaints. Phylogenetic analyses showed that HPeV-10, -13 and -15 strains found in this study have 9-13%, 16.8% and 21.8% nucleotide divergence respectively from the prototype strains and were clustered to distinct genetic lineages. This is the first report of HPeV-15 infection in humans although first identified in rhesus macaques. The arginine-glycine-aspartic acid (RGD) motif present at the C-terminal of VP1 responsible for the viral attachment to cellular integrins was not found in all of these strains. In conclusion, these findings enhance our knowledge related to the epidemiology and genetic diversity of the HPeV in Pakistan and support the need for continued laboratory based surveillance programs especially in infants and neonatal clinical settings. Further, the parechovirus pathogenesis, cross-species transmission and disease reservoirs must be ascertained to adopt better prevention measures.


Subject(s)
Dehydration/complications , Gastroenteritis/complications , Gastroenteritis/virology , Genotype , Parechovirus/genetics , Parechovirus/physiology , Acute Disease/epidemiology , Child, Preschool , Feces/virology , Female , Gastroenteritis/epidemiology , Humans , Infant , Male , Pakistan/epidemiology , Parechovirus/classification , Phylogeny , Retrospective Studies
14.
Virol J ; 9: 247, 2012 Oct 27.
Article in English | MEDLINE | ID: mdl-23102237

ABSTRACT

Acute respiratory infections are a major cause of morbidity in children both in developed and developing countries. A wide range of respiratory viruses, including respiratory syncytial virus (RSV), influenza A and B viruses, parainfluenza viruses (PIVs), adenovirus, rhinovirus (HRV), have repeatedly been detected in acute lower respiratory tract infections (LRTI) in children in the past decades. However, in the last ten years thanks to progress in molecular technologies, newly discovered viruses have been identified including human Metapneumovirus (hMPV), coronaviruses NL63 (HcoV-NL63) and HKU1 (HcoV-HKU1), human Bocavirus (HBoV), new enterovirus (HEV), parechovirus (HpeV) and rhinovirus (HRV) strains, polyomaviruses WU (WUPyV) and KI (KIPyV) and the pandemic H1N1v influenza A virus. These discoveries have heavily modified previous knowledge on respiratory infections mainly highlighting that pediatric population is exposed to a variety of viruses with similar seasonal patterns. In this context establishing a causal link between a newly identified virus and the disease as well as an association between mixed infections and an increase in disease severity can be challenging. This review will present an overview of newly recognized as well as the main emerging respiratory viruses and seek to focus on the their contribution to infection and co-infection in LRTIs in childhood.


Subject(s)
Respiratory Tract Infections/virology , Bocavirus/physiology , Child , Coinfection , Coronavirus/physiology , Coronavirus Infections/virology , Enterovirus/physiology , Enterovirus Infections/virology , Humans , Metapneumovirus/physiology , Paramyxoviridae Infections/virology , Parechovirus/physiology , Parvoviridae Infections/virology , Picornaviridae Infections/virology , Polyomavirus/physiology , Polyomavirus Infections/virology , Rhinovirus/physiology , Tumor Virus Infections/virology
15.
J Gen Virol ; 93(Pt 11): 2363-2370, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22837420

ABSTRACT

Human parechoviruses (HPeVs) are picornaviruses frequently infecting humans. While HPeV1 is associated with mild disease, HPeV3 is the cause of neonatal sepsis and meningitis. To test whether in vitro replication kinetics of HPeV1 and HPeV3 could be related to pathogenicity, HPeV1 and HPeV3 strains isolated from patients were cultured on cell lines of gastrointestinal, respiratory and neural origin, and replication kinetics were measured by real-time PCR. No relationship was found between clinical symptoms and in vitro replication of the HPeV1 strains. In contrast, the HPeV3 strains showed faster replication in neural cells and there was a relationship between higher in vitro replication kinetics and neuropathogenicity in the patient. Furthermore, HPeV1 could be neutralized efficiently by its specific antibody and by intravenous immunoglobulins (IVIG), while most HPeV3 strains could not be neutralized. In IVIG, very low neutralizing antibody (nAb) titres against HPeV3 were found. Additionally, very low nAb titres were observed in sera of two HPeV3-infected donors, while high nAb titres against HPeV1 could be detected. Our data suggest that the mild clinical course of HPeV1 infection is primarily influenced by strong nAb responses, while HPeV3 might be difficult to neutralize in vivo and therefore the course of infection will mainly be determined by in vivo cell tropism.


Subject(s)
Antibodies, Neutralizing/immunology , Parechovirus/classification , Parechovirus/physiology , RNA, Viral/metabolism , Viral Tropism/physiology , Animals , Antibodies, Viral/immunology , Cell Line , Gene Expression Regulation, Viral , Genotype , Humans , Parechovirus/pathogenicity , Picornaviridae Infections/virology , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Virus Replication
16.
Arch Virol ; 154(8): 1271-84, 2009.
Article in English | MEDLINE | ID: mdl-19585077

ABSTRACT

Ljungan virus is a picornavirus isolated from Swedish and North American rodents. Replication of Ljungan virus in cultured cells normally induces a weak and delayed cytopathic effect compared to that of many other picornaviruses. However, efficiently replicating Ljungan virus variants may evolve during serial passages in cell culture. In this study, we evaluate the significance of three substitutions in capsid protein VP0 and VP1 of a cell-culture-adapted variant of the Swedish Ljungan virus 145SL strain. In contrast to the parental strain, this 145SLG variant grows to high titers in green monkey kidney cells and induces a distinct cytopathic effect. Reverse genetic analyses demonstrated that each one of the individual capsid substitutions contributes to lytic replication in cell culture, but also that expression of all three substitutions results in a 100- to 500-fold increase in viral titers compared to viruses encoding single capsid substitutions. In addition, as indicated by detection of activated caspase-3 and DNA fragmentation, there seems to be an association between increased replication efficiency of lytic Ljungan virus variants and induction of an apoptotic response in infected green monkey kidney cells.


Subject(s)
Amino Acid Substitution , Capsid Proteins/physiology , Parechovirus/physiology , Virus Replication , 5' Untranslated Regions/genetics , Amino Acid Sequence , Animals , Apoptosis , Capsid Proteins/genetics , Cell Line , Cytopathogenic Effect, Viral , Molecular Sequence Data , Parechovirus/pathogenicity , Sweden , Virus Replication/genetics
18.
J Virol ; 82(3): 1581-90, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18032503

ABSTRACT

Calpains are calcium-dependent cysteine proteases that degrade cytoskeletal and cytoplasmic proteins. We have studied the role of calpains in the life cycle of human echovirus 1 (EV1). The calpain inhibitors, including calpeptin, calpain inhibitor 1, and calpain inhibitor 2 as well as calpain 1 and calpain 2 short interfering RNAs, completely blocked EV1 infection in the host cells. The effect of the inhibitors was not specific for EV1, because they also inhibited infection by other picornaviruses, namely, human parechovirus 1 and coxsackievirus B3. The importance of the calpains in EV1 infection also was supported by the fact that EV1 increased calpain activity 3 h postinfection. Confocal microscopy and immunoelectron microscopy showed that the EV1/caveolin-1-positive vesicles also contain calpain 1 and 2. Our results indicate that calpains are not required for virus entry but that they are important at a later stage of infection. Calpain inhibitors blocked the production of EV1 particles after microinjection of EV1 RNA into the cells, and they effectively inhibited the synthesis of viral RNA in the host cells. Thus, both calpain 1 and calpain 2 are essential for the replication of EV1 RNA.


Subject(s)
Calpain/metabolism , Enterovirus B, Human/physiology , RNA, Viral/biosynthesis , Virus Replication/physiology , Cell Line , Cytoplasmic Vesicles/chemistry , Cytoplasmic Vesicles/virology , Enzyme Inhibitors/pharmacology , Gene Silencing , Humans , Microscopy, Confocal , Microscopy, Immunoelectron , Parechovirus/physiology
19.
Microbiol Immunol ; 51(9): 841-50, 2007.
Article in English | MEDLINE | ID: mdl-17895601

ABSTRACT

It is of great importance to know how a virus particle is affected by environmental conditions. Physicochemical properties of the virion will affect the virus viability in different environments, viral transmission between hosts, and will also be important for safe handling of the virus. The physicochemical properties of the Ljungan virus (LV) prototype, 87-012, adapted to grow in cell culture were evaluated using both LV in crude cell extracts and purified virions. Replication of LV was completely inhibited by heat. Titers of LV were unaffected by acidic pH, reduced but not completely abolished by alkaline pH, and unaffected by exposure to the detergents Triton X-100 and SDS. Surprisingly, viable LV was still detected after incubation in the acidic, oxidising and detergent-containing environment produced by the commonly used disinfectant Virkon. In conclusion, LV is resilient to extreme pH, detergents and also to oxidising environments, but is sensitive to heat treatment.


Subject(s)
Detergents , Disinfectants , Hot Temperature , Parechovirus/physiology , Peroxides , Sulfuric Acids , Virion/physiology , Animals , Hydrogen-Ion Concentration , Octoxynol , Oxidation-Reduction , Parechovirus/isolation & purification , Sodium Dodecyl Sulfate , Virus Replication
20.
Gen Comp Endocrinol ; 154(1-3): 41-7, 2007.
Article in English | MEDLINE | ID: mdl-17686482

ABSTRACT

Bank voles (Clethrionomys glareolus) kept in captivity develop diabetes mellitus to a significant extent. Also in wild bank voles, elevated blood glucose has been observed. A newly isolated picornavirus named Ljungan virus (LV) has been found in the pancreas of these bank voles. Moreover, LV infection in combination with environmental factors may cause glucose intolerance/diabetes (GINT/D) in normal mice. The aim of the present study was to investigate the functional characteristics of pancreatic islets, isolated from bank voles, bred in the laboratory but considered LV infected. About 20% of all males and females were classified as GINT/D following a glucose tolerance test. Of these animals the majority had become diabetic by 20 weeks of age, with a tendency towards an earlier onset in the males. GINT/D animals had increased serum insulin levels. Islets were tested on the day of isolation (day 0) and after 1 week of culture for their insulin content and their capacity to synthesize (pro)insulin, secrete insulin and metabolize glucose. Functional differences could be observed between normal and GINT/D animals as well as between genders. An elevated basal insulin secretion was observed on day 0 indicating beta-cell dysfunction among islets isolated from diabetic males. In vitro culture could reverse some functional changes. The increased serum insulin level and the increased basal islet insulin secretion may suggest that the animals had developed a type 2 diabetes-like condition. It is likely that the putative stress imposed in the laboratory, maybe in combination with LV infection, can lead to an increased functional demand on the beta-cells.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/pathology , Glucose Intolerance/pathology , Insulin-Secreting Cells/physiology , Parechovirus/physiology , Animals , Arvicolinae , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 2/etiology , Disease Models, Animal , Female , Glucose/metabolism , Glucose/pharmacology , Glucose Intolerance/etiology , Insulin/analysis , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/chemistry , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Oxidation-Reduction , Picornaviridae Infections/complications , Picornaviridae Infections/veterinary , Protein Biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...