Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
PLoS One ; 16(4): e0249811, 2021.
Article in English | MEDLINE | ID: mdl-33861763

ABSTRACT

In Appalachia, La Crosse virus (LACV) is a leading pediatric arbovirus and public health concern for children under 16 years. LACV is transmitted via the bite of an infected Aedes mosquito. Thus, it is imperative to understand the dynamics of the local vector population in order to assess risk and transmission. Using entomological data collected from Knox County, Tennessee in 2013, we formulate an environmentally-driven system of ordinary differential equations to model mosquito population dynamics over a single season. Further, we include infected compartments to represent LACV transmission within the mosquito population. Findings suggest that the model, with dependence on degree days and accumulated precipitation, can closely describe field data. This model confirms the need to include these environmental variables when planning control strategies.


Subject(s)
Aedes/virology , Encephalitis, California/epidemiology , La Crosse virus/pathogenicity , Mosquito Vectors/pathogenicity , Population Dynamics/statistics & numerical data , Animals , Appalachian Region/epidemiology , Child , Child, Preschool , Encephalitis, California/transmission , Encephalitis, California/virology , Humans , La Crosse virus/isolation & purification , Models, Theoretical , Mosquito Vectors/virology , Seasons , Tennessee/epidemiology
2.
Emerg Infect Dis ; 23(12): 2075-2077, 2017 12.
Article in English | MEDLINE | ID: mdl-29148398

ABSTRACT

Infection with La Crosse virus can cause meningoencephalitis, but it is not known to cause acute flaccid paralysis (AFP). During 2008-2014, nine confirmed or probable La Crosse virus disease cases with possible AFP were reported in Ohio, USA. After an epidemiologic and clinical investigation, we determined no patients truly had AFP.


Subject(s)
Diagnostic Errors , Encephalitis, California/physiopathology , La Crosse virus/pathogenicity , Acute Disease , Adolescent , Aged , Animals , Child , Child, Preschool , Encephalitis, California/pathology , Encephalitis, California/virology , Female , Fever/physiopathology , Headache/physiopathology , Humans , La Crosse virus/physiology , Male , Medical Records , Muscle Weakness/physiopathology , Ohio , Paraplegia/diagnosis
3.
Acta Neuropathol ; 130(2): 233-45, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25956408

ABSTRACT

Viral neuroinvasion is a critical step in the pathogenesis of viral encephalitis. Multiple mechanisms of neuroinvasion have been identified, but their relative contribution to central nervous system (CNS) infection remains unclear for many viruses. In this study, we examined neuroinvasion of the mosquito-borne bunyavirus La Crosse (LACV), the leading cause of pediatric viral encephalitis in the USA. We found that the olfactory bulb (OB) and tract were the initial areas of CNS virus infection in mice. Removal of the OB reduced the incidence of LACV-induced disease demonstrating the importance of this area to neuroinvasion. However, we determined that infection of the OB was not due to axonal transport of virus from olfactory sensory neurons as ablation of these cells did not affect viral pathogenesis. Instead, we found that OB capillaries were compromised allowing leakage of virus-sized particles into the brain. Analysis of OB capillaries demonstrated specific alterations in cytoskeletal and Rho GTPase protein expression not observed in capillaries from other brain areas such as the cortex where leakage did not occur. Collectively, these findings indicate that LACV neuroinvasion occurs through hematogenous spread in specific brain regions where capillaries are prone to virus-induced activation such as the OB. Capillaries in these areas may be "hot spots" that are more susceptible to neuroinvasion not only for LACV, but other neurovirulent viruses as well.


Subject(s)
Capillaries/metabolism , Capillary Permeability/physiology , Cerebral Cortex/metabolism , Encephalitis, California/metabolism , La Crosse virus/pathogenicity , Olfactory Bulb/blood supply , Olfactory Bulb/virology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Capillaries/pathology , Capillaries/virology , Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Cerebral Cortex/virology , Cytoskeleton/metabolism , Disease Models, Animal , Encephalitis, California/pathology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Viral Load , Virus Internalization
4.
Pediatrics ; 134(3): e642-50, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25113294

ABSTRACT

OBJECTIVE: To describe the epidemiologic and clinical syndromes associated with pediatric neuroinvasive arboviral infections among children in the United States from 2003 through 2012. METHODS: We reviewed data reported by state health departments to ArboNET, the national arboviral surveillance system, for 2003 through 2012. Children (<18 years) with neuroinvasive arboviral infections (eg, meningitis, encephalitis, or acute flaccid paralysis) were included. Demographic, clinical syndrome, outcome, geographic, and temporal data were analyzed for all cases. RESULTS: During the study period, 1217 cases and 22 deaths due to pediatric neuroinvasive arboviral infection were reported from the 48 contiguous states. La Crosse virus (665 cases; 55%) and West Nile virus (505 cases; 41%) were the most common etiologies identified. Although less common, Eastern equine encephalitis virus (30 cases; 2%) resulted in 10 pediatric deaths. La Crosse virus primarily affected younger children, whereas West Nile virus was more common in older children and adolescents. West Nile virus disease cases occurred throughout the country, whereas La Crosse and the other arboviruses were more focally distributed. CONCLUSIONS: Neuroinvasive arboviral infections were an important cause of pediatric disease from 2003 through 2012. Differences in the epidemiology and clinical disease result from complex interactions among virus, vector, host, and the environment. Decreasing the morbidity and mortality from these agents depends on vector control, personal protection to reduce mosquito and tick bites, and blood donor screening. Effective surveillance is critical to inform clinicians and public health officials about the epidemiologic features of these diseases and to direct prevention efforts.


Subject(s)
Encephalitis, California/epidemiology , La Crosse virus , Population Surveillance/methods , West Nile Fever/epidemiology , Arbovirus Infections/diagnosis , Arbovirus Infections/epidemiology , Arbovirus Infections/virology , Child , Encephalitis, California/diagnosis , Encephalitis, California/virology , Humans , La Crosse virus/pathogenicity , United States/epidemiology , West Nile Fever/diagnosis , West Nile Fever/virology
5.
PLoS Pathog ; 8(7): e1002783, 2012.
Article in English | MEDLINE | ID: mdl-22792064

ABSTRACT

Ubiquitin (Ub) is a vital regulatory component in various cellular processes, including cellular responses to viral infection. As obligate intracellular pathogens, viruses have the capacity to manipulate the ubiquitin (Ub) cycle to their advantage by encoding Ub-modifying proteins including deubiquitinases (DUBs). However, how cellular DUBs modulate specific viral infections, such as norovirus, is poorly understood. To examine the role of DUBs during norovirus infection, we used WP1130, a small molecule inhibitor of a subset of cellular DUBs. Replication of murine norovirus in murine macrophages and the human norovirus Norwalk virus in a replicon system were significantly inhibited by WP1130. Chemical proteomics identified the cellular DUB USP14 as a target of WP1130 in murine macrophages, and pharmacologic inhibition or siRNA-mediated knockdown of USP14 inhibited murine norovirus infection. USP14 is a proteasome-associated DUB that also binds to inositol-requiring enzyme 1 (IRE1), a critical mediator of the unfolded protein response (UPR). WP1130 treatment of murine macrophages did not alter proteasome activity but activated the X-box binding protein-1 (XBP-1) through an IRE1-dependent mechanism. In addition, WP1130 treatment or induction of the UPR also reduced infection of other RNA viruses including encephalomyocarditis virus, Sindbis virus, and La Crosse virus but not vesicular stomatitis virus. Pharmacologic inhibition of the IRE1 endonuclease activity partially rescued the antiviral effect of WP1130. Taken together, our studies support a model whereby induction of the UPR through cellular DUB inhibition blocks specific viral infections, and suggest that cellular DUBs and the UPR represent novel targets for future development of broad spectrum antiviral therapies.


Subject(s)
Antiviral Agents/pharmacology , Nitriles/pharmacology , Norovirus/drug effects , Pyridines/pharmacology , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/metabolism , Unfolded Protein Response/drug effects , Animals , Caliciviridae Infections/drug therapy , Caliciviridae Infections/virology , Cell Line , Cell Line, Tumor , Cyanoacrylates , DNA-Binding Proteins/metabolism , Encephalomyocarditis virus/drug effects , Encephalomyocarditis virus/pathogenicity , Enzyme Inhibitors/pharmacology , Humans , La Crosse virus/drug effects , La Crosse virus/pathogenicity , Macrophages/virology , Membrane Proteins/metabolism , Mice , Norovirus/physiology , Norwalk virus/drug effects , Norwalk virus/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering , Regulatory Factor X Transcription Factors , Sindbis Virus/drug effects , Sindbis Virus/pathogenicity , Transcription Factors/metabolism , Ubiquitin/metabolism , Ubiquitin Thiolesterase/genetics , Virus Replication/drug effects , X-Box Binding Protein 1
6.
J Biol Chem ; 286(5): 3681-92, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21118815

ABSTRACT

La Crosse encephalitis virus (LACV) is a mosquito-borne member of the negative-strand RNA virus family Bunyaviridae. We have previously shown that the virulence factor NSs of LACV is an efficient inhibitor of the antiviral type I interferon system. A recombinant virus unable to express NSs (rLACVdelNSs) strongly induced interferon transcription, whereas the corresponding wt virus (rLACV) suppressed it. Here, we show that interferon induction by rLACVdelNSs mainly occurs through the signaling pathway leading from the pattern recognition receptor RIG-I to the transcription factor IRF-3. NSs expressed by rLACV, however, acts downstream of IRF-3 by specifically blocking RNA polymerase II-dependent transcription. Further investigations revealed that NSs induces proteasomal degradation of the mammalian RNA polymerase II subunit RPB1. NSs thereby selectively targets RPB1 molecules of elongating RNA polymerase II complexes, the so-called IIo form. This phenotype has similarities to the cellular DNA damage response, and NSs was indeed found to transactivate the DNA damage response gene pak6. Moreover, NSs expressed by rLACV boosted serine 139 phosphorylation of histone H2A.X, one of the earliest cellular reactions to damaged DNA. However, other DNA damage response markers such as up-regulation and serine 15 phosphorylation of p53 or serine 1524 phosphorylation of BRCA1 were not triggered by LACV infection. Collectively, our data indicate that the strong suppression of interferon induction by LACV NSs is based on a shutdown of RNA polymerase II transcription and that NSs achieves this by exploiting parts of the cellular DNA damage response pathway to degrade IIo-borne RPB1 subunits.


Subject(s)
La Crosse virus/pathogenicity , RNA Polymerase II/metabolism , Transcription, Genetic , Viral Nonstructural Proteins/physiology , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , DNA Damage , Enzyme Stability , Humans , Interferons/antagonists & inhibitors , RNA Polymerase II/antagonists & inhibitors , Transcriptional Activation , Vero Cells
7.
J Neuroimmune Pharmacol ; 5(3): 428-42, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20652430

ABSTRACT

Arthropod-borne viruses (arboviruses) are of paramount concern as a group of pathogens at the forefront of emerging and re-emerging diseases. Although some arboviral infections are asymptomatic or present with a mild influenza-like illness, many are important human and veterinary pathogens causing serious illness ranging from rash and arthritis to encephalitis and hemorrhagic fever. Here, we discuss arboviruses from diverse families (Flaviviruses, Alphaviruses, and the Bunyaviridae) that are causative agents of encephalitis in humans. An understanding of the natural history of these infections as well as shared mechanisms of neuroinvasion and neurovirulence is critical to control the spread of these viruses and for the development of effective vaccines and treatment modalities.


Subject(s)
Arboviruses/pathogenicity , Encephalitis, Arbovirus/virology , Alphavirus/pathogenicity , Alphavirus Infections/transmission , Alphavirus Infections/virology , Bunyaviridae/pathogenicity , Bunyaviridae Infections/transmission , Bunyaviridae Infections/virology , Chikungunya virus/pathogenicity , Communicable Diseases, Emerging/transmission , Communicable Diseases, Emerging/virology , Encephalitis, Arbovirus/transmission , Encephalitis, California/transmission , Encephalitis, California/virology , Flavivirus/pathogenicity , Flavivirus Infections/transmission , Flavivirus Infections/virology , Humans , La Crosse virus/pathogenicity , Rift Valley Fever/transmission , Rift Valley Fever/virology , Rift Valley fever virus/pathogenicity , West Nile Fever/transmission , West Nile Fever/virology , West Nile virus/pathogenicity
8.
PLoS One ; 4(9): e6954, 2009 Sep 09.
Article in English | MEDLINE | ID: mdl-19742311

ABSTRACT

BACKGROUND: Focal arboviral infections affecting a subset of the overall population present an often overlooked set of challenges in the assessment and reporting of risk and the detection of spatial patterns. Our objective was to assess the variation in risk when using different at-risk populations and geographic scales for the calculation of incidence risk and the detection of geographic hot-spots of infection. We explored these variations using a pediatric arbovirus, La Crosse virus (LACV), as our model. METHODS AND FINDINGS: Descriptive and cluster analyses were performed on probable and confirmed cases of LACV infections reported to the Tennessee Department of Health from 1997 to 2006, using three at-risk populations (the total population, the population 18 years and younger, and the population 15 years and younger) and at two geographic levels (county and census tract) to assess the variation in incidence risk and to investigate evidence of clustering using both global and local spatial statistics. We determined that the most appropriate at-risk population to calculate incidence risk and to assess the evidence of clustering was the population 15 years and younger. Based on our findings, the most appropriate geographical level to conduct spatial analyses and report incidence risk is the census tract level. The incidence risk in the population 15 years and younger at the county level ranged from 0 to 226.5 per 100,000 persons (median 41.5) in those counties reporting cases (n = 14) and at the census tract level it ranged from 50.9 to 673.9 per 100,000 persons (median 126.7) in those census tracts reporting cases (n = 51). To our knowledge, this is the highest reported incidence risk for this population at the county level for Tennessee and at the census tract level nationally. CONCLUSION: The results of this study indicate the possibility of missing disease clusters resulting from performing incidence risk investigations of focal diseases using inappropriate at-risk populations and/or at large geographic scales. Improved disease surveillance and health planning will result through the use of well defined at-risk populations and the use of appropriate geographic scales for the analysis and reporting of diseases. The finding of a high incidence risk of LACV infections in eastern Tennessee demonstrates that the vast majority of these infections continue to be under-diagnosed and/or underreported in this region. Persistent prevention and surveillance efforts will be required to reduce exposure to infectious vectors and to detect new cases of infection in this region. Application of this study's observations in future investigations will enhance the quantification of incidence risk and the identification of high-risk groups within the population.


Subject(s)
Arbovirus Infections/diagnosis , Arbovirus Infections/epidemiology , La Crosse virus/pathogenicity , Adolescent , Animals , Child , Cluster Analysis , Disease Vectors , Environmental Exposure , Humans , Incidence , La Crosse virus/metabolism , Population Surveillance , Risk , Risk Assessment , Tennessee , Treatment Outcome
9.
Virol J ; 5: 25, 2008 Feb 11.
Article in English | MEDLINE | ID: mdl-18267012

ABSTRACT

BACKGROUND: La Crosse virus (LACV), family Bunyaviridae, was first identified as a human pathogen in 1960 after its isolation from a 4 year-old girl with fatal encephalitis in La Crosse, Wisconsin. LACV is a major cause of pediatric encephalitis in North America and infects up to 300,000 persons each year of which 70-130 result in severe disease of the central nervous system (CNS). As an initial step in the establishment of useful animal models to support vaccine development, we examined LACV infectivity, pathogenesis, and immunogenicity in both weanling mice and rhesus monkeys. RESULTS: Following intraperitoneal inoculation of mice, LACV replicated in various organs before reaching the CNS where it replicates to high titer causing death from neurological disease. The peripheral site where LACV replicates to highest titer is the nasal turbinates, and, presumably, LACV can enter the CNS via the olfactory neurons from nasal olfactory epithelium. The mouse infectious dose50 and lethal dose50 was similar for LACV administered either intranasally or intraperitoneally. LACV was highly infectious for rhesus monkeys and infected 100% of the animals at 10 PFU. However, the infection was asymptomatic, and the monkeys developed a strong neutralizing antibody response. CONCLUSION: In mice, LACV likely gains access to the CNS via the blood stream or via olfactory neurons. The ability to efficiently infect mice intranasally raises the possibility that LACV might use this route to infect its natural hosts. Rhesus monkeys are susceptible to LACV infection and develop strong neutralizing antibody responses after inoculation with as little as 10 PFU. Mice and rhesus monkeys are useful animal models for LACV vaccine immunologic testing although the rhesus monkey model is not optimal.


Subject(s)
Disease Models, Animal , Encephalitis, California/virology , La Crosse virus/immunology , La Crosse virus/pathogenicity , Animals , Antibodies, Viral/immunology , Antigens, Viral/immunology , Cell Line , Central Nervous System/pathology , Central Nervous System/virology , Chlorocebus aethiops , Female , Humans , La Crosse virus/physiology , Lethal Dose 50 , Macaca mulatta , Mice , Neutralization Tests , Nose/virology , Peritoneum/virology , Vero Cells , Virus Replication
10.
Virol J ; 4: 41, 2007 May 08.
Article in English | MEDLINE | ID: mdl-17488515

ABSTRACT

BACKGROUND: La Crosse virus (LACV), family Bunyaviridae, is a mosquito-borne virus recognized as a major cause of pediatric encephalitis in North America with 70-130 symptomatic cases each year. The virus was first identified as a human pathogen in 1960 after its isolation from a 4 year-old girl who suffered encephalitis and died in La Crosse, Wisconsin. The majority of LACV infections are mild and never reported, however, serologic studies estimate infection rates of 10-30/100,000 in endemic areas. RESULTS: In the present study, sequence analysis of the complete LACV genomes of low-passage LACV/human/1960, LACV/mosquito/1978, and LACV/human/1978 strains and of biologically cloned derivatives of each strain, indicates that circulating LACVs are genetically stable over time and geographic distance with 99.6-100%, 98.9-100%, 97.8-99.6%, and 99.2-99.7% amino acid identity for N, NsS, M polyprotein, and L proteins respectively. We identified 5 amino acid differences in the RNA polymerase and 4 nucleotide differences in the non-coding region of the L segment specific to the human virus isolates, which may result in altered disease outcomes. CONCLUSION: All three wild type viruses had similar in vitro growth kinetics and phenotypes in mosquito C6/36 and Vero cells, and similar levels of neurovirulence and neuroinvasiveness in Swiss Webster mice. The biologically cloned derivative of LACV/human/1960 was significantly less neuroinvasive than its uncloned parent and differed in sequence at one amino acid position in the GN glycoprotein, identifying this residue as an attenuating mutation.


Subject(s)
Genome, Viral , La Crosse virus/genetics , RNA, Viral/genetics , Amino Acid Substitution , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , Culicidae , DNA, Complementary , Disease Models, Animal , Encephalitis, California/virology , Humans , La Crosse virus/growth & development , La Crosse virus/pathogenicity , Mice , Molecular Sequence Data , Phenotype , Point Mutation , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Vero Cells , Viral Proteins/genetics , Virulence/genetics
11.
J Virol ; 81(10): 4991-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17344298

ABSTRACT

La Crosse virus (LACV) is a mosquito-transmitted member of the Bunyaviridae family that causes severe encephalitis in children. For the LACV nonstructural protein NSs, previous overexpression studies with mammalian cells had suggested two different functions, namely induction of apoptosis and inhibition of RNA interference (RNAi). Here, we demonstrate that mosquito cells persistently infected with LACV do not undergo apoptosis and mount a specific RNAi response. Recombinant viruses that either express (rLACV) or lack (rLACVdelNSs) the NSs gene similarly persisted and were prone to the RNAi-mediated resistance to superinfection. Furthermore, in mosquito cells overexpressed LACV NSs was unable to inhibit RNAi against Semliki Forest virus. In mammalian cells, however, the rLACVdelNSs mutant virus strongly activated the antiviral type I interferon (IFN) system, whereas rLACV as well as overexpressed NSs suppressed IFN induction. Consequently, rLACVdelNSs was attenuated in IFN-competent mouse embryo fibroblasts and animals but not in systems lacking the type I IFN receptor. In situ analyses of mouse brains demonstrated that wild-type and mutant LACV mainly infect neuronal cells and that NSs is able to suppress IFN induction in the central nervous system. Thus, our data suggest little relevance of the NSs-induced apoptosis or RNAi inhibition for growth or pathogenesis of LACV in the mammalian host and indicate that NSs has no function in the insect vector. Since deletion of the viral NSs gene can be fully complemented by inactivation of the host's IFN system, we propose that the major biological function of NSs is suppression of the mammalian innate immune response.


Subject(s)
Interferon Type I/antagonists & inhibitors , La Crosse virus/immunology , RNA Interference , Viral Nonstructural Proteins/physiology , Animals , Apoptosis , Brain/pathology , Brain/virology , Cell Line , Cricetinae , Culicidae , Disease Models, Animal , Encephalitis, California/virology , Humans , Immunohistochemistry , La Crosse virus/pathogenicity , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Neurons/virology , Semliki forest virus/growth & development
12.
J Virol ; 79(16): 10420-8, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16051834

ABSTRACT

La Crosse virus (LACV) belongs to the Bunyaviridae family and causes severe encephalitis in children. It has a negative-sense RNA genome which consists of the three segments L, M, and S. We successfully rescued LACV by transfection of just three plasmids, using a system which was previously established for Bunyamwera virus (Lowen et al., Virology 330:493-500, 2004). These cDNA plasmids represent the three viral RNA segments in the antigenomic orientation, transcribed intracellularly by the T7 RNA polymerase and with the 3' ends trimmed by the hepatitis delta virus ribozyme. As has been shown for Bunyamwera virus, the antigenomic plasmids could serve both as donors for the antigenomic RNA and as support plasmids to provide small amounts of viral proteins for RNA encapsidation and particle formation. In contrast to other rescue systems, however, transfection of additional support plasmids completely abrogated the rescue, indicating that LACV is highly sensitive to overexpression of viral proteins. The BSR-T7/5 cell line, which constitutively expresses T7 RNA polymerase, allowed efficient rescue of LACV, generating approximately 10(8) infectious viruses per milliliter. The utility of this system was demonstrated by the generation of a wild-type virus containing a genetic marker (rLACV) and of a mutant with a deleted NSs gene on the S segment (rLACVdelNSs). The NSs-expressing rLACV formed clear plaques, displayed an efficient host cell shutoff, and was strongly proapoptotic. The rLACVdelNSs mutant, by contrast, exhibited a turbid-plaque phenotype and a less-pronounced shutoff and induced little apoptosis. Nevertheless, both viruses grew in Vero cells to similar titers. Our reverse genetics system now enables us to manipulate the genome of LACV in order to characterize its virulence factors and to develop potential vaccine candidates.


Subject(s)
La Crosse virus/genetics , Viral Nonstructural Proteins/physiology , Animals , Base Sequence , Cells, Cultured , Cloning, Molecular , Genome, Viral , Humans , La Crosse virus/pathogenicity , La Crosse virus/physiology , Molecular Sequence Data , Plasmids , Recombination, Genetic , Viral Nonstructural Proteins/genetics
13.
Virology ; 338(1): 121-32, 2005 Jul 20.
Article in English | MEDLINE | ID: mdl-15923017

ABSTRACT

Members of the California serogroup of orthobunyaviruses, particularly La Crosse (LAC) and Tahyna (TAH) viruses, are significant human pathogens in areas where their mosquito vectors are endemic. Previous studies using wild-type LAC and TAH181/57, a highly neurovirulent strain with low neuroinvasiveness (Janssen, R., Gonzalez-Scarano, F., Nathanson, N., 1984. Mechanisms of bunyavirus virulence. Comparative pathogenesis of a virulent strain of La Crosse and an avirulent strain of Tahyna virus. Lab. Invest. 50 (4), 447-455), have demonstrated that the neuroinvasive phenotype maps to the M segment, the segment that encodes the two viral glycoproteins Gn (G2) and Gc (G1), as well as a non-structural protein NSm. To further define the role of Gn and Gc in fusion and entry, we prepared a panel of recombinant M segment constructs using LAC, TAH181/57, and V22F, a monoclonal-resistant variant of LAC with deficient fusion function. These M segment constructs were then tested in two surrogate assays for virus entry: a cell-to-cell fusion assay based on T7-luciferase expression, and a pseudotype transduction assay based on the incorporation of the bunyavirus glycoproteins on an MLV backbone. Both assays demonstrated that Gc is the principal determinant of virus fusion and cell entry, and furthermore that the region delineated by amino acids 860-1442, corresponding to the membrane proximal two-thirds of Gc, is key to these processes. These results, coupled with structural modeling suggesting homologies between the carboxy region of Gc and Sindbis virus E1, suggest that the LAC Gc functions as a type II fusion protein.


Subject(s)
Encephalitis Virus, California/pathogenicity , La Crosse virus/pathogenicity , Viral Proteins/physiology , Animals , Cell Fusion , Cell Line , Cricetinae , Encephalitis Virus, California/classification , Encephalitis Virus, California/genetics , Gene Expression , Genes, Viral , Humans , Hydrogen-Ion Concentration , La Crosse virus/classification , La Crosse virus/genetics , Leukemia Virus, Murine/genetics , Models, Molecular , Protein Conformation , Quail , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transduction, Genetic , Transfection , Viral Proteins/chemistry , Viral Proteins/genetics , Virulence
14.
J Virol ; 79(1): 234-44, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15596819

ABSTRACT

Through a process known as RNA interference (RNAi), double-stranded short interfering RNAs (siRNAs) silence gene expression in a sequence-specific manner. Recently, several viral proteins, including the nonstructural protein NSs of tomato spotted wilt virus (a plant-infecting bunyavirus), the interferon antagonist protein NS1 of influenza virus, and the E3L protein of vaccinia virus, have been shown to function as suppressors of RNAi, presumably as a counterdefense against cellular mechanisms that decrease viral production. La Crosse virus (LACV), a member of the California serogroup of orthobunyaviruses, has a trisegmented negative-stranded genome comprised of large (L), medium (M), and small (S) segments. To develop a strategy for segment-specific inhibition of transcription, we designed 13 synthetic siRNAs targeting specific RNA segments of the LACV genome that decreased LACV replication and antigen expression in mammalian (293T) and insect (C6/36) cells. Furthermore, NSs, a LACV nonstructural protein, markedly inhibited RNAi directed both against an LACV M segment construct and against a host gene (glyeraldehyde-3-phosphate dehydrogenase), suggesting a possible role for this viral protein in the suppression of RNA silencing. Segment-specific siRNAs will be useful as a tool to analyze LACV transcription and replication and to obtain recombinant viruses. Additionally, NSs will help us to identify molecular pathways involved in RNAi and further define its role in the innate immune system.


Subject(s)
Gene Expression Regulation, Viral , La Crosse virus/pathogenicity , MicroRNAs/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , La Crosse virus/genetics , La Crosse virus/metabolism , MicroRNAs/genetics , Molecular Sequence Data , Mutation , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Transfection , Vero Cells , Viral Nonstructural Proteins/genetics , Viral Plaque Assay , Virus Replication
15.
Am J Infect Control ; 31(8): 508-10, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14647115

ABSTRACT

BACKGROUND: La Crosse encephalitis (LAC) is a mosquito-borne illness that primarily affects children. In 1997, an initial cluster of cases was identified by the regional pediatric referral center in East Tennessee. Since that time, public health officials, pediatric infectious disease physicians, infection control nurses, laboratory personnel, entomologists, and Centers for Disease Control and Prevention consultants have collaborated to provide ongoing surveillance activities. Studies comparing LAC cases with non-LAC (no etiologic diagnosis) central nervous system infections have yielded no statistical significance in signs and symptoms or laboratory values. OBJECTIVE: To determine any differences in signs, symptoms, and/or diagnostic laboratory values between LAC cases and enteroviral central nervous system (EV-CNS) infections. METHODS: In 2001, descriptive public health surveillance for LAC was performed concurrent with an outbreak investigation of EV-CNS infections at a pediatric referral center in East Tennessee. All patients being evaluated for suspected meningitis and/or encephalitis were interviewed for signs and symptoms of illness. Patients with positive test results for LAC and negative results for EV or positive results for EV and negative results for LAC were included in the study. RESULTS: Compared with patients with EV-CNS infection, patients with LAC were significantly more likely to have aphasia (P=.001), loss of consciousness (P=.0003), seizure (P=.0003), and admission to the pediatric intensive care unit (P=.02). Presence of fever, headache, vomiting, stiff neck (subjective), photophobia, behavioral changes, confusion, need for mechanical ventilation, age, and sex showed no statistical significance (P>.05). Statistical differences were not demonstrated in cerebrospinal fluid laboratory values (P>.05). CONCLUSION: Patients with LAC demonstrated more severe symptoms on presentation to the hospital than did patients with EV-CNS infection. A possible advantage of identifying specific viral etiologic factors of pediatric CNS disease by clinical characteristics may be the ability to take advantage of emerging antiviral therapies.


Subject(s)
Central Nervous System Viral Diseases/epidemiology , Encephalitis, California/epidemiology , Enterovirus Infections/epidemiology , La Crosse virus , Adolescent , Aedes/virology , Animals , Central Nervous System Viral Diseases/complications , Central Nervous System Viral Diseases/diagnosis , Child , Child, Preschool , Encephalitis, California/complications , Encephalitis, California/diagnosis , Enterovirus/isolation & purification , Enterovirus/pathogenicity , Enterovirus Infections/complications , Enterovirus Infections/diagnosis , Female , Humans , Infant , Insect Vectors/virology , La Crosse virus/isolation & purification , La Crosse virus/pathogenicity , Male , Population Surveillance , Severity of Illness Index , Tennessee/epidemiology
17.
Virus Res ; 48(2): 143-8, 1997 May.
Article in English | MEDLINE | ID: mdl-9175252

ABSTRACT

La Crosse (LAC) virus belongs to the California (CAL) serogroup of the genus Bunyavirus, family Bunyaviridae. It is considered one of the most important mosquito-borne pathogens in North America, especially in the upper Mid-West, where it is associated with encephalitis during the time of year when mosquitoes are active. Infections occur most frequently in children and young adults and, while most cases are resolved after a period of intense illness, a small fraction (< 1%) are fatal. At present there have only been three isolates of LAC virus from humans all made from brain tissue postmortem. The cases yielding viruses are separated chronologically by 33 years and geographically from Minnesota/Wisconsin (1960, 1978) to Missouri (1993). The M RNA sequence of the first two isolates was previously reported. The present study extends the observations to the isolate from the 1993 case and includes several mosquito isolates as well. A comparison of the M RNAs of these viruses shows that for the human isolates both nucleotide sequence and the deduced amino-acid sequence of the encoded proteins are highly conserved, showing a maximum variation of only 0.91% and 0.69%, respectively. This high degree of conservation over time and space leads to the hypothesis that human infections with this particular genotype of LAC virus are those most likely to have a fatal outcome. It is also shown that a virus with this genotype could be found circulating in mosquitoes in an area more or less intermediate between the locations of the first and second fatal cases.


Subject(s)
Encephalitis, California/genetics , La Crosse virus/genetics , Aedes/virology , Amino Acid Sequence , Animals , Encephalitis, California/mortality , Encephalitis, California/virology , Genotype , Geography , La Crosse virus/chemistry , La Crosse virus/pathogenicity , Molecular Sequence Data , RNA, Viral/genetics , Sequence Analysis, DNA , Virulence/genetics
18.
Am J Trop Med Hyg ; 56(2): 171-6, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9080876

ABSTRACT

The teratogenic potential of three bunyaviruses, two California serogroup bunyaviruses, LaCrosse virus and San Angelo virus, and a Bunyamwera serogroup member, Main Drain virus, in sheep was studied following in utero inoculation of ewes in early gestation. Although Main Drain virus appeared to be most teratogenic, all three viruses induced a range of lesions including arthrogryposis, hydrocephalus, fetal death, axial skeletal deviations, anasarca, and oligohydramnios. The teratogenic effects of these viruses are identical to those described in ovine infections by Cache Valley and Akabane viruses. Demonstration of a common bunyaviral tropism for fetal tissue infection that results in congenital brain and musculoskeletal malformations provides evidence that human in utero infection by bunyaviruses could result in similar malformations in human infants.


Subject(s)
Abnormalities, Multiple/veterinary , Bunyamwera virus/pathogenicity , Bunyaviridae Infections/veterinary , Encephalitis Virus, California/pathogenicity , Fetus/abnormalities , Pregnancy Complications, Infectious/veterinary , Sheep Diseases/virology , Abnormalities, Multiple/embryology , Abnormalities, Multiple/virology , Animals , Arthrogryposis/embryology , Arthrogryposis/veterinary , Arthrogryposis/virology , Bunyamwera virus/isolation & purification , Bunyaviridae Infections/complications , Bunyaviridae Infections/embryology , Chlorocebus aethiops , Encephalitis Virus, California/isolation & purification , Encephalitis, California/complications , Encephalitis, California/embryology , Encephalitis, California/veterinary , Female , Fetal Death/veterinary , Fetal Death/virology , Hydrocephalus/embryology , Hydrocephalus/veterinary , Hydrocephalus/virology , La Crosse virus/isolation & purification , La Crosse virus/pathogenicity , Oligohydramnios/veterinary , Oligohydramnios/virology , Pregnancy , Sheep , Sheep Diseases/embryology , Vero Cells
19.
Am J Trop Med Hyg ; 55(5): 567-9, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8940992

ABSTRACT

We examined the usefulness of mongolian gerbils (Meriones unguiculatus) as a new animal model for La Crosse virus (LACV) studies. Gerbils were exposed to LACV by either intramuscular (im) inoculation or exposure to transovarially infected Aedes triseriatus. Our studies indicate that gerbils may be a suitable animal model for LACV infection. Gerbils were susceptible to LACV, survived viral infection, and developed viremias and neutralizing antibody titers following exposure by im injection and by the bite of infected mosquitoes. Moreover, they are attractive to mosquito vectors. Gerbils have other advantages as laboratory vertebrate hosts for LACV; they are inexpensive, breed in captivity, and are usually mild-mannered and easy to handle. Thus, gerbils are a suitable model in the study of LACV pathogenesis as well as of transplacental and vector transmission.


Subject(s)
Encephalitis, California/virology , Gerbillinae/virology , La Crosse virus/pathogenicity , Viremia/virology , Aedes/virology , Animals , Disease Models, Animal , Disease Transmission, Infectious , Encephalitis, California/transmission , Infectious Disease Transmission, Vertical , Insect Vectors/virology , Neutralization Tests , Viremia/transmission
20.
J Virol ; 69(6): 3475-81, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7745694

ABSTRACT

La Crosse virus, a member of the California serogroup of bunyaviruses, is an important cause of pediatric encephalitis in the midwestern United States. Like all bunyaviruses, La Crosse virus contains two glycoproteins, G1 and G2, the larger of which, G1, is the target of neutralizing antibodies. To develop an understanding of the role of each of the glycoproteins in the generation of a protective immune response, we immunized 1-week-old mice with three different preparations: a vaccinia virus recombinant (VV.ORF) that expresses both G1 and G2, a vaccinia virus recombinant (VV.G1) that expresses G1 only, and a truncated soluble G1 (sG1) protein prepared in a baculovirus system. Whereas VV.ORF generated a protective response that was mostly directed against G1, VV.G1 was only partially effective at inducing a neutralizing response and at protecting mice from a potentially lethal challenge with La Crosse virus. Nevertheless, a single immunization with the sG1 preparation resulted in a robust immune response and protection against La Crosse virus. These results indicate that (i) the G1 protein by itself can induce an immune response sufficient for protection from a lethal challenge with La Crosse virus, (ii) a neutralizing humoral response correlates with protection, and (iii) the context in which G1 is presented affects its immunogenicity. The key step in the defense against central nervous system infection appeared to be interruption of a transient viremia that occurred just after La Crosse virus inoculation.


Subject(s)
Antibodies, Viral/immunology , Encephalitis, California/prevention & control , Glycoproteins/immunology , La Crosse virus/immunology , Viral Proteins/immunology , Aging/immunology , Animals , Cattle , Cell Line , Disease Susceptibility/immunology , Encephalitis, California/immunology , Glycoproteins/genetics , Glycoproteins/therapeutic use , Immunotherapy, Adoptive , La Crosse virus/metabolism , La Crosse virus/pathogenicity , Mice , Neutralization Tests , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Viral Proteins/genetics , Viral Proteins/therapeutic use , Viral Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...