Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
PLoS Genet ; 18(12): e1010548, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36574452

RESUMEN

Variation in immune homeostasis, the state in which the immune system is maintained in the absence of stimulation, is highly variable across populations. This variation is attributed to both genetic and environmental factors. However, the identity and function of specific regulators have been difficult to identify in humans. We evaluated homeostatic antibody levels in the serum of the Collaborative Cross (CC) mouse genetic reference population. We found heritable variation in all antibody isotypes and subtypes measured. We identified 4 quantitative trait loci (QTL) associated with 3 IgG subtypes: IgG1, IgG2b, and IgG2c. While 3 of these QTL map to genome regions of known immunological significance (major histocompatibility and immunoglobulin heavy chain locus), Qih1 (associated with variation in IgG1) mapped to a novel locus on Chromosome 18. We further associated this locus with B cell proportions in the spleen and identify Methyl-CpG binding domain protein 1 under this locus as a novel regulator of homeostatic IgG1 levels in the serum and marginal zone B cells (MZB) in the spleen, consistent with a role in MZB differentiation to antibody secreting cells.


Asunto(s)
Ratones de Colaboración Cruzada , Sitios de Carácter Cuantitativo , Ratones , Humanos , Animales , Sitios de Carácter Cuantitativo/genética , Ratones de Colaboración Cruzada/genética , Activación de Linfocitos , Inmunoglobulina G/genética , Homeostasis/genética , Proteínas de Unión al ADN/genética , Factores de Transcripción/genética
2.
PLoS Genet ; 11(10): e1005504, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26452100

RESUMEN

New systems genetics approaches are needed to rapidly identify host genes and genetic networks that regulate complex disease outcomes. Using genetically diverse animals from incipient lines of the Collaborative Cross mouse panel, we demonstrate a greatly expanded range of phenotypes relative to classical mouse models of SARS-CoV infection including lung pathology, weight loss and viral titer. Genetic mapping revealed several loci contributing to differential disease responses, including an 8.5Mb locus associated with vascular cuffing on chromosome 3 that contained 23 genes and 13 noncoding RNAs. Integrating phenotypic and genetic data narrowed this region to a single gene, Trim55, an E3 ubiquitin ligase with a role in muscle fiber maintenance. Lung pathology and transcriptomic data from mice genetically deficient in Trim55 were used to validate its role in SARS-CoV-induced vascular cuffing and inflammation. These data establish the Collaborative Cross platform as a powerful genetic resource for uncovering genetic contributions of complex traits in microbial disease severity, inflammation and virus replication in models of outbred populations.


Asunto(s)
Interacciones Huésped-Patógeno , Inflamación/genética , Síndrome Respiratorio Agudo Grave/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Humanos , Inflamación/patología , Inflamación/virología , Ratones , Fenotipo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Síndrome Respiratorio Agudo Grave/patología , Síndrome Respiratorio Agudo Grave/virología , Replicación Viral/genética
3.
J Virol ; 90(1): 433-43, 2016 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26491151

RESUMEN

UNLABELLED: Chikungunya virus (CHIKV) is an alphavirus responsible for causing epidemic outbreaks of polyarthralgia in humans. Because CHIKV is initially introduced via the skin, where γδ T cells are prevalent, we evaluated the response of these cells to CHIKV infection. CHIKV infection led to a significant increase in γδ T cells in the infected foot and draining lymph node that was associated with the production of proinflammatory cytokines and chemokines in C57BL/6J mice. γδ T cell(-/-) mice demonstrated exacerbated CHIKV disease characterized by less weight gain and greater foot swelling than occurred in wild-type mice, as well as a transient increase in monocytes and altered cytokine/chemokine expression in the foot. Histologically, γδ T cell(-/-) mice had increased inflammation-mediated oxidative damage in the ipsilateral foot and ankle joint compared to wild-type mice which was independent of differences in CHIKV replication. These results suggest that γδ T cells play a protective role in limiting the CHIKV-induced inflammatory response and subsequent tissue and joint damage. IMPORTANCE: Recent epidemics, including the 2004 to 2007 outbreak and the spread of CHIKV to naive populations in the Caribbean and Central and South America with resultant cases imported into the United States, have highlighted the capacity of CHIKV to cause explosive epidemics where the virus can spread to millions of people and rapidly move into new areas. These studies identified γδ T cells as important to both recruitment of key inflammatory cell populations and dampening the tissue injury due to oxidative stress. Given the importance of these cells in the early response to CHIKV, this information may inform the development of CHIKV vaccines and therapeutics.


Asunto(s)
Fiebre Chikungunya/inmunología , Virus Chikungunya/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/análisis , Linfocitos T/inmunología , Animales , Peso Corporal , Modelos Animales de Enfermedad , Miembro Posterior/patología , Histocitoquímica , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Linfocitos T/química
4.
PLoS Pathog ; 9(2): e1003196, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23468633

RESUMEN

Genetic variation contributes to host responses and outcomes following infection by influenza A virus or other viral infections. Yet narrow windows of disease symptoms and confounding environmental factors have made it difficult to identify polymorphic genes that contribute to differential disease outcomes in human populations. Therefore, to control for these confounding environmental variables in a system that models the levels of genetic diversity found in outbred populations such as humans, we used incipient lines of the highly genetically diverse Collaborative Cross (CC) recombinant inbred (RI) panel (the pre-CC population) to study how genetic variation impacts influenza associated disease across a genetically diverse population. A wide range of variation in influenza disease related phenotypes including virus replication, virus-induced inflammation, and weight loss was observed. Many of the disease associated phenotypes were correlated, with viral replication and virus-induced inflammation being predictors of virus-induced weight loss. Despite these correlations, pre-CC mice with unique and novel disease phenotype combinations were observed. We also identified sets of transcripts (modules) that were correlated with aspects of disease. In order to identify how host genetic polymorphisms contribute to the observed variation in disease, we conducted quantitative trait loci (QTL) mapping. We identified several QTL contributing to specific aspects of the host response including virus-induced weight loss, titer, pulmonary edema, neutrophil recruitment to the airways, and transcriptional expression. Existing whole-genome sequence data was applied to identify high priority candidate genes within QTL regions. A key host response QTL was located at the site of the known anti-influenza Mx1 gene. We sequenced the coding regions of Mx1 in the eight CC founder strains, and identified a novel Mx1 allele that showed reduced ability to inhibit viral replication, while maintaining protection from weight loss.


Asunto(s)
Variación Genética , Interacciones Huésped-Patógeno/genética , Gripe Humana/virología , Modelos Genéticos , Infecciones por Orthomyxoviridae/virología , Enfermedades de los Roedores/virología , Animales , Cruzamientos Genéticos , Femenino , Humanos , Virus de la Influenza A , Gripe Humana/genética , Gripe Humana/patología , Pulmón/patología , Ratones , Ratones Endogámicos , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Fenotipo , Virus Reordenados/genética , Virus Reordenados/patogenicidad , Recombinación Genética , Enfermedades de los Roedores/genética , Enfermedades de los Roedores/patología , Especificidad de la Especie , Replicación Viral
5.
J Virol ; 87(10): 5697-706, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23487448

RESUMEN

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus responsible for recent epidemic outbreaks of debilitating disease in humans. Alphaviruses are known to interact with members of the C-type lectin receptor family of pattern recognition proteins, and given that the dendritic cell immunoreceptor (DCIR) is known to act as a negative regulator of the host inflammatory response and has previously been associated with rheumatoid arthritis, we evaluated DCIR's role in response to CHIKV infection. Although we observed an increase in the proportion of dendritic cells at the site of CHIKV infection at 24 to 36 h postinfection, these cells showed decreased cell surface DCIR, suggestive of DCIR triggering and internalization. In vitro, bone marrow-derived dendritic cells from DCIR-deficient (DCIR(-/-)) mice exhibited altered cytokine expression following exposure to CHIKV. DCIR(-/-) mice exhibited more severe disease signs than wild-type C57BL6/J mice following CHIKV infection, including a more rapid and more severe onset of virus-induced edema and enhanced weight loss. Histological examination revealed that DCIR-deficient animals exhibited increased inflammation and damage in both the fascia of the inoculated foot and the ankle joint, and DCIR deficiency skewed the CHIKV-induced cytokine response at the site of infection at multiple times postinfection. Early differences in virus-induced disease between C57BL6/J and DCIR(-/-) mice were independent of viral replication, while extended viral replication correlated with enhanced foot swelling and tissue inflammation and damage in DCIR(-/-) compared to C57BL6/J mice at 6 to 7 days postinfection. These results suggest that DCIR plays a protective role in limiting the CHIKV-induced inflammatory response and subsequent tissue and joint damage.


Asunto(s)
Virus Chikungunya/inmunología , Virus Chikungunya/patogenicidad , Células Dendríticas/inmunología , Células Dendríticas/virología , Lectinas Tipo C/metabolismo , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/patología , Animales , Articulación del Tobillo/patología , Fiebre Chikungunya , Modelos Animales de Enfermedad , Pie/patología , Histocitoquímica , Lectinas Tipo C/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
6.
PLoS Pathog ; 8(3): e1002586, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22457620

RESUMEN

Mosquito-borne alphaviruses such as chikungunya virus and Ross River virus (RRV) are emerging pathogens capable of causing large-scale epidemics of virus-induced arthritis and myositis. The pathology of RRV-induced disease in both humans and mice is associated with induction of the host inflammatory response within the muscle and joints, and prior studies have demonstrated that the host complement system contributes to development of disease. In this study, we have used a mouse model of RRV-induced disease to identify and characterize which complement activation pathways mediate disease progression after infection, and we have identified the mannose binding lectin (MBL) pathway, but not the classical or alternative complement activation pathways, as essential for development of RRV-induced disease. MBL deposition was enhanced in RRV infected muscle tissue from wild type mice and RRV infected MBL deficient mice exhibited reduced disease, tissue damage, and complement deposition compared to wild-type mice. In contrast, mice deficient for key components of the classical or alternative complement activation pathways still developed severe RRV-induced disease. Further characterization of MBL deficient mice demonstrated that similar to C3(-/-) mice, viral replication and inflammatory cell recruitment were equivalent to wild type animals, suggesting that RRV-mediated induction of complement dependent immune pathology is largely MBL dependent. Consistent with these findings, human patients diagnosed with RRV disease had elevated serum MBL levels compared to healthy controls, and MBL levels in the serum and synovial fluid correlated with severity of disease. These findings demonstrate a role for MBL in promoting RRV-induced disease in both mice and humans and suggest that the MBL pathway of complement activation may be an effective target for therapeutic intervention for humans suffering from RRV-induced arthritis and myositis.


Asunto(s)
Infecciones por Alphavirus/complicaciones , Artritis Reactiva/virología , Lectina de Unión a Manosa/metabolismo , Miositis/virología , Virus del Río Ross/fisiología , Infecciones por Alphavirus/metabolismo , Infecciones por Alphavirus/patología , Animales , Artritis Reactiva/metabolismo , Artritis Reactiva/patología , Activación de Complemento , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/virología , Miositis/metabolismo , Miositis/patología , Virus del Río Ross/patogenicidad , Líquido Sinovial/metabolismo , Replicación Viral
7.
J Virol ; 86(19): 10675-85, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22837203

RESUMEN

Arthralgia-associated alphaviruses, including chikungunya virus (CHIKV) and Ross River virus (RRV), pose significant public health threats because of their ability to cause explosive outbreaks of debilitating arthralgia and myalgia in human populations. Although the host inflammatory response is known to contribute to the pathogenesis of alphavirus-induced arthritis and myositis, the role that Toll-like receptors (TLRs), which are major regulators of host antiviral and inflammatory responses, play in the pathogenesis of alphavirus-induced arthritis and myositis has not been extensively studied. Using a mouse model of RRV-induced myositis/arthritis, we found that myeloid differentiation primary response gene 88 (Myd88)-dependent TLR7 signaling is involved in protection from severe RRV-associated disease. Infections of Myd88- and TLR7-deficient mouse strains with RRV revealed that both Myd88 and TLR7 significantly contributed to protection from RRV-induced mortality, and both mouse strains exhibited more severe tissue damage than wild-type (WT) mice following RRV infection. While viral loads were unchanged in either Myd88 or TLR7 knockout mice compared to WT mice at early times postinfection, both Myd88 and TLR7 knockout mice exhibited higher viral loads than WT mice at late times postinfection. Furthermore, while high levels of RRV-specific antibody were produced in TLR7-deficient mice, this antibody had very little neutralizing activity and had lower affinity than WT antibody. Additionally, TLR7- and Myd88-deficient mice showed defects in germinal center activity, suggesting that TLR7-dependent signaling is critical for the development of protective antibody responses against RRV.


Asunto(s)
Infecciones por Alphavirus/metabolismo , Glicoproteínas de Membrana/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Virus del Río Ross/metabolismo , Receptor Toll-Like 7/metabolismo , Animales , Anticuerpos Neutralizantes/química , Bioensayo/métodos , Ensayo de Inmunoadsorción Enzimática/métodos , Citometría de Flujo/métodos , Inflamación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Vero
8.
J Virol ; 85(20): 10682-90, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21849461

RESUMEN

Venezuelan equine encephalitis (VEE) virus is a mosquito-borne alphavirus associated with sporadic outbreaks in human and equid populations in the Western Hemisphere. After the bite of an infected mosquito, the virus initiates a biphasic disease: a peripheral phase with viral replication in lymphoid and myeloid tissues, followed by a neurotropic phase with infection of central nervous system (CNS) neurons, causing neuropathology and in some cases fatal encephalitis. The mechanisms allowing VEE virus to enter the CNS are currently poorly understood. Previous data have shown that the virus gains access to the CNS by infecting olfactory sensory neurons in the nasal mucosa of mice. However, at day 5 after inoculation, the infection of the brain is multifocal, indicating that virus particles are able to cross the blood-brain barrier (BBB). To better understand the role of the BBB during VEE virus infection, we used a well-characterized mouse model system. Using VEE virus replicon particles (VRP), we modeled the early events of neuroinvasion, showing that the replication of VRP in the nasal mucosa induced the opening of the BBB, allowing peripherally administered VRP to invade the brain. Peripheral VEE virus infection was characterized by a biphasic opening of the BBB. Further, inhibition of BBB opening resulted in a delayed viral neuroinvasion and pathogenesis. Overall, these results suggest that VEE virus initially enters the CNS through the olfactory pathways and initiates viral replication in the brain, which induces the opening of the BBB, allowing a second wave of invading virus from the periphery to enter the brain.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Barrera Hematoencefálica/virología , Virus de la Encefalitis Equina Venezolana/patogenicidad , Encefalomielitis Equina Venezolana/patología , Encefalomielitis Equina Venezolana/virología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos BALB C , Neuronas Receptoras Olfatorias/virología , Enfermedades de los Roedores/patología , Enfermedades de los Roedores/virología
9.
Am J Pathol ; 178(1): 32-40, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21224040

RESUMEN

Chikungunya virus (CHIKV), an emerging mosquito-borne Alphavirus, causes debilitating rheumatic disease in humans that can last for weeks to months. Starting in 2004, a CHIKV outbreak in the Indian Ocean region affected millions of people, and infected travelers introduced CHIKV to new regions. The pathogenesis of CHIKV is poorly understood, and no approved vaccines or specific therapies exist. A major challenge to the study of CHIKV disease is the lack of a small animal model that recapitulates the major outcomes of human infection. In this study, the pathogenesis of CHIKV in C57BL/6J mice was investigated using biological and molecular clones of CHIKV isolated from human serum (CHIKV SL15649). After 14-day-old mice were inoculated with CHIKV SL15649 in the footpad, they displayed reduced weight gain and swelling of the inoculated limb. Histologic analysis of hind limb sections revealed severe necrotizing myositis, mixed inflammatory cell arthritis, chronic active tenosynovitis, and multifocal vasculitis. Interestingly, these disease signs and viral RNA persisted in musculoskeletal tissues for at least 3 weeks after inoculation. This work demonstrates the development of a mouse model of CHIKV infection with clinical manifestations and histopathologic findings that are consistent with the disease signs of CHIKV-infected humans, providing a useful tool for studying viral and host factors that drive CHIKV pathogenesis and for evaluating potential therapeutics against this emerging viral disease.


Asunto(s)
Artritis Reumatoide/virología , Virus Chikungunya , Modelos Animales de Enfermedad , Ratones , Miositis/virología , Tenosinovitis/virología , Infecciones por Alphavirus/patología , Animales , Artritis Reumatoide/patología , Fiebre Chikungunya , Miembro Posterior/patología , Humanos , Ratones Endogámicos C57BL , Músculo Esquelético/patología , Miositis/patología , Tenosinovitis/patología
10.
J Virol ; 84(9): 4556-68, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20181704

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is a mosquito-borne RNA virus of the genus Alphavirus that is responsible for a significant disease burden in Central and South America through sporadic outbreaks into human and equid populations. For humans, 2 to 4% of cases are associated with encephalitis, and there is an overall case mortality rate of approximately 1%. In mice, replication of the virus within neurons of the central nervous system (CNS) leads to paralyzing, invariably lethal encephalomyelitis. However, mice infected with certain attenuated mutants of the virus are able to control the infection within the CNS and recover. To better define what role T cell responses might be playing in this process, we infected B cell-deficient microMT mice with a VEEV mutant that induces mild, sublethal illness in immune competent mice. Infected microMT mice rapidly developed the clinical signs of severe paralyzing encephalomyelitis but were eventually able to control the infection and recover fully from clinical illness. Recovery in this system was T cell dependent and associated with a dramatic reduction in viral titers within the CNS, followed by viral persistence in the brain. Further comparison of the relative roles of T cell subpopulations within this system revealed that CD4(+) T cells were better producers of gamma interferon (IFN-gamma) than CD8(+) T cells and were more effective at controlling VEEV within the CNS. Overall, these results suggest that T cells, especially CD4(+) T cells, can successfully control VEEV infection within the CNS and facilitate recovery from a severe viral encephalomyelitis.


Asunto(s)
Anticuerpos Antivirales/inmunología , Virus de la Encefalitis Equina Venezolana/inmunología , Encefalomielitis Equina Venezolana/inmunología , Linfocitos T/inmunología , Animales , Encéfalo/virología , Encefalomielitis Equina Venezolana/patología , Femenino , Humanos , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subgrupos de Linfocitos T/inmunología , Carga Viral
11.
J Virol ; 83(9): 4275-86, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19225006

RESUMEN

Venezuelan equine encephalitis virus (VEE) replicon particles (VRP) were used to model the initial phase of VEE-induced encephalitis in the mouse brain. VRP can target and infect cells as VEE, but VRP do not propagate beyond the first infected cell due to the absence of the structural genes. Direct intracranial inoculation of VRP into mice induced acute encephalitis with signs similar to the neuronal phase of wild-type VEE infection and other models of virus-induced encephalitis. Using the previously established VRP-mRNP tagging system, a new method to distinguish the host responses in infected cells from those in uninfected bystander cell populations, we detected a robust and rapid innate immune response in the central nervous system (CNS) by infected neurons and uninfected bystander cells. Moreover, this innate immune response in the CNS compromised blood-brain barrier integrity, created an inflammatory response, and directed an adaptive immune response characterized by proliferation and activation of microglia cells and infiltration of inflammatory monocytes, in addition to CD4(+) and CD8(+) T lymphocytes. Taken together, these data suggest that a naïve CNS has an intrinsic potential to induce an innate immune response that could be crucial to the outcome of the infection by determining the composition and dynamics of the adaptive immune response. Furthermore, these results establish a model for neurotropic virus infection to identify host and viral factors that contribute to invasion of the brain, the mechanism(s) whereby the adaptive immune response can clear the infection, and the role of the host innate response in these processes.


Asunto(s)
Modelos Animales de Enfermedad , Virus de la Encefalitis Equina Venezolana/crecimiento & desarrollo , Virus de la Encefalitis Equina Venezolana/patogenicidad , Encefalomielitis Equina Venezolana/virología , Virión/crecimiento & desarrollo , Animales , Proliferación Celular , Citocinas/metabolismo , Virus de la Encefalitis Equina Venezolana/genética , Virus de la Encefalitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/patología , Femenino , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Endogámicos BALB C , Microglía/metabolismo , Microglía/patología , ARN Viral/genética , Virión/genética
12.
J Virol ; 83(23): 12432-42, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19793821

RESUMEN

The host innate immune response provides a critical first line of defense against invading pathogens, inducing an antiviral state to impede the spread of infection. While numerous studies have documented antiviral responses within actively infected tissues, few have described the earliest innate response induced systemically by infection. Here, utilizing Venezuelan equine encephalitis virus (VEE) replicon particles (VRP) to limit infection to the initially infected cells in vivo, a rapid activation of the antiviral response was demonstrated not only within the murine draining lymph node, where replication was confined, but also within distal tissues. In the liver and brain, expression of interferon-stimulated genes was detected by 1 to 3 h following VRP footpad inoculation, reaching peak expression of >100-fold over that in mock-infected animals. Moreover, mice receiving a VRP footpad inoculation 6, 12, or 24 h prior to an otherwise lethal VEE footpad challenge were completely protected from death, including a drastic reduction in challenge virus titers. VRP pretreatment also provided protection from intranasal VEE challenge and extended the average survival time following intracranial challenge. Signaling through the interferon receptor was necessary for antiviral gene induction and protection from VEE challenge. However, VRP pretreatment failed to protect mice from a heterologous, lethal challenge with vesicular stomatitis virus, yet conferred protection following challenge with influenza virus. Collectively, these results document a rapid modulation of the host innate response within hours of infection, capable of rapidly alerting the entire animal to pathogen invasion and leading to protection from viral disease.


Asunto(s)
Encéfalo/inmunología , Virus de la Encefalitis Equina Venezolana/inmunología , Encefalomielitis Equina Venezolana/prevención & control , Inmunidad Innata , Hígado/inmunología , Ganglios Linfáticos/inmunología , Animales , Encéfalo/virología , Femenino , Perfilación de la Expresión Génica , Interferones/inmunología , Hígado/virología , Ganglios Linfáticos/virología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Rhabdoviridae/prevención & control , Análisis de Supervivencia
13.
J Virol ; 83(20): 10571-81, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19656875

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is an important human and veterinary pathogen causing sporadic epizootic outbreaks of potentially fatal encephalitis. The type I interferon (IFN) system plays a central role in controlling VEEV and other alphavirus infections, and IFN evasion is likely an important determinant of whether these viruses disseminate and cause disease within their hosts. Alphaviruses are thought to limit the induction of type I IFNs and IFN-stimulated genes by shutting off host cell macromolecular synthesis, which in the case of VEEV is partially mediated by the viral capsid protein. However, more specific strategies by which alphaviruses inhibit type I IFN signaling have not been characterized. Analyses of cells infected with VEEV and VEEV replicon particles (VRP) demonstrate that viral infection rapidly disrupts tyrosine phosphorylation and nuclear translocation of the transcription factor STAT1 in response to both IFN-beta and IFN-gamma. This effect was independent of host shutoff and expression of viral capsid, suggesting that VEEV uses novel mechanisms to interfere with type I and type II IFN signaling. Furthermore, at times when STAT1 activation was efficiently inhibited, VRP infection did not limit tyrosine phosphorylation of Jak1, Tyk2, or STAT2 after IFN-beta treatment but did inhibit Jak1 and Jak2 activation in response to IFN-gamma, suggesting that VEEV interferes with STAT1 activation by the type I and II receptor complexes through distinct mechanisms. Identification of the viral requirements for this novel STAT1 inhibition will further our understanding of alphavirus molecular pathogenesis and may provide insights into effective alphavirus-based vaccine design.


Asunto(s)
Virus de la Encefalitis Equina Venezolana/patogenicidad , Factor de Transcripción STAT1/antagonistas & inhibidores , Transducción de Señal , Animales , Chlorocebus aethiops , Cricetinae , Células HeLa , Humanos , Interferón beta/antagonistas & inhibidores , Interferón beta/inmunología , Interferón gamma/antagonistas & inhibidores , Interferón gamma/inmunología , Fosforilación , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/fisiología , Tirosina/metabolismo , Células Vero
14.
Cell Rep ; 31(4): 107587, 2020 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-32348764

RESUMEN

Host genetic factors play a fundamental role in regulating humoral immunity to viral infection, including influenza A virus (IAV). Here, we utilize the Collaborative Cross (CC), a mouse genetic reference population, to study genetic regulation of variation in antibody response following IAV infection. CC mice show significant heritable variation in the magnitude, kinetics, and composition of IAV-specific antibody response. We map 23 genetic loci associated with this variation. Analysis of a subset of these loci finds that they broadly affect the antibody response to IAV as well as other viruses. Candidate genes are identified based on predicted variant consequences and haplotype-specific expression patterns, and several show overlap with genes identified in human mapping studies. These findings demonstrate that the host antibody response to IAV infection is under complex genetic control and highlight the utility of the CC in modeling and identifying genetic factors with translational relevance to human health and disease.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Gripe Humana/genética , Replicación Viral/genética , Humanos
15.
J Virol ; 81(24): 13412-23, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17913817

RESUMEN

A novel genetic vaccine that is based on a Venezuelan equine encephalitis virus (VEE) replicon launched from plasmid DNA is described. The plasmid encodes a VEE replicon under the transcriptional control of the cytomegalovirus immediate-early promoter (VEE DNA). The VEE DNA consistently expressed 3- to 15-fold more green fluorescent protein in vitro than did a conventional DNA vaccine. Furthermore, transfection with the DNA-launched VEE replicon induced apoptosis and type I interferon production. Inoculation of mice with VEE DNA encoding human immunodeficiency virus type 1 gp160 significantly increased humoral responses by several orders of magnitude compared to an equal dose of a conventional DNA vaccine. These increases were also observed at 10- and 100-fold-lower doses of the VEE DNA. Cellular immune responses measured by gamma interferon and interleukin 2 enzyme-linked immunospot assay were significantly higher in mice immunized with the VEE DNA at decreased doses. The immune responses induced by the VEE DNA-encoded antigen, however, were independent of an intact type I interferon signaling pathway. Moreover, the DNA-launched VEE replicon induced an efficient prime to a VEE replicon particle (VRP) boost, increasing humoral and cellular immunity by at least 1 order of magnitude compared to VEE DNA only. Importantly, immunization with VEE DNA, as opposed to VRP, did not induce any anti-VRP neutralizing antibodies. Increased potency of DNA vaccines and reduced vector immunity may ultimately have an impact on the design of vaccination strategies in humans.


Asunto(s)
Anticuerpos Antivirales/sangre , Virus de la Encefalitis Equina Venezolana/genética , Virus de la Encefalitis Equina Venezolana/inmunología , Interferón Tipo I/biosíntesis , Interleucina-2/biosíntesis , Plásmidos/genética , Replicón/inmunología , Vacunas de ADN/inmunología , Animales , Apoptosis , Línea Celular , Chlorocebus aethiops , Vectores Genéticos , Proteínas gp160 de Envoltorio del VIH/genética , Proteínas gp160 de Envoltorio del VIH/inmunología , VIH-1/genética , Humanos , Inmunización , Inmunoglobulina G/sangre , Células L , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células 3T3 NIH , Regiones Promotoras Genéticas , Replicón/genética , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Células Vero
16.
Virology ; 515: 250-260, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29324290

RESUMEN

Mannose binding lectin (MBL) generally plays a protective role during viral infection, yet MBL-mediated complement activation promotes Ross River virus (RRV)-induced inflammatory tissue destruction, contributing to arthritis and myositis. As MBL binds to carbohydrates, we hypothesized that N-linked glycans on the RRV envelope glycoproteins act as ligands for MBL. Using a panel of RRV mutants lacking the envelope N-linked glycans, we found that MBL deposition onto infected cells was dependent on the E2 glycans. Moreover, the glycan-deficient viruses exhibited reduced disease and tissue damage in a mouse model of RRV-induced myositis compared to wild-type RRV, despite similar viral load and inflammatory infiltrates within the skeletal muscle. Instead, the reduced disease induced by glycan-deficient viruses was linked to decreased MBL deposition and complement activation within inflamed tissues. These results demonstrate that the viral N-linked glycans promote MBL deposition and complement activation onto RRV-infected cells, contributing to the development of RRV-induced myositis.


Asunto(s)
Infecciones por Alphavirus/inmunología , Proteínas del Sistema Complemento/inmunología , Polisacáridos/inmunología , Virus del Río Ross/inmunología , Proteínas del Envoltorio Viral/inmunología , Infecciones por Alphavirus/virología , Animales , Activación de Complemento , Modelos Animales de Enfermedad , Humanos , Lectina de Unión a Manosa/inmunología , Ratones Endogámicos C57BL , Polisacáridos/química , Virus del Río Ross/genética , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
17.
G3 (Bethesda) ; 8(2): 427-445, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29187420

RESUMEN

Influenza A virus (IAV) is a respiratory pathogen that causes substantial morbidity and mortality during both seasonal and pandemic outbreaks. Infection outcomes in unexposed populations are affected by host genetics, but the host genetic architecture is not well understood. Here, we obtain a broad view of how heritable factors affect a mouse model of response to IAV infection using an 8 × 8 diallel of the eight inbred founder strains of the Collaborative Cross (CC). Expanding on a prior statistical framework for modeling treatment response in diallels, we explore how a range of heritable effects modify acute host response to IAV through 4 d postinfection. Heritable effects in aggregate explained ∼57% of the variance in IAV-induced weight loss. Much of this was attributable to a pattern of additive effects that became more prominent through day 4 postinfection and was consistent with previous reports of antiinfluenza myxovirus resistance 1 (Mx1) polymorphisms segregating between these strains; these additive effects largely recapitulated haplotype effects observed at the Mx1 locus in a previous study of the incipient CC, and are also replicated here in a CC recombinant intercross population. Genetic dominance of protective Mx1 haplotypes was observed to differ by subspecies of origin: relative to the domesticus null Mx1 allele, musculus acts dominantly whereas castaneus acts additively. After controlling for Mx1, heritable effects, though less distinct, accounted for ∼34% of the phenotypic variance. Implications for future mapping studies are discussed.


Asunto(s)
Teorema de Bayes , Predisposición Genética a la Enfermedad/genética , Proteínas de Resistencia a Mixovirus/genética , Infecciones por Orthomyxoviridae/genética , Animales , Modelos Animales de Enfermedad , Haplotipos , Humanos , Virus de la Influenza A/fisiología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Endogámicos , Infecciones por Orthomyxoviridae/virología , Fenotipo , Especificidad de la Especie
18.
mBio ; 8(6)2017 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-29138302

RESUMEN

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus responsible for several significant outbreaks of debilitating acute and chronic arthritis and arthralgia over the past decade. These include a recent outbreak in the Caribbean islands and the Americas that caused more than 1 million cases of viral arthralgia. Despite the major impact of CHIKV on global health, viral determinants that promote CHIKV-induced disease are incompletely understood. Most CHIKV strains contain a conserved opal stop codon at the end of the viral nsP3 gene. However, CHIKV strains that encode an arginine codon in place of the opal stop codon have been described, and deep-sequencing analysis of a CHIKV isolate from the Caribbean identified both arginine and opal variants within this strain. Therefore, we hypothesized that the introduction of the arginine mutation in place of the opal termination codon may influence CHIKV virulence. We tested this by introducing the arginine mutation into a well-characterized infectious clone of a CHIKV strain from Sri Lanka and designated this virus Opal524R. This mutation did not impair viral replication kinetics in vitro or in vivo Despite this, the Opal524R virus induced significantly less swelling, inflammation, and damage within the feet and ankles of infected mice. Further, we observed delayed induction of proinflammatory cytokines and chemokines, as well as reduced CD4+ T cell and NK cell recruitment compared to those in the parental strain. Therefore, the opal termination codon plays an important role in CHIKV pathogenesis, independently of effects on viral replication.IMPORTANCE Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that causes significant outbreaks of viral arthralgia. Studies with CHIKV and other alphaviruses demonstrated that the opal termination codon within nsP3 is highly conserved. However, some strains of CHIKV and other alphaviruses contain mutations in the opal termination codon. These mutations alter the virulence of related alphaviruses in mammalian and mosquito hosts. Here, we report that a clinical isolate of a CHIKV strain from the recent outbreak in the Caribbean islands contains a mixture of viruses encoding either the opal termination codon or an arginine mutation. Mutating the opal stop codon to an arginine residue attenuates CHIKV-induced disease in a mouse model. Compared to infection with the opal-containing parental virus, infection with the arginine mutant causes limited swelling and inflammation, as well as dampened recruitment of immune mediators of pathology, including CD4+ T cells and NK cells. We propose that the opal termination codon plays an essential role in the induction of severe CHIKV disease.


Asunto(s)
Artritis/patología , Fiebre Chikungunya/patología , Virus Chikungunya/patogenicidad , Codón de Terminación , Mutación , Proteínas no Estructurales Virales/genética , Factores de Virulencia/genética , Animales , Arginina/genética , Artritis/virología , Fiebre Chikungunya/virología , Virus Chikungunya/fisiología , Modelos Animales de Enfermedad , Ratones , Replicación Viral
19.
G3 (Bethesda) ; 4(8): 1429-44, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24902603

RESUMEN

Genetic variation between diverse mouse species is well-characterized, yet existing knowledge of the mouse transcriptome comes largely from one mouse strain (C57BL/6J). As such, it is unlikely to reflect the transcriptional complexity of the mouse species. Gene transcription is dynamic and condition-specific; therefore, to better understand the mouse transcriptional response to respiratory virus infection, we infected the eight founder strains of the Collaborative Cross with either influenza A virus or severe acute respiratory syndrome coronavirus and sequenced lung RNA samples at 2 and 4 days after infection. We found numerous instances of transcripts that were not present in the C57BL/6J reference annotation, indicating that a nontrivial proportion of the mouse genome is transcribed but poorly annotated. Of these novel transcripts, 2150 could be aligned to human or rat genomes, but not to existing mouse genomes, suggesting functionally conserved sequences not yet recorded in mouse genomes. We also found that respiratory virus infection induced differential expression of 4287 splicing junctions, resulting in strain-specific isoform expression. Of these, 59 were influenced by strain-specific mutations within 2 base pairs of key intron-exon boundaries, suggesting cis-regulated expression. Our results reveal the complexity of the transcriptional response to viral infection, previously undocumented genomic elements, and extensive diversity in the response across mouse strains. These findings identify hitherto unexplored transcriptional patterns and undocumented transcripts in genetically diverse mice. Host genetic variation drives the complexity and diversity of the host response by eliciting starkly different transcriptional profiles in response to a viral infection.


Asunto(s)
Infecciones por Orthomyxoviridae/genética , Síndrome Respiratorio Agudo Grave/genética , Animales , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genoma , Virus de la Influenza A , Pulmón/metabolismo , Pulmón/virología , Ratones , Infecciones por Orthomyxoviridae/virología , Fenotipo , ARN/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , Análisis de Secuencia de ARN , Síndrome Respiratorio Agudo Grave/virología , Especificidad de la Especie , Carga Viral
20.
Cancer Discov ; 4(12): 1387-97, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25266735

RESUMEN

UNLABELLED: Whether dietary fiber protects against colorectal cancer is controversial because of conflicting results from human epidemiologic studies. However, these studies and mouse models of colorectal cancer have not controlled the composition of gut microbiota, which ferment fiber into short-chain fatty acids such as butyrate. Butyrate is noteworthy because it has energetic and epigenetic functions in colonocytes and tumor-suppressive properties in colorectal cancer cell lines. We used gnotobiotic mouse models colonized with wild-type or mutant strains of a butyrate-producing bacterium to demonstrate that fiber does have a potent tumor-suppressive effect but in a microbiota- and butyrate-dependent manner. Furthermore, due to the Warburg effect, butyrate was metabolized less in tumors where it accumulated and functioned as a histone deacetylase (HDAC) inhibitor to stimulate histone acetylation and affect apoptosis and cell proliferation. To support the relevance of this mechanism in human cancer, we demonstrate that butyrate and histone-acetylation levels are elevated in colorectal adenocarcinomas compared with normal colonic tissues. SIGNIFICANCE: These results, which link diet and microbiota to a tumor-suppressive metabolite, provide insight into conflicting epidemiologic findings and suggest that probiotic/prebiotic strategies can modulate an endogenous HDAC inhibitor for anticancer chemoprevention without the adverse effects associated with synthetic HDAC inhibitors used in chemotherapy.


Asunto(s)
Butiratos/metabolismo , Transformación Celular Neoplásica , Neoplasias Colorrectales/etiología , Fibras de la Dieta , Vida Libre de Gérmenes , Microbiota , Animales , Carcinógenos/administración & dosificación , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Humanos , Mucosa Intestinal/patología , Ratones , Clasificación del Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA