Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Appl Microbiol ; 135(2)2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38323434

RESUMO

Arthritis and periodontitis are inflammatory diseases that share several immunopathogenic features. The expansion in the study of virus-induced arthritis has shed light on how this condition could impact other parts of the human body, including the mouth. Viral arthritis is an inflammatory joint disease caused by several viruses, most notably the alphaviruses Chikungunya virus (CHIKV), Sindbis virus (SINV), Ross River virus (RRV), Mayaro virus (MAYV), and O'nyong'nyong virus (ONNV). These viruses can induce an upsurge of matrix metalloproteinases and immune-inflammatory mediators such as Interleukin-6 (IL6), IL-1ß, tumor necrosis factor, chemokine ligand 2, and receptor activator of nuclear factor kappa-B ligand in the joint and serum of infected individuals. This can lead to the influx of inflammatory cells to the joints and associated muscles as well as osteoclast activation and differentiation, culminating in clinical signs of swelling, pain, and bone resorption. Moreover, several data indicate that these viral infections can affect other sites of the body, including the mouth. The human oral cavity is a rich and diverse microbial ecosystem, and viral infection can disrupt the balance of microbial species, causing local dysbiosis. Such events can result in oral mucosal damage and gingival bleeding, which are indicative of periodontitis. Additionally, infection by RRV, CHIKV, SINV, MAYV, or ONNV can trigger the formation of osteoclasts and upregulate pro-osteoclastogenic inflammatory mediators, interfering with osteoclast activation. As a result, these viruses may be linked to systemic conditions, including oral manifestations. Therefore, this review focuses on the involvement of alphavirus infections in joint and oral health, acting as potential agents associated with oral mucosal inflammation and alveolar bone loss. The findings of this review demonstrate how alphavirus infections could be linked to the comorbidity between arthritis and periodontitis and may provide a better understanding of potential therapeutic management for both conditions.


Assuntos
Infecções por Alphavirus , Artrite , Vírus Chikungunya , Periodontite , Humanos , Infecções por Alphavirus/tratamento farmacológico , Infecções por Alphavirus/patologia , Vírus Chikungunya/fisiologia , Mediadores da Inflamação/uso terapêutico , Ligantes , Ross River virus/fisiologia
2.
PLoS Pathog ; 16(5): e1008517, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32365139

RESUMO

Ross River fever is a mosquito-transmitted viral disease that is endemic to Australia and the surrounding Pacific Islands. Ross River virus (RRV) belongs to the arthritogenic group of alphaviruses, which largely cause disease characterized by debilitating polyarthritis, rash, and fever. There is no specific treatment or licensed vaccine available, and the mechanisms of protective humoral immunity in humans are poorly understood. Here, we describe naturally occurring human mAbs specific to RRV, isolated from subjects with a prior natural infection. These mAbs potently neutralize RRV infectivity in cell culture and block infection through multiple mechanisms, including prevention of viral attachment, entry, and fusion. Some of the most potently neutralizing mAbs inhibited binding of RRV to Mxra8, a recently discovered alpahvirus receptor. Epitope mapping studies identified the A and B domains of the RRV E2 protein as the major antigenic sites for the human neutralizing antibody response. In experiments in mice, these mAbs were protective against cinical disease and reduced viral burden in multiple tissues, suggesting a potential therapeutic use for humans.


Assuntos
Infecções por Alphavirus/prevenção & controle , Anticorpos Monoclonais , Anticorpos Neutralizantes , Anticorpos Antivirais , Proteínas do Capsídeo/imunologia , Epitopos/imunologia , Ross River virus/imunologia , Proteínas do Envelope Viral/imunologia , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/patologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/farmacologia , Chlorocebus aethiops , Feminino , Humanos , Camundongos , Pessoa de Meia-Idade , Células Vero
3.
mBio ; 11(2)2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32127460

RESUMO

Arthritogenic alphaviruses such as Ross River and Chikungunya viruses cause debilitating muscle and joint pain and pose significant challenges in the light of recent outbreaks. How host immune responses are orchestrated after alphaviral infections and lead to musculoskeletal inflammation remains poorly understood. Here, we show that myositis induced by Ross River virus (RRV) infection is driven by CD11bhi Ly6Chi inflammatory monocytes and followed by the establishment of a CD11bhi Ly6Clo CX3CR1+ macrophage population in the muscle upon recovery. Selective modulation of CD11bhi Ly6Chi monocyte migration to infected muscle using immune-modifying microparticles (IMP) reduced disease score, tissue damage, and inflammation and promoted the accumulation of CX3CR1+ macrophages, enhancing recovery and resolution. Here, we detail the role of immune pathology, describing a poorly characterized muscle macrophage subset as part of the dynamics of alphavirus-induced myositis and tissue recovery and identify IMP as an effective immunomodulatory approach. Given the lack of specific treatments available for alphavirus-induced pathologies, this study highlights a therapeutic potential for simple immune modulation by IMP in infected individuals in the event of large alphavirus outbreaks.IMPORTANCE Arthritogenic alphaviruses cause debilitating inflammatory disease, and current therapies are restricted to palliative approaches. Here, we show that following monocyte-driven muscle inflammation, tissue recovery is associated with the accumulation of CX3CR1+ macrophages in the muscle. Modulating inflammatory monocyte infiltration using immune-modifying microparticles (IMP) reduced tissue damage and inflammation and enhanced the formation of tissue repair-associated CX3CR1+ macrophages in the muscle. This shows that modulating key effectors of viral inflammation using microparticles can alter the outcome of disease by facilitating the accumulation of macrophage subsets associated with tissue repair.


Assuntos
Infecções por Alphavirus/metabolismo , Infecções por Alphavirus/virologia , Receptor 1 de Quimiocina CX3C/genética , Monócitos/metabolismo , Miosite/etiologia , Miosite/metabolismo , Cicatrização , Infecções por Alphavirus/patologia , Animais , Biomarcadores , Biópsia , Receptor 1 de Quimiocina CX3C/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Imunomodulação/genética , Imunofenotipagem , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Transgênicos , Monócitos/imunologia , Monócitos/virologia , Miosite/patologia
4.
J Virol ; 92(23)2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30232189

RESUMO

Sindbis virus (SINV) is a representative member of the Alphavirus genus in the Togaviridae family. The hallmark of SINV replication in vertebrate cells is a rapid development of the cytopathic effect (CPE), which usually occurs within 24 h postinfection. Mechanistic understanding of CPE might lead to development of new prophylactic vaccines and therapeutic means against alphavirus infections. However, development of noncytopathic SINV variants and those of other Old World alphaviruses was always highly inefficient and usually resulted in selection of mutants demonstrating poor replication of the viral genome and transcription of subgenomic RNA. This likely caused a nonspecific negative effect on the rates of CPE development. The results of this study demonstrate that CPE induced by SINV and likely by other Old World alphaviruses is a multicomponent process, in which transcriptional and translational shutoffs are the key contributors. Inhibition of cellular transcription and translation is determined by SINV nsP2 and nsP3 proteins, respectively. Defined mutations in the nsP2-specific peptide between amino acids (aa) 674 and 688 prevent virus-induced degradation of the catalytic subunit of cellular-DNA-dependent RNA polymerase II and transcription inhibition and make SINV a strong type I interferon (IFN) inducer without affecting its replication rates. Mutations in the nsP3 macrodomain, which were demonstrated to inhibit its mono-ADP-ribosylhydrolase activity, downregulate the second component of CPE development, inhibition of cellular translation, and also have no effect on virus replication rates. Only the combination of nsP2- and nsP3-specific mutations in the SINV genome has a dramatic negative effect on the ability of virus to induce CPE.IMPORTANCE Alphaviruses are a group of important human and animal pathogens with worldwide distribution. Their characteristic feature is a highly cytopathic phenotype in cells of vertebrate origin. The molecular mechanism of CPE remains poorly understood. In this study, by using Sindbis virus (SINV) as a model of the Old World alphaviruses, we demonstrated that SINV-specific CPE is redundantly determined by viral nsP2 and nsP3 proteins. NsP2 induces the global transcriptional shutoff, and this nuclear function can be abolished by the mutations of the small, surface-exposed peptide in the nsP2 protease domain. NsP3, in turn, determines the development of translational shutoff, and this activity depends on nsP3 macrodomain-associated mono-ADP-ribosylhydrolase activity. A combination of defined mutations in nsP2 and nsP3, which abolish SINV-induced transcription and translation inhibition, in the same viral genome does not affect SINV replication rates but makes it noncytopathic and a potent inducer of type I interferon.


Assuntos
Infecções por Alphavirus/patologia , Cisteína Endopeptidases/metabolismo , Efeito Citopatogênico Viral , Biossíntese de Proteínas , Sindbis virus/fisiologia , Transcrição Gênica , Proteínas não Estruturais Virais/metabolismo , Infecções por Alphavirus/genética , Infecções por Alphavirus/metabolismo , Infecções por Alphavirus/virologia , Animais , Cisteína Endopeptidases/genética , Genoma Viral , Camundongos , Células NIH 3T3 , Proteínas não Estruturais Virais/genética , Vírion , Replicação Viral
5.
mBio ; 9(4)2018 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-30131356

RESUMO

Infection with Ross River virus (RRV) causes debilitating polyarthritis and arthralgia in individuals. Alphaviruses are highly sensitive to type I interferon (IFN). Mutations at the conserved P3 position of the cleavage site between nonstructural protein 1 (nsP1) and nsP2 (1/2 site) modulate type I IFN induction for both RRV and Sindbis virus (SINV). We constructed and characterized RRV-T48A534V, a mutant harboring an A534V substitution in the P1 position of the 1/2 site, and compared it to parental RRV-T48 and to RRV-T48A532V, SINVI538 and SINVT538 harboring different substitutions in the same region. A534V substitution resulted in impaired processing of RRV nonstructural polyprotein and in elevated production of replicase-generated pathogen-associated molecular pattern (PAMP) RNAs that induce expression of type I IFN. Both A532V and A534V substitutions affected synthesis of viral RNAs, though the effects of these closely located mutations were drastically different affecting mostly either the viral negative-strand RNA or genomic and subgenomic RNA levels, respectively. Synthesis of PAMP RNAs was also observed for SINV replicase, and it was increased by I538T substitution. In comparison to RRV-T48, RRV-T48A534V was attenuated in vitro and in vivo Interestingly, when type I IFN-deficient cells and type I IFN receptor-deficient mice were infected with RRV-T48 or RRV-T48A534V, differences between these viruses were no longer apparent. Compared to RRV-T48, RRV-T48A534V infection was associated with increased upregulation of type I IFN signaling proteins. We demonstrate novel mechanisms by which the A534V mutation affect viral nonstructural polyprotein processing that can impact PAMP RNA production, type I IFN induction/sensitivity, and disease.IMPORTANCE This study gives further insight into mechanisms of type I IFN modulation by the medically important alphaviruses Ross River virus (RRV) and Sindbis virus (SINV). By characterizing attenuated RRV mutants, the crucial role of amino acid residues in P1 and P3 positions (the first and third amino acid residues preceding the scissile bond) of the cleavage site between nsP1 and nsP2 regions was highlighted. The study uncovers a unique relationship between alphavirus nonstructural polyprotein processing, RNA replication, production of different types of pathogen-associated molecular pattern (PAMP) RNAs, type I IFN induction, and disease pathogenesis. This study also highlights the importance of the host innate immune response in RRV infections. The viral determinants of type I IFN modulation provide potential drug targets for clinical treatment of alphaviral disease and offer new approaches for rational attenuation of alphaviruses for construction of vaccine candidates.


Assuntos
Interferons/metabolismo , Proteínas Mutantes/imunologia , Mutação de Sentido Incorreto , Poliproteínas/metabolismo , RNA Viral/imunologia , Ross River virus/patogenicidade , Proteínas não Estruturais Virais/metabolismo , Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Animais , Antivirais/metabolismo , Modelos Animais de Doenças , Camundongos , Proteínas Mutantes/genética , Poliproteínas/genética , RNA Viral/metabolismo , Ross River virus/genética , Ross River virus/imunologia , Sindbis virus/genética , Sindbis virus/imunologia , Sindbis virus/patogenicidade , Proteínas não Estruturais Virais/genética , Virulência
6.
J Virol ; 92(7)2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29321331

RESUMO

Sindbis virus (SINV) infection of neurons in the brain and spinal cord in mice provides a model system for investigating recovery from encephalomyelitis and antibody-mediated clearance of virus from the central nervous system (CNS). To determine the roles of IgM and IgG in recovery, we compared the responses of immunoglobulin-deficient activation-induced adenosine deaminase-deficient (AID-/-), secretory IgM-deficient (sIgM-/-), and AID-/- sIgM-/- double-knockout (DKO) mice with those of wild-type (WT) C57BL/6 mice for disease, clearance of infectious virus and viral RNA from brain and spinal cord, antibody responses, and B cell infiltration into the CNS. Because AID is essential for immunoglobulin class switch recombination and somatic hypermutation, AID-/- mice produce only germ line IgM, while sIgM-/- mice secrete IgG but no IgM and DKO mice produce no secreted immunoglobulin. After intracerebral infection with the TE strain of SINV, most mice recovered. Development of neurologic disease occurred slightly later in sIgM-/- mice, but disease severity, weight loss, and survival were similar between the groups. AID-/- mice produced high levels of SINV-specific IgM, while sIgM-/- mice produced no IgM and high levels of IgG2a compared to WT mice. All mice cleared infectious virus from the spinal cord, but DKO mice failed to clear infectious virus from brain and had higher levels of viral RNA in the CNS late after infection. The numbers of infected cells and the amount of cell death in brain were comparable. We conclude that antibody is required and that either germ line IgM or IgG is sufficient for clearance of virus from the CNS.IMPORTANCE Mosquito-borne alphaviruses that infect neurons can cause fatal encephalomyelitis. Recovery requires a mechanism for the immune system to clear virus from infected neurons without harming the infected cells. Antiviral antibody has previously been shown to be a noncytolytic means for alphavirus clearance. Antibody-secreting cells enter the nervous system after infection and produce antiviral IgM before IgG. Clinical studies of human viral encephalomyelitis suggest that prompt production of IgM is associated with recovery, but it was not known whether IgM is effective for clearance. Our studies used mice deficient in production of IgM, IgG, or both to characterize the antibody necessary for alphavirus clearance. All mice developed similar signs of neurologic disease and recovered from infection. Antibody was necessary for virus clearance from the brain, and either early germ line IgM or IgG was sufficient. These studies support the clinical observation that prompt production of antiviral antibody is a determinant of outcome.


Assuntos
Infecções por Alphavirus/imunologia , Anticorpos Antivirais/imunologia , Encéfalo/imunologia , Infecções do Sistema Nervoso Central/imunologia , Imunoglobulina M/imunologia , Sindbis virus/imunologia , Infecções por Alphavirus/genética , Infecções por Alphavirus/patologia , Animais , Anticorpos Antivirais/genética , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular , Infecções do Sistema Nervoso Central/genética , Infecções do Sistema Nervoso Central/patologia , Cricetinae , Citidina Desaminase/deficiência , Feminino , Imunoglobulina M/genética , Camundongos , Camundongos Knockout , Sindbis virus/genética
7.
Microbes Infect ; 19(11): 496-504, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28754345

RESUMO

Ross River virus (RRV) is an arthitogenic alphavirus capable of causing outbreaks of debilitating musculoskeletal inflammatory disease in humans. RRV is the most common mosquito-borne disease in Australia, with outbreaks of RRV generally occurring during seasonal wet and warm conditions. Patients with Ross River virus disease (RRVD) typically present with fever, polyarthralgia, myalgia and a maculopapular erythematous rash. Treatment of the disease is usually palliative with no licensed vaccines or antiviral therapies currently available. In an effort to better inform therapeutic design, much progress has been made to understand the pathogenesis of RRVD. Progress has been largely driven by clinical evaluations supported by research using established murine models of RRVD, able to accurately replicate human disease. In this review we describe RRVD pathogenesis and the role of the host immune response, with particular focus on insights from studying animal models. We also discuss prospects for effective vaccines, preclinical development of therapeutic strategies and raise important questions for future RRV research.


Assuntos
Infecções por Alphavirus/diagnóstico , Ross River virus , Infecções por Alphavirus/patologia , Infecções por Alphavirus/terapia , Infecções por Alphavirus/virologia , Animais , Modelos Animais de Doenças , Humanos , Cuidados Paliativos/métodos
8.
J Neuroimmunol ; 308: 118-130, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28501330

RESUMO

Viral infections of the central nervous system (CNS) are often associated with blood-brain barrier (BBB) disruption, yet the impact of virus replication and immune cell recruitment on BBB integrity are incompletely understood. Using two-photon microscopy, we demonstrate that Venezuelan equine encephalitis virus (VEEV) strain TC83-GFP, a GFP expressing, attenuated strain with a G3A mutation within the 5' UTR that is associated with increased sensitivity to type I interferons (IFNs), does not directly impact BBB permeability. Following intranasal infection of both wild-type and IFN-induced protein with tetratricopeptide repeats 1 (IFIT1)-deficient mice, which fail to block TC83-specific RNA translation, virus spreads to the olfactory bulb and cortex via migration along axonal tracts of neurons originating from the olfactory neuroepithelium. Global dissemination of virus in the CNS by 2days post-infection (dpi) was associated with increased BBB permeability in the olfactory bulb, but not in the cortex or hindbrain, where permeability only increased after the recruitment of CX3CR1+ and CCR2+ mononuclear cells on 6 dpi, which corresponded with tight junction loss and claudin 5 redistribution. Importantly, despite higher levels of viral replication, similar results were obtained in IFIT1-deficient mice. These findings indicate that TC83 gains CNS access via anterograde axonal migration without directly altering BBB function and that mononuclear and endothelial cell interactions may underlie BBB disruption during alphavirus encephalitis.


Assuntos
Infecções por Alphavirus/patologia , Barreira Hematoencefálica/fisiopatologia , Encéfalo/metabolismo , Encéfalo/virologia , Replicação Viral/fisiologia , Complexo 1 de Proteínas Adaptadoras/genética , Complexo 1 de Proteínas Adaptadoras/metabolismo , Infecções por Alphavirus/genética , Animais , Animais Recém-Nascidos , Barreira Hematoencefálica/ultraestrutura , Barreira Hematoencefálica/virologia , Receptor 1 de Quimiocina CX3C , Permeabilidade Capilar/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Cricetinae , Modelos Animais de Doenças , Vírus da Encefalite Equina Venezuelana/fisiologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Células Epiteliais/ultraestrutura , Células Epiteliais/virologia , Regulação da Expressão Gênica/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Internalização do Vírus
9.
Dis Model Mech ; 10(7): 847-857, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28483796

RESUMO

Alphaviruses, such as chikungunya virus (CHIKV) and Sindbis virus (SINV), are vector-borne pathogens that cause acute illnesses in humans and are sometimes associated with neuropathies, especially in infants and elderly patients. Little is known about their mechanism of entry into the central nervous system (CNS), even for SINV, which has been used extensively as a model for viral encephalopathies. We previously established a CHIKV infection model in the optically transparent zebrafish larva; here we describe a new SINV infection model in this host. We imaged in vivo the onset and progression of the infection caused by intravenous SINV inoculation. Similar to that described for CHIKV, infection in the periphery was detected early and was transient, whereas CNS infection started at later time points and was persistent or progressive. We then tested the possible mechanisms of neuroinvasion by CHIKV and SINV. Neither virus relied on macrophage-mediated transport to access the CNS. CHIKV, but not SINV, always infects endothelial cells of the brain vasculature. By contrast, axonal transport was much more efficient with SINV than CHIKV, both from the periphery to the CNS and between neural tissues. Thus, the preferred mechanisms of neuroinvasion by these two related viruses are distinct, providing a powerful imaging-friendly system to compare mechanisms and prevention methods of encephalopathies.


Assuntos
Vírus Chikungunya/fisiologia , Imageamento Tridimensional , Sistema Nervoso/virologia , Sindbis virus/fisiologia , Internalização do Vírus , Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Animais , Transporte Axonal , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/virologia , Febre de Chikungunya/patologia , Febre de Chikungunya/virologia , Células Endoteliais/patologia , Células Endoteliais/virologia , Larva/virologia , Macrófagos/metabolismo , Microvasos/patologia , Sistema Nervoso/patologia , Tropismo/fisiologia , Replicação Viral/fisiologia , Peixe-Zebra
10.
J Virol ; 90(19): 8780-94, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27440901

RESUMO

UNLABELLED: Host cells respond to viral infections by producing type I interferon (IFN), which induces the expression of hundreds of interferon-stimulated genes (ISGs). Although ISGs mediate a protective state against many pathogens, the antiviral functions of the majority of these genes have not been identified. IFITM3 is a small transmembrane ISG that restricts a broad range of viruses, including orthomyxoviruses, flaviviruses, filoviruses, and coronaviruses. Here, we show that alphavirus infection is increased in Ifitm3(-/-) and Ifitm locus deletion (Ifitm-del) fibroblasts and, reciprocally, reduced in fibroblasts transcomplemented with Ifitm3. Mechanistic studies showed that Ifitm3 did not affect viral binding or entry but inhibited pH-dependent fusion. In a murine model of chikungunya virus arthritis, Ifitm3(-/-) mice sustained greater joint swelling in the ipsilateral ankle at days 3 and 7 postinfection, and this correlated with higher levels of proinflammatory cytokines and viral burden. Flow cytometric analysis suggested that Ifitm3(-/-) macrophages from the spleen were infected at greater levels than observed in wild-type (WT) mice, results that were supported by experiments with Ifitm3(-/-) bone marrow-derived macrophages. Ifitm3(-/-) mice also were more susceptible than WT mice to lethal alphavirus infection with Venezuelan equine encephalitis virus, and this was associated with greater viral burden in multiple organs. Collectively, our data define an antiviral role for Ifitm3 in restricting infection of multiple alphaviruses. IMPORTANCE: The interferon-induced transmembrane protein 3 (IFITM3) inhibits infection of multiple families of viruses in cell culture. Compared to other viruses, much less is known about the antiviral effect of IFITM3 on alphaviruses. In this study, we characterized the antiviral activity of mouse Ifitm3 against arthritogenic and encephalitic alphaviruses using cells and animals with a targeted gene deletion of Ifitm3 as well as deficient cells transcomplemented with Ifitm3. Based on extensive virological analysis, we demonstrate greater levels of alphavirus infection and disease pathogenesis when Ifitm3 expression is absent. Our data establish an inhibitory role for Ifitm3 in controlling infection of alphaviruses.


Assuntos
Infecções por Alphavirus/imunologia , Vírus Chikungunya/imunologia , Vírus da Encefalite Equina Venezuelana/imunologia , Fatores Imunológicos/metabolismo , Proteínas de Membrana/metabolismo , Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Animais , Vírus Chikungunya/fisiologia , Citocinas/metabolismo , Modelos Animais de Doenças , Vírus da Encefalite Equina Venezuelana/fisiologia , Fibroblastos/imunologia , Fibroblastos/virologia , Deleção de Genes , Teste de Complementação Genética , Fatores Imunológicos/deficiência , Macrófagos/virologia , Proteínas de Membrana/deficiência , Camundongos , Camundongos Knockout , Carga Viral , Internalização do Vírus/efeitos dos fármacos
11.
J Virol ; 90(5): 2418-33, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26676771

RESUMO

UNLABELLED: Alphaviruses represent a diverse set of arboviruses, many of which are important pathogens. Chikungunya virus (CHIKV), an arthritis-inducing alphavirus, is the cause of a massive ongoing outbreak in the Caribbean and South America. In contrast to CHIKV, other related alphaviruses, such as Venezuelan equine encephalitis virus (VEEV) and Semliki Forest virus (SFV), can cause encephalitic disease. E2, the receptor binding protein, has been implicated as a determinant in cell tropism, host range, pathogenicity, and immunogenicity. Previous reports also have demonstrated that E2 contains residues important for host range expansions and monoclonal antibody binding; however, little is known about what role each protein domain (e.g., A, B, and C) of E2 plays on these factors. Therefore, we constructed chimeric cDNA clones between CHIKV and VEEV or SFV to probe the effect of each domain on pathogenicity in vitro and in vivo. CHIKV chimeras containing each of the domains of the E2 (ΔDomA, ΔDomB, and ΔDomC) from SFV, but not VEEV, were successfully rescued. Interestingly, while all chimeric viruses were attenuated compared to CHIKV in mice, ΔDomB virus showed similar rates of infection and dissemination in Aedes aegypti mosquitoes, suggesting differing roles for the E2 protein in different hosts. In contrast to CHIKV; ΔDomB, and to a lesser extent ΔDomA, caused neuron degeneration and demyelination in mice infected intracranially, suggesting a shift toward a phenotype similar to SFV. Thus, chimeric CHIKV/SFV provide insights on the role the alphavirus E2 protein plays on pathogenesis. IMPORTANCE: Chikungunya virus (CHIKV) has caused large outbreaks of acute and chronic arthritis throughout Africa and Southeast Asia and has now become a massive public health threat in the Americas, causing an estimated 1.2 million human cases in just over a year. No approved vaccines or antivirals exist for human use against CHIKV or any other alphavirus. Despite the threat, little is known about the role the receptor binding protein (E2) plays on disease outcome in an infected host. To study this, our laboratory generated chimeric CHIKV containing corresponding regions of the Semliki Forest virus (SFV) E2 (domains A, B, and C) substituted into the CHIKV genome. Our results demonstrate that each domain of E2 likely plays a critical, but dissimilar role in the viral life cycle. Our experiments show that manipulation of E2 domains can be useful for studies on viral pathogenesis and potentially the production of vaccines and/or antivirals.


Assuntos
Infecções por Alphavirus/patologia , Vírus Chikungunya/patogenicidade , Vírus da Encefalite Equina Venezuelana/patogenicidade , Vírus da Floresta de Semliki/patogenicidade , Proteínas do Envelope Viral/metabolismo , Aedes/virologia , Infecções por Alphavirus/virologia , Animais , Encéfalo/patologia , Vírus Chikungunya/genética , Modelos Animais de Doenças , Vírus da Encefalite Equina Venezuelana/genética , Feminino , Masculino , Camundongos Endogâmicos C57BL , Estrutura Terciária de Proteína , Vírus da Floresta de Semliki/genética , Proteínas do Envelope Viral/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
12.
J Fish Dis ; 38(3): 271-81, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25297529

RESUMO

A challenge model for pancreas disease in Atlantic salmon, Salmo salar L. fry, was developed comparing two salmonid alphavirus (SAV) subtypes: SAV1 and SAV5. Viral doses of 3 × 10(5) TCID50  mL(-1) for SAV1 and 3 × 10(4) for SAV5 were tested in triplicate tanks, each containing 450 salmon fry. Cumulative mortalities of 1.2% were recorded. Titres of virus recovered from the mortalities ranged from 10(2) to 10(7) TCID50  mL(-1) . Fry were sampled at 3, 5 and 7.5 weeks post-challenge. Sampling after 3 weeks revealed a high prevalence of infection in the absence of clinical signs, and infectious virus was recovered from 80% and 43% of sampled fry infected with SAV1 and SAV5, respectively. After 5 weeks pancreas, heart and red skeletal muscle lesions were generally observed, whilst degeneration in white skeletal muscle was observed only in fish infected with SAV1. In situ hybridisation confirmed the presence of viral genome in infected pancreas, heart and muscle. After 7.5 weeks, infectious virus (both isolates) was recovered from 13.3% of the fish sampled, with a viral titre of 10(2) TCID50  mL(-1) . Clearly, salmon fry are susceptible to SAV infection and pancreas disease.


Assuntos
Infecções por Alphavirus/veterinária , Doenças dos Peixes/transmissão , Doenças dos Peixes/virologia , Pancreatopatias/veterinária , Salmo salar , Alphavirus/isolamento & purificação , Alphavirus/fisiologia , Infecções por Alphavirus/mortalidade , Infecções por Alphavirus/patologia , Infecções por Alphavirus/transmissão , Infecções por Alphavirus/virologia , Animais , Doenças dos Peixes/mortalidade , Doenças dos Peixes/patologia , Água Doce , Genoma Viral/genética , Pancreatopatias/mortalidade , Pancreatopatias/patologia , Pancreatopatias/virologia , Carga Viral
13.
Proc Natl Acad Sci U S A ; 111(45): 16053-8, 2014 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-25362048

RESUMO

Mosquito-borne alphaviruses are important causes of epidemic encephalomyelitis. Neuronal cell death during fatal alphavirus encephalomyelitis is immune-mediated; however, the types of cells involved and their regulation have not been determined. We show that the virus-induced inflammatory response was accompanied by production of the regulatory cytokine IL-10, and in the absence of IL-10, paralytic disease occurred earlier and mice died faster. To determine the reason for accelerated disease in the absence of IL-10, immune responses in the CNS of IL-10(-/-) and wild-type (WT) mice were compared. There were no differences in the amounts of brain inflammation or peak virus replication; however, IL-10(-/-) animals had accelerated and increased infiltration of CD4(+)IL-17A(+) and CD4(+)IL-17A(+)IFNγ(+) cells compared with WT animals. Th17 cells infiltrating the brain demonstrated a pathogenic phenotype with the expression of the transcription factor, Tbet, and the production of granzyme B, IL-22, and GM-CSF, with greater production of GM-CSF in IL-10(-/-) mice. Therefore, in fatal alphavirus encephalomyelitis, pathogenic Th17 cells enter the CNS at the onset of neurologic disease and, in the absence of IL-10, appear earlier, develop into Th1/Th17 cells more often, and have greater production of GM-CSF. This study demonstrates a role for pathogenic Th17 cells in fatal viral encephalitis.


Assuntos
Infecções por Alphavirus/imunologia , Encefalomielite/imunologia , Interleucina-10/imunologia , Sindbis virus/imunologia , Células Th17/imunologia , Infecções por Alphavirus/genética , Infecções por Alphavirus/patologia , Animais , Encéfalo/imunologia , Encéfalo/patologia , Encéfalo/virologia , Encefalomielite/genética , Encefalomielite/patologia , Encefalomielite/virologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Granzimas/genética , Granzimas/imunologia , Interferon gama/genética , Interferon gama/imunologia , Interleucina-10/genética , Interleucinas/genética , Interleucinas/imunologia , Camundongos , Camundongos Knockout , Células Th1/imunologia , Células Th1/patologia , Células Th17/patologia , Interleucina 22
14.
J Immunol ; 193(5): 2455-68, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25070856

RESUMO

Previous studies using mannose-binding lectin (MBL) and complement C4-deficient mice have suggested that the lectin pathway (LP) is not required for the development of inflammatory arthritis in the collagen Ab-induced arthritis (CAIA) model. MBL, ficolins and collectin-11 are key LP pattern recognition molecules that associate with three serine proteases-MASP-1, MASP-2, and MASP-3-and with two MBL-associated proteins designated sMAP and MBL-associated protein of 44kDA (MAp44). Recent studies have shown that MAp44, an alternatively spliced product of the MASP-1/3 gene, is a competitive inhibitor of the binding of the recognition molecules to all three MASPs. In these studies, we examined the effect of treatment of mice with adenovirus (Ad) programmed to express human MAp44 (AdhMAp44) on the development of CAIA. AdhMAp44 and Ad programming GFP (AdGFP) expression were injected i.p. in C57BL/6 wild type mice prior to the induction of CAIA. AdhMAp44 significantly reduced the clinical disease activity (CDA) score by 81% compared with mice injected with AdGFP. Similarly, histopathologic injury scores for inflammation, pannus, cartilage and bone damage, as well as C3 deposition in the cartilage and synovium, were significantly reduced by AdhMAp44 pretreatment. Mice treated with AdmMAp44, programming expression of mouse MAp44, also showed significantly decreased CDA score and histopathologic injury scores. In addition, administration of AdhMAp44 significantly diminished the severity of Ross River virus-induced arthritis, an LP-dependent model. Our study provides conclusive evidence that an intact complement LP is essential to initiate CAIA, and that MAp44 may be an appropriate treatment for inflammatory arthritis.


Assuntos
Adenoviridae , Artrite Experimental/imunologia , Complemento C3/imunologia , Lectina de Ligação a Manose da Via do Complemento/imunologia , Serina Proteases Associadas a Proteína de Ligação a Manose/imunologia , Infecções por Alphavirus/genética , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/patologia , Processamento Alternativo/genética , Processamento Alternativo/imunologia , Animais , Artrite Experimental/genética , Artrite Experimental/patologia , Complemento C3/genética , Lectina de Ligação a Manose da Via do Complemento/genética , Humanos , Serina Proteases Associadas a Proteína de Ligação a Manose/genética , Camundongos , Camundongos Knockout , Ross River virus/imunologia , Transdução Genética
15.
Proc Natl Acad Sci U S A ; 111(16): 6040-5, 2014 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-24733914

RESUMO

Arthritogenic alphaviruses including Ross River virus (RRV), Sindbis virus, and chikungunya virus cause worldwide outbreaks of musculoskeletal disease. The ability of alphaviruses to induce bone pathologies remains poorly defined. Here we show that primary human osteoblasts (hOBs) can be productively infected by RRV. RRV-infected hOBs produced high levels of inflammatory cytokine including IL-6. The RANKL/OPG ratio was disrupted in the synovial fluid of RRV patients, and this was accompanied by an increase in serum Tartrate-resistant acid phosphatase 5b (TRAP5b) levels. Infection of bone cells with RRV was validated using an established RRV murine model. In wild-type mice, infectious virus was detected in the femur, tibia, patella, and foot, together with reduced bone volume in the tibial epiphysis and vertebrae detected by microcomputed tomographic (µCT) analysis. The RANKL/OPG ratio was also disrupted in mice infected with RRV; both this effect and the bone loss were blocked by treatment with an IL-6 neutralizing antibody. Collectively, these findings provide previously unidentified evidence that alphavirus infection induces bone loss and that OBs are capable of producing proinflammatory mediators during alphavirus-induced arthralgia. The perturbed RANKL/OPG ratio in RRV-infected OBs may therefore contribute to bone loss in alphavirus infection.


Assuntos
Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Artrite/virologia , Reabsorção Óssea/patologia , Reabsorção Óssea/virologia , Osteoblastos/patologia , Ross River virus/fisiologia , Fosfatase Ácida/sangue , Adulto , Infecções por Alphavirus/sangue , Animais , Anticorpos Neutralizantes/farmacologia , Artrite/sangue , Artrite/patologia , Reabsorção Óssea/sangue , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/patologia , Osso e Ossos/virologia , Feminino , Lâmina de Crescimento/efeitos dos fármacos , Lâmina de Crescimento/patologia , Lâmina de Crescimento/virologia , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-6/biossíntese , Isoenzimas/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Testes de Neutralização , Osteoblastos/efeitos dos fármacos , Osteoblastos/virologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/patologia , Osteoclastos/virologia , Osteogênese/efeitos dos fármacos , Osteoprotegerina/metabolismo , Fenótipo , Ligante RANK/metabolismo , Ross River virus/efeitos dos fármacos , Líquido Sinovial/metabolismo , Fosfatase Ácida Resistente a Tartarato , Replicação Viral/efeitos dos fármacos , Microtomografia por Raio-X
16.
J Clin Virol ; 59(4): 270-3, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24556566

RESUMO

The re-emerging invalidating chikungunya disease has recently extended to temperate areas. Other alphaviruses can also present with febrile arthalgias. Dengue virus transmitted by the same species of mosquitoes may cocirculate, leading to dual infections and concurrent epidemics. Although these diseases share similar clinical features, their prognoses considerably differ. Prominent and prolonged articular disorders are more consistent with chikungunya virus, whereas haemorrhages make the gravity of dengue infection. Specific symptoms are required, especially when diagnostic tests are not available or performable at a large scale. Indeed, early clinical suspicion of a vector-borne disease is crucial to isolate the first cases in the course of an outbreak, and discrimination between arboviruses help to optimal management of patients. No specific chikungunya clinical sign has been yet reported. We highlight here the high prevalence (about 25%) of acute ear redness in infected people during the 2008 chikungunya outbreak in Jahor Bahru in Malaysia. Nine consenting patients are more precisely described. Ear chondritis could be sensitive diagnostic criterion of the acute stage of chikungunya, every physician - even in occidental non endemic areas - should be aware of.


Assuntos
Infecções por Alphavirus/complicações , Infecções por Alphavirus/epidemiologia , Surtos de Doenças , Orelha Externa/patologia , Otite Externa/epidemiologia , Otite Externa/etiologia , Adolescente , Adulto , Idoso , Infecções por Alphavirus/diagnóstico , Infecções por Alphavirus/patologia , Doenças das Cartilagens/diagnóstico , Doenças das Cartilagens/patologia , Febre de Chikungunya , Criança , Pré-Escolar , Feminino , Humanos , Malásia/epidemiologia , Masculino , Pessoa de Meia-Idade , Otite Externa/diagnóstico , Otite Externa/patologia , Adulto Jovem
17.
Asian Pac J Trop Med ; 6(8): 631-4, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23790334

RESUMO

OBJECTIVE: To investigate cytokine profile in patients with chikungunya virus (CHIKV) infection. METHODS: Twenty eight pairs of serum samples collected from CHIKV infected patients during the outbreak of chikungunya fever in South Thailand in 2008 were obtained. A multiple cytokine assay for detection of 17 cytokines was performed. RESULTS: In the acute stage of CHIKV infection, the patients had significantly higher levels of interleukin-6, granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, monocyte chemotactic protein 1 and tumor necrosis factor alpha than the control (P<0.001, P=0.023, P=0.015, P<0.001 and P=0.024, respectively). When the disease developed to the recovery stage, the patients had significantly lower levels of interleukin-6, granulocyte-macrophage colony-stimulating factor, monocyte chemotactic protein 1 and macrophage inflammatory protein beta than in the acute stage (P<0.001). CONCLUSIONS: This study provides additional information that these cytokines could play roles in pathogenesis of CHIKV infection and could be used as disease biomarkers or drug targets.


Assuntos
Infecções por Alphavirus/imunologia , Infecções por Alphavirus/patologia , Vírus Chikungunya/imunologia , Citocinas/sangue , Infecções por Alphavirus/epidemiologia , Febre de Chikungunya , Surtos de Doenças , Humanos , Soro/química , Tailândia/epidemiologia
18.
J Virol ; 87(15): 8511-23, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23720714

RESUMO

Alphavirus dogma has long dictated the production of a discrete set of structural proteins during infection of a cell: capsid, pE2, 6K, and E1. However, bioinformatic analyses of alphavirus genomes (A. E. Firth, B. Y. Chung, M. N. Fleeton, and J. F. Atkins, Virol. J. 5:108, 2008) suggested that a ribosomal frameshifting event occurs during translation of the alphavirus structural polyprotein. Specifically, a frameshift event is suggested to occur during translation of the 6K gene, yielding production of a novel protein, termed transframe (TF), comprised of a C-terminal extension of the 6K protein in the -1 open reading frame (ORF). Here, we validate the findings of Firth and colleagues with respect to the production of the TF protein and begin to characterize the function of TF. Using a mass spectrometry-based approach, we identified TF in purified preparations of both Sindbis and Chikungunya virus particles. We next constructed a panel of Sindbis virus mutants with mutations which alter the production, size, or sequence of TF. We demonstrate that TF is not absolutely required in culture, although disrupting TF production leads to a decrease in virus particle release in both mammalian and insect cells. In a mouse neuropathogenesis model, mortality was <15% in animals infected with the TF mutants, whereas mortality was 95% in animals infected with the wild-type virus. Using a variety of additional assays, we demonstrate that TF retains ion-channel activity analogous to that of 6K and that lack of production of TF does not affect genome replication, particle infectivity, or envelope protein transit to the cell surface. The TF protein therefore represents a previously uncharacterized factor important for alphavirus assembly.


Assuntos
Vírus Chikungunya/fisiologia , Regulação Viral da Expressão Gênica , Sindbis virus/fisiologia , Proteínas Virais/biossíntese , Montagem de Vírus , Infecções por Alphavirus/mortalidade , Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Animais , Linhagem Celular , Vírus Chikungunya/genética , Vírus Chikungunya/patogenicidade , Modelos Animais de Doenças , Insetos , Camundongos , Sindbis virus/genética , Análise de Sobrevida , Replicação Viral
19.
J Virol ; 87(10): 5970-84, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23514884

RESUMO

Arthritogenic alphaviruses are human pathogens maintained in nature through alternating replication in vertebrates and mosquitoes. Using chimeric viruses, we previously reported that replacement of the PE2 coding region of the T48 strain of Ross River virus (RRV-T48) with that from the attenuated DC5692 strain, which differ by 7 amino acids, resulted in an attenuated disease phenotype in a mouse model of RRV-induced rheumatic disease. Here, we demonstrate that introduction of one of these amino acid differences, a tyrosine (Y)-to-histidine (H) change at position 18 of the E2 glycoprotein (E2 Y18H), into the RRV-T48 genetic background was sufficient to generate a virus that caused dramatically less severe musculoskeletal disease in mice. The attenuated phenotype of RRV-T48 E2 Y18H was associated with reduced viral loads in musculoskeletal tissues, reduced viremia, and less efficient virus spread. Consistent with these findings, RRV-T48 E2 Y18H replicated less well in mammalian cells in vitro due to significantly reduced PFU released per infected cell. In contrast, RRV-T48 E2 Y18H replicated more efficiently than RRV-T48 in C6/36 mosquito cells. Competition studies confirmed that RRV-T48 E2 Y18H had a fitness advantage in mosquito cells and a fitness disadvantage in mammalian cells. Interestingly, all sequenced Ross River viruses encode either a tyrosine or a histidine at E2 position 18, and this holds true for other alphaviruses in the Semliki Forest antigenic complex. Taken together, these findings suggest that a tyrosine-to-histidine switch at E2 position 18 functions as a regulator of RRV fitness in vertebrate and invertebrate cells.


Assuntos
Substituição de Aminoácidos , Proteínas do Capsídeo/metabolismo , Histidina/genética , Ross River virus/patogenicidade , Tirosina/genética , Proteínas do Envelope Viral/metabolismo , Fatores de Virulência/metabolismo , Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Animais , Proteínas do Capsídeo/genética , Linhagem Celular , Culicidae , Modelos Animais de Doenças , Histidina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/patologia , Músculo Esquelético/virologia , Ross River virus/genética , Ross River virus/fisiologia , Tirosina/metabolismo , Proteínas do Envelope Viral/genética , Carga Viral , Viremia , Virulência , Fatores de Virulência/genética , Replicação Viral
20.
Viral Immunol ; 26(1): 75-83, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23409931

RESUMO

Chikungunya virus (CHIKV) is an important emerging mosquito-borne alphavirus, indigenous to tropical Africa and Asia. It can cause epidemic fever and acute illness characterized by fever and arthralgias. The epidemic cycle of this infection is similar to dengue and urban yellow fever viral infections. The generation of an efficient vaccine against CHIKV is necessary to prevent and/or control the disease manifestations of the infection. In this report, we studied immune response against a CHIKV-envelope DNA vaccine (pEnv) and the role of the CHIKV nonstructural gene 2 (nsP2) as an adjuvant for the induction of protective immune responses in a relevant mouse challenge model. When injected with the CHIKV pEnv alone, 70% of the immunized mice survived CHIKV challenge, whereas when co-injected with pEnv+pnsP2, 90% of the mice survived viral challenge. Mice also exhibited a delayed onset signs of illness, and a marked decrease in morbidity, suggesting a nsP2 mediated adjuvant effect. Co-injection of the pnsP2 adjuvant with pEnv also qualitatively and quantitatively increased antigen specific neutralizing antibody responses compared to vaccination with pEnv alone. In sum, these novel data imply that the addition of nsP2 to the pEnv vaccine enhances anti-CHIKV-Env immune responses and maybe useful to include in future CHIKV clinical vaccination strategies.


Assuntos
Adjuvantes Imunológicos/metabolismo , Infecções por Alphavirus/prevenção & controle , Vírus Chikungunya/imunologia , Vacinas de DNA/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas não Estruturais Virais/metabolismo , Adjuvantes Imunológicos/genética , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/patologia , Animais , Febre de Chikungunya , Vírus Chikungunya/genética , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Índice de Gravidade de Doença , Análise de Sobrevida , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Proteínas do Envelope Viral/genética , Proteínas não Estruturais Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA