Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 18(2): e1010185, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35143591

RESUMO

Arthritogenic alphaviruses are mosquito-borne viruses that are a major cause of infectious arthropathies worldwide, and recent outbreaks of chikungunya virus and Ross River virus (RRV) infections highlight the need for robust intervention strategies. Alphaviral arthritis can persist for months after the initial acute disease, and is mediated by cellular immune responses. A common strategy to limit inflammation and pathology is to dampen the overwhelming inflammatory responses by modulating proinflammatory cytokine pathways. Here, we investigate the contribution of interleukin-17 (IL-17), a cytokine involved in arthropathies such as rheumatoid arthritis, in the development RRV-induced arthritis and myositis. IL-17 was quantified in serum from RRV-infected patients, and mice were infected with RRV and joints and muscle tissues collected to analyse cellular infiltrates, tissue mRNA, cytokine expression, and joint and muscle histopathology. IL-17 expression was increased in musculoskeletal tissues and serum of RRV-infected mice and humans, respectively. IL-17-producing T cells and neutrophils contributed to the cellular infiltrate in the joint and muscle tissue during acute RRV disease in mice. Blockade of IL-17A/F using a monoclonal antibody (mAb) reduced disease severity in RRV-infected mice and led to decreased proinflammatory proteins, cellular infiltration in synovial tissues and cartilage damage, without affecting viral titers in inflamed tissues. IL-17A/F blockade triggered a shift in transcriptional profile of both leukocyte infiltrates and musculoskeletal stromal cells by downregulating proinflammatory genes. This study highlights a previously uncharacterized role for an effector cytokine in alphaviral pathology and points towards potential therapeutic benefit in targeting IL-17 to treat patients presenting with RRV-induced arthropathy.


Assuntos
Artrite Reumatoide/imunologia , Imunidade Celular , Inflamação/imunologia , Interleucina-17/imunologia , Miosite/imunologia , Ross River virus/imunologia , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Animais , Artrite Reumatoide/virologia , Chlorocebus aethiops , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miosite/virologia , Células Vero , Carga Viral
2.
J Virol ; 96(17): e0099922, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-36000846

RESUMO

Arthritogenic alphaviruses are mosquito-borne arboviruses that include several re-emerging human pathogens, including the chikungunya (CHIKV), Ross River (RRV), Mayaro (MAYV), and o'nyong-nyong (ONNV) virus. Arboviruses are transmitted via a mosquito bite to the skin. Herein, we describe intradermal RRV infection in a mouse model that replicates the arthritis and myositis seen in humans with Ross River virus disease (RRVD). We show that skin infection with RRV results in the recruitment of inflammatory monocytes and neutrophils, which together with dendritic cells migrate to draining lymph nodes (LN) of the skin. Neutrophils and monocytes are productively infected and traffic virus from the skin to LN. We show that viral envelope N-linked glycosylation is a key determinant of skin immune responses and disease severity. RRV grown in mammalian cells elicited robust early antiviral responses in the skin, while RRV grown in mosquito cells stimulated poorer early antiviral responses. We used glycan mass spectrometry to characterize the glycan profile of mosquito and mammalian cell-derived RRV, showing deglycosylation of the RRV E2 glycoprotein is associated with curtailed skin immune responses and reduced disease following intradermal infection. Altogether, our findings demonstrate skin infection with an arthritogenic alphavirus leads to musculoskeletal disease and envelope glycoprotein glycosylation shapes disease outcome. IMPORTANCE Arthritogenic alphaviruses are transmitted via mosquito bites through the skin, potentially causing debilitating diseases. Our understanding of how viral infection starts in the skin and how virus systemically disseminates to cause disease remains limited. Intradermal arbovirus infection described herein results in musculoskeletal pathology, which is dependent on viral envelope N-linked glycosylation. As such, intradermal infection route provides new insights into how arboviruses cause disease and could be extended to future investigations of skin immune responses following infection with other re-emerging arboviruses.


Assuntos
Infecções por Alphavirus , Artrite , Miosite , Polissacarídeos , Ross River virus , Pele , Infecções por Alphavirus/complicações , Infecções por Alphavirus/imunologia , Animais , Antivirais/imunologia , Artrite/complicações , Artrite/imunologia , Culicidae/virologia , Células Dendríticas , Modelos Animais de Doenças , Glicosilação , Humanos , Espectrometria de Massas , Camundongos , Monócitos , Miosite/complicações , Miosite/imunologia , Neutrófilos , Polissacarídeos/química , Polissacarídeos/imunologia , Ross River virus/imunologia , Pele/imunologia , Pele/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia
3.
PLoS Pathog ; 16(8): e1008743, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32760128

RESUMO

Arthritogenic alphaviruses cause debilitating musculoskeletal disease and historically have circulated in distinct regions. With the global spread of chikungunya virus (CHIKV), there now is more geographic overlap, which could result in heterologous immunity affecting natural infection or vaccination. Here, we evaluated the capacity of a cross-reactive anti-CHIKV monoclonal antibody (CHK-265) to protect against disease caused by the distantly related alphavirus, Ross River virus (RRV). Although CHK-265 only moderately neutralizes RRV infection in cell culture, it limited clinical disease in mice independently of Fc effector function activity. Despite this protective phenotype, RRV escaped from CHK-265 neutralization in vivo, with resistant variants retaining pathogenic potential. Near the inoculation site, CHK-265 reduced viral burden in a type I interferon signaling-dependent manner and limited immune cell infiltration into musculoskeletal tissue. In a parallel set of experiments, purified human CHIKV immune IgG also weakly neutralized RRV, yet when transferred to mice, resulted in improved clinical outcome during RRV infection despite the emergence of resistant viruses. Overall, this study suggests that weakly cross-neutralizing antibodies can protect against heterologous alphavirus disease, even if neutralization escape occurs, through an early viral control program that tempers inflammation.


Assuntos
Infecções por Alphavirus/complicações , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Reações Cruzadas/imunologia , Doenças Musculoesqueléticas/prevenção & controle , Ross River virus/isolamento & purificação , Carga Viral/imunologia , Infecções por Alphavirus/virologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Musculoesqueléticas/imunologia , Doenças Musculoesqueléticas/virologia , Receptores Fc/fisiologia , Ross River virus/imunologia , Virulência
4.
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
5.
J Virol ; 94(8)2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-31996431

RESUMO

Ross River virus (RRV) belongs to the genus Alphavirus and is prevalent in Australia. RRV infection can cause arthritic symptoms in patients and may include rash, fever, arthralgia, and myalgia. Type I interferons (IFN) are the primary antiviral cytokines and trigger activation of the host innate immune system to suppress the replication of invading viruses. Alphaviruses are able to subvert the type I IFN system, but the mechanisms used are ill defined. In this study, seven RRV field strains were analyzed for induction of and sensitivity to type I IFN. The sensitivities of these strains to human IFN-ß varied significantly and were highest for the RRV 2548 strain. Compared to prototype laboratory strain RRV-T48, RRV 2548 also induced higher type I IFN levels both in vitro and in vivo and caused milder disease. To identify the determinants involved in type I IFN modulation, the region encoding the nonstructural proteins (nsPs) of RRV 2548 was sequenced, and 42 amino acid differences from RRV-T48 were identified. Using fragment swapping and site-directed mutagenesis, we discovered that substitutions E402A and R522Q in nsP1 as well as Q619R in nsP2 were responsible for increased sensitivity of RRV 2548 to type I IFN. In contrast, substitutions A31T, N219T, S580L, and Q619R in nsP2 led to induction of higher levels of type I IFN. With exception of E402A, all these variations are common for naturally occurring RRV strains. However, they are different from all known determinants of type I IFN modulation reported previously in nsPs of alphaviruses.IMPORTANCE By identifying natural Ross River virus (RRV) amino acid determinants for type I interferon (IFN) modulation, this study gives further insight into the mechanism of type I IFN modulation by alphaviruses. Here, the crucial role of type I IFN in the early stages of RRV disease pathogenesis is further demonstrated. This study also provides a comparison of the roles of different parts of the RRV nonstructural region in type I IFN modulation, highlighting the importance of nonstructural protein 1 (nsP1) and nsP2 in this process. Three substitutions in nsP1 and nsP2 were found to be independently associated with enhanced type I IFN sensitivity, and four independent substitutions in nsP2 were important in elevated type I IFN induction. Such evidence has clear implications for RRV immunobiology, persistence, and pathology. The identification of viral proteins that modulate type I IFN may also have importance for the pathogenesis of other alphaviruses.


Assuntos
Antivirais/farmacologia , Interferon Tipo I/imunologia , Interferon Tipo I/farmacologia , Ross River virus/efeitos dos fármacos , Ross River virus/imunologia , Alphavirus/genética , Alphavirus/imunologia , Infecções por Alphavirus/virologia , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Citocinas , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Sítio-Dirigida , Ross River virus/genética , Células Vero , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia , Virulência , Replicação Viral/efeitos dos fármacos
6.
Emerg Infect Dis ; 25(4): 827-830, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30882332

RESUMO

A unique outbreak of Ross River virus (RRV) infection was reported in Fiji in 1979. In 2013, RRV seroprevalence among residents was 46.5% (362/778). Of the residents who were seronegative in 2013 and retested in 2015, 10.9% (21/192) had seroconverted to RRV, suggesting ongoing endemic circulation of RRV in Fiji.


Assuntos
Infecções por Alphavirus/diagnóstico , Ross River virus/imunologia , Infecções por Alphavirus/sangue , Infecções por Alphavirus/epidemiologia , Anticorpos Antivirais/sangue , Fiji/epidemiologia , Humanos , Ross River virus/isolamento & purificação , Estudos Soroepidemiológicos
7.
Emerg Infect Dis ; 25(12): 2243-2252, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31742522

RESUMO

Australia experienced its largest recorded outbreak of Ross River virus (RRV) during the 2014-15 reporting year, comprising >10,000 reported cases. We investigated epidemiologic, entomologic, and virologic factors that potentially contributed to the scale of the outbreak in Queensland, the state with the highest number of notifications (6,371). Spatial analysis of human cases showed that notifications were geographically widespread. In Brisbane, human case notifications and virus detections in mosquitoes occurred across inland and coastal locations. Viral sequence data demonstrated 2 RRV lineages (northeastern genotypes I and II) were circulating, and a new strain containing 3 unique amino acid changes in the envelope 2 protein was identified. Longitudinal mosquito collections demonstrated unusually high relative abundance of Culex annulirostris and Aedes procax mosquitoes, attributable to extensive freshwater larval habitats caused by early and persistent rainfall during the reporting year. Increased prevalence of these mosquitoes probably contributed to the scale of this outbreak.


Assuntos
Infecções por Alphavirus/epidemiologia , Infecções por Alphavirus/virologia , Ross River virus , Infecções por Alphavirus/história , Infecções por Alphavirus/transmissão , Surtos de Doenças , Genes Virais , Geografia Médica , História do Século XXI , Humanos , Mosquitos Vetores/virologia , Filogenia , Vigilância em Saúde Pública , Queensland/epidemiologia , Ross River virus/classificação , Ross River virus/genética , Ross River virus/imunologia
8.
PLoS Pathog ; 13(12): e1006788, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29281739

RESUMO

Chikungunya virus (CHIKV) belongs to a group of mosquito-borne alphaviruses associated with acute and chronic arthropathy, with peripheral and limb joints most commonly affected. Using a mouse model of CHIKV infection and arthritic disease, we show that CHIKV replication and the ensuing foot arthropathy were dramatically reduced when mice were housed at 30°C, rather than the conventional 22°C. The effect was not associated with a detectable fever, but was dependent on type I interferon responses. Bioinformatics analyses of RNA-Seq data after injection of poly(I:C)/jetPEI suggested the unfolded protein response and certain type I interferon responses are promoted when feet are slightly warmer. The ambient temperature thus appears able profoundly to effect anti-viral activity in the periphery, with clear consequences for alphaviral replication and the ensuing arthropathy. These observations may provide an explanation for why alphaviral arthropathies are largely restricted to joints of the limbs and the extremities.


Assuntos
Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Artrite Experimental/imunologia , Artrite Experimental/virologia , Artrite Infecciosa/imunologia , Artrite Infecciosa/virologia , Interferon Tipo I/metabolismo , Infecções por Alphavirus/patologia , Animais , Artrite Experimental/patologia , Artrite Infecciosa/patologia , Febre de Chikungunya/imunologia , Febre de Chikungunya/patologia , Febre de Chikungunya/virologia , Vírus Chikungunya/imunologia , Vírus Chikungunya/patogenicidade , Vírus Chikungunya/fisiologia , Feminino , , Interações Hospedeiro-Patógeno/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ross River virus/imunologia , Ross River virus/patogenicidade , Ross River virus/fisiologia , Temperatura , Carga Viral , Replicação Viral/imunologia , Replicação Viral/fisiologia
9.
PLoS Pathog ; 13(12): e1006748, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29244871

RESUMO

Chikungunya virus (CHIKV) and Ross River virus (RRV) are mosquito-transmitted alphaviruses that cause debilitating acute and chronic musculoskeletal disease. Monocytes are implicated in the pathogenesis of these infections; however, their specific roles are not well defined. To investigate the role of inflammatory Ly6ChiCCR2+ monocytes in alphavirus pathogenesis, we used CCR2-DTR transgenic mice, enabling depletion of these cells by administration of diptheria toxin (DT). DT-treated CCR2-DTR mice displayed more severe disease following CHIKV and RRV infection and had fewer Ly6Chi monocytes and NK cells in circulation and muscle tissue compared with DT-treated WT mice. Furthermore, depletion of CCR2+ or Gr1+ cells, but not NK cells or neutrophils alone, restored virulence and increased viral loads in mice infected with an RRV strain encoding attenuating mutations in nsP1 to levels detected in monocyte-depleted mice infected with fully virulent RRV. Disease severity and viral loads also were increased in DT-treated CCR2-DTR+;Rag1-/- mice infected with the nsP1 mutant virus, confirming that these effects are independent of adaptive immunity. Monocytes and macrophages sorted from muscle tissue of RRV-infected mice were viral RNA positive and had elevated expression of Irf7, and co-culture of Ly6Chi monocytes with RRV-infected cells resulted in induction of type I IFN gene expression in monocytes that was Irf3;Irf7 and Mavs-dependent. Consistent with these data, viral loads of the attenuated nsP1 mutant virus were equivalent to those of WT RRV in Mavs-/- mice. Finally, reconstitution of Irf3-/-;Irf7-/- mice with CCR2-DTR bone marrow rescued mice from severe infection, and this effect was reversed by depletion of CCR2+ cells, indicating that CCR2+ hematopoietic cells are capable of inducing an antiviral response. Collectively, these data suggest that MAVS-dependent production of type I IFN by monocytes is critical for control of acute alphavirus infection and that determinants in nsP1, the viral RNA capping protein, counteract this response.


Assuntos
Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Monócitos/imunologia , Monócitos/virologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Antígenos Ly/metabolismo , Vírus Chikungunya/imunologia , Vírus Chikungunya/patogenicidade , Toxina Diftérica/farmacologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/imunologia , Humanos , Inflamação/virologia , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 7 de Interferon/deficiência , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/imunologia , Interferon Tipo I/biossíntese , Interferon Tipo I/genética , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Monócitos/efeitos dos fármacos , Receptores CCR2/genética , Receptores CCR2/metabolismo , Ross River virus/genética , Ross River virus/imunologia , Ross River virus/patogenicidade , Carga Viral , Virulência/genética , Virulência/imunologia
11.
J Virol ; 90(8): 4150-4159, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26865723

RESUMO

UNLABELLED: The alphaviral6kgene region encodes the two structural proteins 6K protein and, due to a ribosomal frameshift event, the transframe protein (TF). Here, we characterized the role of the6kproteins in the arthritogenic alphavirus Ross River virus (RRV) in infected cells and in mice, using a novel6kin-frame deletion mutant. Comprehensive microscopic analysis revealed that the6kproteins were predominantly localized at the endoplasmic reticulum of RRV-infected cells. RRV virions that lack the6kproteins 6K and TF [RRV-(Δ6K)] were more vulnerable to changes in pH, and the corresponding virus had increased sensitivity to a higher temperature. While the6kdeletion did not reduce RRV particle production in BHK-21 cells, it affected virion release from the host cell. Subsequentin vivostudies demonstrated that RRV-(Δ6K) caused a milder disease than wild-type virus, with viral titers being reduced in infected mice. Immunization of mice with RRV-(Δ6K) resulted in a reduced viral load and accelerated viral elimination upon secondary infection with wild-type RRV or another alphavirus, chikungunya virus (CHIKV). Our results show that the6kproteins may contribute to alphaviral disease manifestations and suggest that manipulation of the6kgene may be a potential strategy to facilitate viral vaccine development. IMPORTANCE: Arthritogenic alphaviruses, such as chikungunya virus (CHIKV) and Ross River virus (RRV), cause epidemics of debilitating rheumatic disease in areas where they are endemic and can emerge in new regions worldwide. RRV is of considerable medical significance in Australia, where it is the leading cause of arboviral disease. The mechanisms by which alphaviruses persist and cause disease in the host are ill defined. This paper describes the phenotypic properties of an RRV6kdeletion mutant. The absence of the6kgene reduced virion release from infected cells and also reduced the severity of disease and viral titers in infected mice. Immunization with the mutant virus protected mice against viremia not only upon exposure to RRV but also upon challenge with CHIKV. These findings could lead to the development of safer and more immunogenic alphavirus vectors for vaccine delivery.


Assuntos
Infecções por Alphavirus/virologia , Ross River virus/genética , Ross River virus/imunologia , Proteínas Estruturais Virais/genética , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/fisiopatologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Vírus Chikungunya/imunologia , Chlorocebus aethiops , Cricetinae , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Mutação , Fases de Leitura , Ross River virus/patogenicidade , Deleção de Sequência , Células Vero , Carga Viral , Proteínas Estruturais Virais/análise , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/imunologia , Replicação Viral
12.
PLoS Pathog ; 11(10): e1005191, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26436766

RESUMO

Arthritogenic alphaviruses, including Ross River virus (RRV) and chikungunya virus (CHIKV), are responsible for explosive epidemics involving millions of cases. These mosquito-transmitted viruses cause inflammation and injury in skeletal muscle and joint tissues that results in debilitating pain. We previously showed that arginase 1 (Arg1) was highly expressed in myeloid cells in the infected and inflamed musculoskeletal tissues of RRV- and CHIKV-infected mice, and specific deletion of Arg1 from myeloid cells resulted in enhanced viral control. Here, we show that Arg1, along with other genes associated with suppressive myeloid cells, is induced in PBMCs isolated from CHIKV-infected patients during the acute phase as well as the chronic phase, and that high Arg1 expression levels were associated with high viral loads and disease severity. Depletion of both CD4 and CD8 T cells from RRV-infected Arg1-deficient mice restored viral loads to levels detected in T cell-depleted wild-type mice. Moreover, Arg1-expressing myeloid cells inhibited virus-specific T cells in the inflamed and infected musculoskeletal tissues, but not lymphoid tissues, following RRV infection in mice, including suppression of interferon-γ and CD69 expression. Collectively, these data enhance our understanding of the immune response following arthritogenic alphavirus infection and suggest that immunosuppressive myeloid cells may contribute to the duration or severity of these debilitating infections.


Assuntos
Infecções por Alphavirus/imunologia , Arginase/imunologia , Células Mieloides/imunologia , Linfócitos T/imunologia , Carga Viral/imunologia , Transferência Adotiva , Animais , Western Blotting , Febre de Chikungunya/imunologia , Vírus Chikungunya , Citometria de Fluxo , Humanos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Ross River virus/imunologia
13.
J Immunol ; 194(2): 678-89, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25488988

RESUMO

Ross River virus (RRV), chikungunya virus, and related alphaviruses cause debilitating polyarthralgia and myalgia. Mouse models of RRV and chikungunya virus have demonstrated a role for the adaptive immune response in the control of these infections. However, questions remain regarding the role for T cells in viral control, including the magnitude, location, and dynamics of CD8(+) T cell responses. To address these questions, we generated a recombinant RRV expressing the H-2(b)-restricted glycoprotein 33 (gp33) determinant derived from the glycoprotein of lymphocytic choriomeningitis virus. Using tetramers, we tracked gp33-specific CD8(+) T cells during RRV-lymphocytic choriomeningitis virus infection. We found that acute RRV infection induces activation of CD8(+) T cell responses in lymphoid and musculoskeletal tissues that peak from 10-14 d postinoculation, suggesting that CD8(+) T cells contribute to control of acute RRV infection. Mice genetically deficient for CD8(+) T cells or wild-type mice depleted of CD8(+) T cells had elevated RRV loads in skeletal muscle tissue, but not joint-associated tissues, at 14 d postinoculation, suggesting that the ability of CD8(+) T cells to control RRV infection is tissue dependent. Finally, adoptively transferred T cells were capable of reducing RRV loads in skeletal muscle tissue of Rag1(-/-) mice, indicating that T cells can contribute to the control of RRV infection in the absence of B cells and Ab. Collectively, these data demonstrate a role for T cells in the control of RRV infection and suggest that the antiviral capacity of T cells is controlled in a tissue-specific manner.


Assuntos
Infecções por Alphavirus/imunologia , Linfócitos T CD8-Positivos/imunologia , Imunidade Celular , Músculo Esquelético/imunologia , Ross River virus/imunologia , Infecções por Alphavirus/genética , Infecções por Alphavirus/patologia , Animais , Linfócitos T CD8-Positivos/patologia , Antígeno de Histocompatibilidade H-2D/genética , Antígeno de Histocompatibilidade H-2D/imunologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Camundongos , Camundongos Knockout , Músculo Esquelético/patologia , Músculo Esquelético/virologia , Fatores de Tempo , Proteínas Virais/genética , Proteínas Virais/metabolismo
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.
J Virol ; 88(7): 3719-32, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24429363

RESUMO

UNLABELLED: Ross River virus (RRV) is one of a group of mosquito-transmitted alphaviruses that cause debilitating, and often chronic, musculoskeletal disease in humans. Previously, we reported that replacement of the nonstructural protein 1 (nsP1) gene of the mouse-virulent RRV strain T48 with that from the mouse-avirulent strain DC5692 generated a virus that was attenuated in a mouse model of disease. Here we find that the six nsP1 nonsynonymous nucleotide differences between strains T48 and DC5692 are determinants of RRV virulence, and we identify two nonsynonymous nucleotide changes as sufficient for the attenuated phenotype. RRV T48 carrying the six nonsynonymous DC5692 nucleotide differences (RRV-T48-nsP1(6M)) was attenuated in both wild-type and Rag1(-/-) mice. Despite the attenuated phenotype, RRV T48 and RRV-T48-nsP1(6M) loads in tissues of wild-type and Rag1(-/-) mice were indistinguishable from 1 to 3 days postinoculation. RRV-T48-nsP1(6M) loads in skeletal muscle tissue, but not in other tissues, decreased dramatically by 5 days postinoculation in both wild-type and Rag1(-/-) mice, suggesting that the RRV-T48-nsP1(6M) mutant is more sensitive to innate antiviral effectors than RRV T48 in a tissue-specific manner. In vitro, we found that the attenuating mutations in nsP1 conferred enhanced sensitivity to type I interferon. In agreement with these findings, RRV T48 and RRV-T48-nsP1(6M) loads were similar in mice deficient in the type I interferon receptor. Our findings suggest that the type I IFN response controls RRV infection in a tissue-specific manner and that specific amino acid changes in nsP1 are determinants of RRV virulence by regulating the sensitivity of RRV to interferon. IMPORTANCE: Arthritogenic alphaviruses, including Ross River virus (RRV), infect humans and cause debilitating pain and inflammation of the musculoskeletal system. In this study, we identified coding changes in the RRV nsP1 gene that control the virulence of RRV and its sensitivity to the antiviral type I interferon response, a major component of antiviral defense in mammals. Furthermore, our studies revealed that the effects of these attenuating mutations are tissue specific. These findings suggest that these mutations in nsP1 influence the sensitivity of RRV to type I interferon only in specific host tissues. The new knowledge gained from these studies contributes to our understanding of host responses that control alphavirus infection and viral determinants that counteract these responses.


Assuntos
Infecções por Alphavirus/virologia , Interações Hospedeiro-Patógeno , Interferon Tipo I/imunologia , Mutação de Sentido Incorreto , Ross River virus/patogenicidade , Proteínas não Estruturais Virais/metabolismo , Fatores de Virulência/metabolismo , Infecções por Alphavirus/patologia , Estruturas Animais/virologia , Animais , Análise Mutacional de DNA , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ross River virus/imunologia , Carga Viral , Proteínas não Estruturais Virais/genética , Virulência , Fatores de Virulência/genética
16.
J Immunol ; 189(8): 4047-59, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22972923

RESUMO

Chikungunya virus (CHIKV) and Ross River virus (RRV) cause a debilitating, and often chronic, musculoskeletal inflammatory disease in humans. Macrophages constitute the major inflammatory infiltrates in musculoskeletal tissues during these infections. However, the precise macrophage effector functions that affect the pathogenesis of arthritogenic alphaviruses have not been defined. We hypothesized that the severe damage to musculoskeletal tissues observed in RRV- or CHIKV-infected mice would promote a wound-healing response characterized by M2-like macrophages. Indeed, we found that RRV- and CHIKV-induced musculoskeletal inflammatory lesions, and macrophages present in these lesions, have a unique gene-expression pattern characterized by high expression of arginase 1 and Ym1/Chi3l3 in the absence of FIZZ1/Relmα that is consistent with an M2-like activation phenotype. Strikingly, mice specifically deleted for arginase 1 in neutrophils and macrophages had dramatically reduced viral loads and improved pathology in musculoskeletal tissues at late times post-RRV infection. These findings indicate that arthritogenic alphavirus infection drives a unique myeloid cell activation program in inflamed musculoskeletal tissues that inhibits virus clearance and impedes disease resolution in an arginase 1-dependent manner.


Assuntos
Infecções por Alphavirus/imunologia , Arginase/genética , Vírus Chikungunya/imunologia , Deleção de Genes , Macrófagos/imunologia , Neutrófilos/imunologia , Ross River virus/imunologia , Regulação para Cima/imunologia , Infecções por Alphavirus/enzimologia , Infecções por Alphavirus/terapia , Animais , Arginase/antagonistas & inibidores , Linhagem Celular , Cricetinae , Humanos , Inflamação/enzimologia , Inflamação/imunologia , Inflamação/patologia , Macrófagos/enzimologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/enzimologia , Células Mieloides/imunologia , Células Mieloides/virologia , Neutrófilos/enzimologia , Neutrófilos/virologia , Carga Viral/imunologia
17.
Commun Dis Intell Q Rep ; 38(2): E114-22, 2014 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-25222206

RESUMO

An increase in off-season (June to September) Ross River virus (RRV) notifications from the greater Perth metropolitan area was observed from 2006 to 2009. We investigated the increase to determine whether it is likely to have reflected a true increase in off-season cases. A single positive RRV IgM test result is sufficient for RRV notification but where follow-up testing was performed, the positive predictive value of an IgM test where IgG was negative was very low in the off-season and also in the season when using the only commercially available test kit. The increase in off-season notifications was not associated with an increase in off-season testing. Some Perth laboratories use more stringent notification criteria than the nationally agreed RRV case definition, and the geographical distribution of samples tested varies between laboratories. Our findings make a strong case to change the nationally agreed case definition for RRV to not accept a single IgM positive test result as laboratory definitive evidence where the IgG is negative. Our study also identified a range of challenges in interpreting changes in seasonal patterns and geographical distribution of RRV. Any such observed changes should be investigated through further data analysis and/or mosquito trapping and testing in order to assess validity.


Assuntos
Infecções por Alphavirus/epidemiologia , Vigilância da População , Ross River virus , Estações do Ano , Infecções por Alphavirus/diagnóstico , Austrália/epidemiologia , Notificação de Doenças , Humanos , Imunoglobulina M/sangue , Imunoglobulina M/imunologia , Ross River virus/classificação , Ross River virus/imunologia
18.
PLoS Negl Trop Dis ; 16(2): e0009848, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35143495

RESUMO

Across the Pacific, and including in the Solomon Islands, outbreaks of arboviruses such as dengue, chikungunya, and Zika are increasing in frequency, scale and impact. Outbreaks of mosquito-borne disease have the potential to overwhelm the health systems of small island nations. This study mapped the seroprevalence of dengue, Zika, chikungunya and Ross River viruses in 5 study sites in the Solomon Islands. Serum samples from 1,021 participants were analysed by ELISA. Overall, 56% of participants were flavivirus-seropositive for dengue (28%), Zika (1%) or both flaviviruses (27%); and 53% of participants were alphavirus-seropositive for chikungunya (3%), Ross River virus (31%) or both alphaviruses (18%). Seroprevalence for both flaviviruses and alphaviruses varied by village and age of the participant. The most prevalent arboviruses in the Solomon Islands were dengue and Ross River virus. The high seroprevalence of dengue suggests that herd immunity may be a driver of dengue outbreak dynamics in the Solomon Islands. Despite being undetected prior to this survey, serology results suggest that Ross River virus transmission is endemic. There is a real need to increase the diagnostic capacities for each of the arboviruses to support effective case management and to provide timely information to inform vector control efforts and other outbreak mitigation interventions.


Assuntos
Infecções por Alphavirus/sangue , Febre de Chikungunya/sangue , Vírus Chikungunya/imunologia , Vírus da Dengue/imunologia , Dengue/sangue , Ross River virus/imunologia , Infecção por Zika virus/sangue , Zika virus/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Infecções por Alphavirus/epidemiologia , Infecções por Alphavirus/virologia , Anticorpos Antivirais/sangue , Febre de Chikungunya/epidemiologia , Febre de Chikungunya/virologia , Vírus Chikungunya/genética , Vírus Chikungunya/isolamento & purificação , Criança , Pré-Escolar , Dengue/epidemiologia , Dengue/virologia , Vírus da Dengue/genética , Vírus da Dengue/isolamento & purificação , Feminino , Humanos , Masculino , Melanesia/epidemiologia , Pessoa de Meia-Idade , Ross River virus/genética , Ross River virus/isolamento & purificação , Estudos Soroepidemiológicos , Adulto Jovem , Zika virus/genética , Zika virus/isolamento & purificação , Infecção por Zika virus/epidemiologia , Infecção por Zika virus/virologia
19.
J Virol ; 84(16): 8021-32, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20519386

RESUMO

Chikungunya virus is a mosquito-borne arthrogenic alphavirus that has recently reemerged to produce the largest epidemic ever documented for this virus. Here we describe a new adult wild-type mouse model of chikungunya virus arthritis, which recapitulates the self-limiting arthritis, tenosynovitis, and myositis seen in humans. Rheumatic disease was associated with a prolific infiltrate of monocytes, macrophages, and NK cells and the production of monocyte chemoattractant protein 1 (MCP-1), tumor necrosis factor alpha (TNF-alpha), and gamma interferon (IFN-gamma). Infection with a virus isolate from the recent Reunion Island epidemic induced significantly more mononuclear infiltrates, proinflammatory mediators, and foot swelling than did an Asian isolate from the 1960s. Primary mouse macrophages were shown to be productively infected with chikungunya virus; however, the depletion of macrophages ameliorated rheumatic disease and prolonged the viremia. Only 1 microg of an unadjuvanted, inactivated, whole-virus vaccine derived from the Asian isolate completely protected against viremia and arthritis induced by the Reunion Island isolate, illustrating that protection is not strain specific and that low levels of immunity are sufficient to mediate protection. IFN-alpha treatment was able to prevent arthritis only if given before infection, suggesting that IFN-alpha is not a viable therapy. Prior infection with Ross River virus, a related arthrogenic alphavirus, and anti-Ross River virus antibodies protected mice against chikungunya virus disease, suggesting that individuals previously exposed to Ross River virus should be protected from chikungunya virus disease. This new mouse model of chikungunya virus disease thus provides insights into pathogenesis and a simple and convenient system to test potential new interventions.


Assuntos
Infecções por Alphavirus/patologia , Infecções por Alphavirus/virologia , Artrite/patologia , Artrite/virologia , Vírus Chikungunya/isolamento & purificação , Vírus Chikungunya/patogenicidade , Modelos Animais de Doenças , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/prevenção & controle , Animais , Artrite/imunologia , Artrite/prevenção & controle , Vírus Chikungunya/imunologia , Citocinas/metabolismo , Feminino , Pé/patologia , Pé/virologia , Histocitoquímica , Humanos , Imuno-Histoquímica , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia , Monócitos/imunologia , Ross River virus/imunologia , Vacinas de Produtos Inativados/imunologia , Carga Viral , Vacinas Virais/imunologia , Viremia
20.
J Exp Med ; 187(11): 1799-811, 1998 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-9607921

RESUMO

The serine proteinase inhibitor (serpin) plasminogen activator inhibitor type 2 (PAI-2) is well characterized as an inhibitor of extracellular urokinase-type plasminogen activator. Here we show that intracellular, but not extracellular, PAI-2 protected cells from the rapid cytopathic effects of alphavirus infection. This protection did not appear to be related to an effect on apoptosis but was associated with a PAI-2-mediated induction of constitutive low-level interferon (IFN)-alpha/beta production and IFN-stimulated gene factor 3 (ISGF3) activation, which primed the cells for rapid induction of antiviral genes. This primed phenotype was associated with a rapid development of resistance to infection by the PAI-2 transfected cells and the establishment of a persistent productive infection. PAI-2 was also induced in macrophages in response to viral RNA suggesting that PAI-2 is a virus response gene. These observations, together with the recently demonstrated PAI-2-mediated inhibition of tumor necrosis factor-alpha induced apoptosis, (a) illustrate that PAI-2 has an additional and distinct function as an intracellular regulator of signal transduction pathway(s) and (b) demonstrate a novel activity for a eukaryotic serpin.


Assuntos
Alphavirus/imunologia , Interferon-alfa/genética , Interferon beta/genética , Inibidor 2 de Ativador de Plasminogênio/farmacologia , Inibidores de Serina Proteinase/farmacologia , Adenovírus Humanos/imunologia , Antivirais , Apoptose , Efeito Citopatogênico Viral/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Vírus da Influenza A/imunologia , Fator Gênico 3 Estimulado por Interferon , Fator Gênico 3 Estimulado por Interferon, Subunidade gama , Interferon-alfa/imunologia , Interferon beta/imunologia , Inibidor 2 de Ativador de Plasminogênio/genética , Poli I-C/imunologia , Reação em Cadeia da Polimerase , Proteínas/metabolismo , RNA Mensageiro , Ross River virus/imunologia , Inibidores de Serina Proteinase/genética , Sindbis virus/imunologia , Fatores de Transcrição/metabolismo , Proteínas Virais/metabolismo , Latência Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA