Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
Int J Mol Sci ; 25(9)2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38732169

RESUMO

Infections may affect the course of autoimmune inflammatory diseases of the central nervous system (CNS), such as multiple sclerosis (MS). Infections with lactate dehydrogenase-elevating virus (LDV) protected mice from developing experimental autoimmune encephalomyelitis (EAE), a mouse counterpart of MS. Uninfected C57BL/6 mice immunized with the myelin oligodendrocyte glycoprotein peptide (MOG35-55) experienced paralysis and lost weight at a greater rate than mice who had previously been infected with LDV. LDV infection decreased the presentation of the MOG peptide by CD11b+CD11c+ dendritic cells (DC) to pathogenic T lymphocytes. When comparing non-infected mice to infected mice, the histopathological examination of the CNS showed more areas of demyelination and CD45+ and CD3+, but not Iba1+ cell infiltration. These results suggest that the protective effect of LDV infection against EAE development is mediated by a suppression of myelin antigen presentation by a specific DC subset to autoreactive T lymphocytes. Such a mechanism might contribute to the general suppressive effect of infections on autoimmune diseases known as the hygiene hypothesis.


Assuntos
Células Dendríticas , Encefalomielite Autoimune Experimental , Vírus Elevador do Lactato Desidrogenase , Esclerose Múltipla , Glicoproteína Mielina-Oligodendrócito , Animais , Feminino , Camundongos , Apresentação de Antígeno/imunologia , Infecções por Cardiovirus/imunologia , Antígeno CD11b/metabolismo , Antígeno CD11b/imunologia , Antígeno CD11c/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/virologia , Vírus Elevador do Lactato Desidrogenase/imunologia , Camundongos Endogâmicos C57BL , Esclerose Múltipla/imunologia , Esclerose Múltipla/virologia , Esclerose Múltipla/patologia , Glicoproteína Mielina-Oligodendrócito/imunologia , Fragmentos de Peptídeos/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
2.
J Virol ; 97(10): e0093023, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37792000

RESUMO

IMPORTANCE: Mouse models of viral infection play an especially large role in virology. In 1960, a mouse virus, lactate dehydrogenase-elevating virus (LDV), was discovered and found to have the peculiar ability to evade clearance by the immune system, enabling it to persistently infect an individual mouse for its entire lifespan without causing overt disease. However, researchers were unable to grow LDV in culture, ultimately resulting in the demise of this system as a model of failed immunity. We solve this problem by identifying the cell-surface molecule CD163 as the critical missing component in cell-culture systems, enabling the growth of LDV in immortalized cell lines for the first time. This advance creates abundant opportunities for further characterizing LDV in order to study both failed immunity and the family of viruses to which LDV belongs, Arteriviridae (aka, arteriviruses).


Assuntos
Antígenos CD , Antígenos de Diferenciação Mielomonocítica , Técnicas de Cultura de Células , Expressão Ectópica do Gene , Vírus Elevador do Lactato Desidrogenase , Receptores de Superfície Celular , Animais , Camundongos , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/genética , Antígenos de Diferenciação Mielomonocítica/metabolismo , Linhagem Celular/virologia , Vírus Elevador do Lactato Desidrogenase/genética , Vírus Elevador do Lactato Desidrogenase/crescimento & desenvolvimento , Vírus Elevador do Lactato Desidrogenase/imunologia , Vírus Elevador do Lactato Desidrogenase/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Fatores de Tempo
3.
Int J Mol Sci ; 22(16)2021 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-34445732

RESUMO

Infection with viruses, such as the lactate dehydrogenase-elevating virus (LDV), is known to trigger the onset of autoimmune anemia through the enhancement of the phagocytosis of autoantibody-opsonized erythrocytes by activated macrophages. Type I interferon receptor-deficient mice show enhanced anemia, which suggests a protective effect of these cytokines, partly through the control of type II interferon production. The development of anemia requires the expression of Fcγ receptors (FcγR) I, III, and IV. Whereas LDV infection decreases FcγR III expression, it enhances FcγR I and IV expression in wild-type animals. The LDV-associated increase in the expression of FcγR I and IV is largely reduced in type I interferon receptor-deficient mice, through both type II interferon-dependent and -independent mechanisms. Thus, the regulation of the expression of FcγR I and IV, but not III, by interferons may partly explain the exacerbating effect of LDV infection on anemia that results from the enhanced phagocytosis of IgG autoantibody-opsonized erythrocytes.


Assuntos
Anemia Hemolítica Autoimune/imunologia , Infecções por Arterivirus/imunologia , Interferons/metabolismo , Vírus Elevador do Lactato Desidrogenase/imunologia , Receptores de IgG/metabolismo , Anemia Hemolítica Autoimune/virologia , Animais , Infecções por Arterivirus/virologia , Interações Hospedeiro-Patógeno , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose
4.
mBio ; 11(2)2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32265335

RESUMO

Coinfections shape immunity and influence the development of inflammatory diseases, resulting in detrimental or beneficial outcome. Coinfections with concurrent Plasmodium species can alter malaria clinical evolution, and malaria infection itself can modulate autoimmune reactions. Yet, the underlying mechanisms remain ill defined. Here, we demonstrate that the protective effects of some rodent malaria strains on T cell-mediated inflammatory pathologies are due to an RNA virus cohosted in malaria-parasitized blood. We show that live and extracts of blood parasitized by Plasmodium berghei K173 or Plasmodium yoelii 17X YM, protect against P. berghei ANKA-induced experimental cerebral malaria (ECM) and myelin oligodendrocyte glycoprotein (MOG)/complete Freund's adjuvant (CFA)-induced experimental autoimmune encephalomyelitis (EAE), and that protection is associated with a strong type I interferon (IFN-I) signature. We detected the presence of the RNA virus lactate dehydrogenase-elevating virus (LDV) in the protective Plasmodium stabilates and we established that LDV infection alone was necessary and sufficient to recapitulate the protective effects on ECM and EAE. In ECM, protection resulted from an IFN-I-mediated reduction in the abundance of splenic conventional dendritic cell and impairment of their ability to produce interleukin (IL)-12p70, leading to a decrease in pathogenic CD4+ Th1 responses. In EAE, LDV infection induced IFN-I-mediated abrogation of IL-23, thereby preventing the differentiation of granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing encephalitogenic CD4+ T cells. Our work identifies a virus cohosted in several Plasmodium stabilates across the community and deciphers its major consequences on the host immune system. More generally, our data emphasize the importance of considering contemporaneous infections for the understanding of malaria-associated and autoimmune diseases.IMPORTANCE Any infection modifies the host immune status, potentially ameliorating or aggravating the pathophysiology of a simultaneous inflammatory condition. In the course of investigating how malaria infection modulates the severity of contemporaneous inflammatory diseases, we identified a nonpathogenic mouse virus in stabilates of two widely used rodent parasite lines: Plasmodium berghei K173 and Plasmodium yoelii 17X YM. We established that the protective effects of these Plasmodium lines on cerebral malaria and multiple sclerosis are exclusively due to this virus. The virus induces a massive type I interferon (IFN-I) response and causes quantitative and qualitative defects in the ability of dendritic cells to promote pathogenic T cell responses. Beyond revealing a possible confounding factor in rodent malaria models, our work uncovers some bases by which a seemingly innocuous viral (co)infection profoundly changes the immunopathophysiology of inflammatory diseases.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Inflamação/imunologia , Interferon Tipo I/imunologia , Vírus Elevador do Lactato Desidrogenase/imunologia , Malária Cerebral/imunologia , Animais , Coinfecção/imunologia , Coinfecção/parasitologia , Coinfecção/virologia , Citocinas/imunologia , Células Dendríticas/imunologia , Inflamação/fisiopatologia , Interferon gama/imunologia , Malária Cerebral/sangue , Malária Cerebral/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei , Plasmodium yoelii , Baço/citologia , Baço/imunologia
5.
J Gen Virol ; 95(Pt 7): 1504-1509, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24739273

RESUMO

Although many cells undergo transformation, few actually develop into tumours, due to successful mechanisms of immunosurveillance. To investigate whether an infectious agent may play a role in this process, the growth of a plasmacytoma was investigated in mice infected by lactate dehydrogenase-elevating virus. Acutely infected animals were significantly protected against tumour development. The mechanisms responsible for this protection were analysed in mice deficient for relevant immune cells or molecules and after in vivo cell depletion. This protection by viral infection correlated with NK cell activation and with IFN-γ production. It might also be related to activation of NK/T-cells, although this remains to be proven formally. Therefore, our results indicated that infections with benign micro-organisms may protect the host against cancer development, through non-specific stimulation of the host's innate immune system and especially of NK cells.


Assuntos
Células Matadoras Naturais/imunologia , Vírus Elevador do Lactato Desidrogenase/imunologia , Vírus Oncolíticos/imunologia , Plasmocitoma/imunologia , Plasmocitoma/prevenção & controle , Animais , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout
6.
Int Immunopharmacol ; 15(3): 544-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23391715

RESUMO

Lactate dehydrogenase-elevating virus (LDV) is an apparently innocuous and persistent virus that can modify mouse immune reactions. We have shown that LDV-infected mice immunized with human growth hormone (hGH) showed a deep modification of the specificity of the anti-hGH antibodies (Ab) in CBA/Ht mice but not BALB/c animals. The aim of this work was to extend the previous observations to another mouse strain, C57BL/6, as well as to an antigen unrelated to hGH, ovalbumin (OVA), and to explore at the same time the production of various cytokines at serum and cellular levels. The amount of Ab directed to hGH or OVA native antigenic determinants versus the concentration of Ab to cryptic epitopes was evaluated by ELISA competition experiments. Results indicated that LDV infection affected Ab specificity solely in CBA/Ht mice. In CBA/Ht the virus infection was associated with a reduction of the Ab titers to hGH native epitopes and with a decrease of IL-13 and IL-17 serum levels, but Ab to native OVA epitopes were increased with a simultaneous increase of IL-17. Accordingly, only lymph node cells from infected CBA/Ht mice immunized with OVA were found to produce INF-γ, IL-13 and IL-17. Thus, a correlation of cytokine production with a change in Ab specificity after a viral infection was found, although this phenomenon was restricted to a given antigen and to the genetic background of immunized animals. These observations suggest that an apparent harmless virus can affect some immunological mechanisms, which could lead, for example, to inflammatory or autoimmune disorders.


Assuntos
Anticorpos Antivirais/imunologia , Especificidade de Anticorpos , Infecções por Arterivirus/imunologia , Citocinas/imunologia , Epitopos Imunodominantes/imunologia , Vírus Elevador do Lactato Desidrogenase/imunologia , Animais , Anticorpos Antivirais/sangue , Hormônio do Crescimento/imunologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Ovalbumina/imunologia , Especificidade da Espécie
7.
J Immunol ; 189(5): 2521-9, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22821964

RESUMO

The immune system is tasked with defending against a myriad of microbial infections, and its response to a given infectious microbe may be strongly influenced by coinfection with another microbe. It was shown that infection of mice with lactate dehydrogenase-elevating virus (LDV) impairs early adaptive immune responses to Friend virus (FV) coinfection. To investigate the mechanism of this impairment, we examined LDV-induced innate immune responses and found LDV-specific induction of IFN-α and IFN-γ. LDV-induced IFN-α had little effect on FV infection or immune responses, but unexpectedly, LDV-induced IFN-γ production dampened Th1 adaptive immune responses and enhanced FV infection. Two distinct effects were identified. First, LDV-induced IFN-γ signaling indirectly modulated FV-specific CD8+ T cell responses. Second, intrinsic IFN-γ signaling in B cells promoted polyclonal B cell activation and enhanced early FV infection, despite promotion of germinal center formation and neutralizing Ab production. Results from this model reveal that IFN-γ production can have detrimental effects on early adaptive immune responses and virus control.


Assuntos
Imunidade Adaptativa , Regulação para Baixo/imunologia , Interferon gama/fisiologia , Vírus da Leucemia Murina/imunologia , Infecções por Retroviridae/imunologia , Imunidade Adaptativa/genética , Animais , Modelos Animais de Doenças , Regulação para Baixo/genética , Feminino , Vírus da Leucemia Murina de Friend/imunologia , Vírus da Leucemia Murina de Friend/patogenicidade , Interferon gama/deficiência , Interferon gama/genética , Vírus Elevador do Lactato Desidrogenase/imunologia , Vírus Elevador do Lactato Desidrogenase/patogenicidade , Vírus da Leucemia Murina/patogenicidade , Leucemia Experimental/genética , Leucemia Experimental/imunologia , Leucemia Experimental/virologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Infecções por Retroviridae/genética , Infecções por Retroviridae/patologia , Vírus Formadores de Foco no Baço/imunologia , Vírus Formadores de Foco no Baço/patogenicidade , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/virologia
8.
J Gen Virol ; 93(Pt 1): 106-112, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21940410

RESUMO

Lactate dehydrogenase-elevating virus (LDV) exacerbates mouse susceptibility to endotoxin shock through enhanced tumour necrosis factor (TNF) production by macrophages exposed to lipopolysaccharide (LPS). However, the in vivo enhancement of TNF production in response to LPS induced by the virus largely exceeds that found in vitro with cells derived from infected animals. Infection was followed by a moderate increase of Toll-like receptor (TLR)-4/MD2, but not of membrane CD14 expression on peritoneal macrophages. Peritoneal macrophages from LDV-infected mice unresponsive to type I interferons (IFNs) did not show enhanced expression of TLR-4/MD2 nor of CD14, and did not produce more TNF in response to LPS than cells from infected normal counterparts, although the in vivo response of these animals to LPS was strongly enhanced. In contrast, the virus triggered a sharp increase of soluble CD14 and of LPS-binding protein serum levels in normal mice. However, production of these LPS soluble receptors was similar in LDV-infected type I IFN-receptor deficient mice and in their normal counterparts. Moreover, serum of LDV-infected mice that contained these soluble receptors had little effect if any on cell response to LPS. These results suggest that enhanced response of LDV-infected mice to LPS results mostly from mechanisms independent of LPS receptor expression.


Assuntos
Infecções por Arterivirus/veterinária , Vírus Elevador do Lactato Desidrogenase/fisiologia , Receptores de Lipopolissacarídeos/genética , Doenças dos Roedores/genética , Doenças dos Roedores/virologia , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/imunologia , Animais , Infecções por Arterivirus/genética , Infecções por Arterivirus/imunologia , Infecções por Arterivirus/virologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Células Cultivadas , Regulação para Baixo , Feminino , Vírus Elevador do Lactato Desidrogenase/imunologia , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/imunologia , Antígeno 96 de Linfócito/genética , Antígeno 96 de Linfócito/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/virologia , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Doenças dos Roedores/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
9.
J Virol ; 83(21): 11211-22, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19692462

RESUMO

Retroviruses can establish persistent infection despite induction of a multipartite antiviral immune response. Whether collective failure of all parts of the immune response or selective deficiency in one crucial part underlies the inability of the host to clear retroviral infections is currently uncertain. We examine here the contribution of virus-specific CD4(+) T cells in resistance against Friend virus (FV) infection in the murine host. We show that the magnitude and duration of the FV-specific CD4(+) T-cell response is directly proportional to resistance against acute FV infection and subsequent disease. Notably, significant protection against FV-induced disease is afforded by FV-specific CD4(+) T cells in the absence of a virus-specific CD8(+) T-cell or B-cell response. Enhanced spread of FV infection in hosts with increased genetic susceptibility or coinfection with Lactate dehydrogenase-elevating virus (LDV) causes a proportional increase in the number of FV-specific CD4(+) T cells required to control FV-induced disease. Furthermore, ultimate failure of FV/LDV coinfected hosts to control FV-induced disease is accompanied by accelerated contraction of the FV-specific CD4(+) T-cell response. Conversely, an increased frequency or continuous supply of FV-specific CD4(+) T cells is both necessary and sufficient to effectively contain acute infection and prevent disease, even in the presence of coinfection. Thus, these results suggest that FV-specific CD4(+) T cells provide significant direct protection against acute FV infection, the extent of which critically depends on the ratio of FV-infected cells to FV-specific CD4(+) T cells.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Vírus da Leucemia Murina de Friend/imunologia , Leucemia Experimental/imunologia , Ativação Linfocitária/imunologia , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , Vírus Elevador do Lactato Desidrogenase/imunologia , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos B/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Interferon/genética , Receptores de Interferon/imunologia , Receptor de Interferon gama
10.
Electromagn Biol Med ; 27(3): 277-88, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18821203

RESUMO

The authors were monitoring adherence ability of T lymphocytes in vitro in patients with laryngeal and pharyngeal carcinoma at the presence of tumor-specific and viral LDH antigen. The results were assessed and expressed in percent of non adherent T lymphocytes (NAL). First, NAL in patients before initiating the treatment was compared with NAL control group (voluntary blood donors). The ability of the adherence in T lymphocytes in the control group is statistically significantly higher. Further on, NAL in the course of a successful oncological treatment was monitored at the interval of 6 months following the treatment, and further on at yearly intervals. NAL level drops statistically significantly within 6 months and then hold on at levels with no statistical difference unlike the control group, however, the ability of T lymphocyte in patients to adhere remains statistically significantly lower. Statistically significantly higher levels of NAL are at the presence of LDH viral antigen. Further on, the authors were following the influence of magnetic sinusoidal field of power frequency (50 Hz) of a low induction (0.5, 0.1, and 0.05 mT) on NAL. NAL values under the influence of an experimental magnetic field before initiating the treatment as well as in the course of a successful oncological treatment are statistically significantly lower. It means that magnetic filed increases the adherence ability of T lymphocytes in patients with laryngeal and pharyngeal cancer in vitro.


Assuntos
Neoplasias Laríngeas/imunologia , Neoplasias Laríngeas/terapia , Leucócitos/patologia , Leucócitos/efeitos da radiação , Magnetismo , Neoplasias Faríngeas/imunologia , Neoplasias Faríngeas/terapia , Animais , Antígenos Virais/imunologia , Estudos de Casos e Controles , Adesão Celular/efeitos da radiação , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/terapia , Humanos , Vírus Elevador do Lactato Desidrogenase/imunologia , Neoplasias Laríngeas/patologia , Camundongos , Neoplasias Faríngeas/patologia , Fatores de Tempo
11.
J Virol ; 82(1): 408-18, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17959678

RESUMO

Friend virus (FV) and lactate dehydrogenase-elevating virus (LDV) are endemic mouse viruses that can cause long-term chronic infections in mice. We found that numerous mouse-passaged FV isolates also contained LDV and that coinfection with LDV delayed FV-specific CD8(+) T-cell responses during acute infection. While LDV did not alter the type of acute pathology induced by FV, which was severe splenomegaly caused by erythroproliferation, the immunosuppression mediated by LDV increased both the severity and the duration of FV infection. Compared to mice infected with FV alone, those coinfected with both FV and LDV had delayed CD8(+) T-cell responses, as measured by FV-specific tetramers. This delayed response accounted for the prolonged and exacerbated acute phase of FV infection. Suppression of FV-specific CD8(+) T-cell responses occurred not only in mice infected concomitantly with LDV but also in mice chronically infected with LDV 8 weeks prior to infection with FV. The LDV-induced suppression was not mediated by T regulatory cells, and no inhibition of the CD4(+) T-cell or antibody responses was observed. Considering that most human adults are carriers of chronically infectious viruses at the time of new virus insults and that coinfections with viruses such as human immunodeficiency virus and hepatitis C virus are currently epidemic, it is of great interest to determine how infection with one virus may impact host responses to a second infection. Coinfection of mice with LDV and FV provides a well-defined, natural host model for such studies.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vírus da Leucemia Murina de Friend/imunologia , Tolerância Imunológica , Vírus Elevador do Lactato Desidrogenase/imunologia , Animais , Anticorpos Antivirais/imunologia , Linfócitos T CD4-Positivos/imunologia , Feminino , Leucemia Eritroblástica Aguda/virologia , Leucemia Experimental/virologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/patologia , Esplenomegalia/virologia , Linfócitos T Reguladores/imunologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/patologia
12.
J Gen Virol ; 88(Pt 11): 3063-3066, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17947531

RESUMO

Two distinct pathways of gamma interferon (IFN-gamma) production have been found in mice infected with lactate dehydrogenase-elevating virus. Both pathways involve natural killer cells. The first is mostly interleukin-12-independent and is not controlled by type I interferons. The second, which is suppressed by type I interferons, leads to increased levels of IFN-gamma production and requires the secretion of interleukin-12. This regulation of IFN-gamma production by type I interferons may help to control indirect pathogenesis induced by this cytokine.


Assuntos
Interferon gama/biossíntese , Células Matadoras Naturais/imunologia , Vírus Elevador do Lactato Desidrogenase/imunologia , Redes e Vias Metabólicas , Animais , Regulação da Expressão Gênica , Interferon Tipo I/imunologia , Interleucina-12/imunologia , Camundongos
13.
Int Immunol ; 19(11): 1303-11, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17914119

RESUMO

The effect of mouse infection with lactate dehydrogenase-elevating virus (LDV), a usually non-pathogenic virus, on concomitant bacterial endotoxin shock was analyzed, in terms of lethality and cytokine production. A strong enhancement of susceptibility to the shock was observed in mice acutely infected with this virus. It correlated with a sharp increase of tumor necrosis factor and leukemia inhibitory factor production and was controlled by the mouse genetic background. The viral infection led to an imbalance in the cytokine response to LPS, with an enhancement of pro-inflammatory cytokines, including IL-18 and IFN-gamma and a delayed secretion of anti-inflammatory IL-10 that could result in exacerbated macrophage activation. Enhanced IFN-gamma production was involved in the virus-induced susceptibility to shock. In sharp contrast with other viral infections, IFN-alpha/beta diminished IFN-gamma production and the resulting increased response to LPS in LDV-infected animals.


Assuntos
Infecções por Arterivirus/imunologia , Endotoxinas/toxicidade , Interferons/imunologia , Vírus Elevador do Lactato Desidrogenase/imunologia , Choque Séptico/imunologia , Animais , Infecções por Arterivirus/virologia , Citocinas/imunologia , Feminino , Vírus Elevador do Lactato Desidrogenase/patogenicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA
14.
Scand J Immunol ; 66(6): 628-35, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17949409

RESUMO

The present study was conducted to determine whether lactate dehydrogenase-elevating virus (LDV) infection at the sensitization and challenge phases affect the development of delayed allergic eosinophilic rhinitis induced by ovalbumin (OVA) in BALB/c mice (DAR group). Compared to the DAR group, LDV infection at the priming (DAR/LDVs group) and immunizing (DAR/LDVc group) phases reduced the induction of eosinophils in the bone marrow (BM) and/or blood. However, the number of eosinophils in the BM was not affected in the DAR/LDVc group. In addition, total blood IgE values were reduced in the DAR/LDVs but not the DAR/LDVc groups. Compared to the production of cytokines from splenic cells and blood IgE values in the DAR group, OVA-specific IL-4 and IFN-gamma productions and IgE values were reduced in the DAR/LDVs, whereas OVA-specific IFN-gamma and IL-4 productions were increased and decreased, respectively in the DAR/LDVc,but not the DAR/LDVs groups. Both DAR/LDVs and DAR/LDVc groups reduced the development of eosinophilic rhinitis associated with reduced VCAM-1 expression on endothelium in blood vessels and ICAM-1 expression on nasal respiratory epithelium at inflamed areas. The present study suggests that LDV infection at the sensitization phase may reduce the development of T helper (Th) 1 and Th2 responses, whereas LDV infection at the challenge phase may inhibit the development of Th2 response by shifting to Th1 response. These may be responsible for the reduction of the development of DAR by LDV infection.


Assuntos
Infecções por Arterivirus/imunologia , Modelos Animais de Doenças , Eosinofilia/imunologia , Imunização , Vírus Elevador do Lactato Desidrogenase/imunologia , Rinite Alérgica Perene/imunologia , Rinite Alérgica Perene/virologia , Animais , Testes de Provocação Brônquica , Eosinofilia/virologia , Feminino , Imunoglobulina E/biossíntese , Fatores Imunológicos/imunologia , Interferon gama/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Mucosa Nasal/imunologia , Mucosa Nasal/virologia , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Rinite Alérgica Perene/prevenção & controle , Especificidade da Espécie , Organismos Livres de Patógenos Específicos/imunologia , Células Th1/imunologia , Células Th1/virologia , Células Th2/imunologia , Células Th2/virologia
15.
Comp Immunol Microbiol Infect Dis ; 27(6): 423-31, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15325515

RESUMO

Lactate dehydrogenase-elevating virus (LDV) has a strict species-specificity and can replicate only in a subset of mouse primary macrophages in vitro. Because it is difficult to grow and purify sufficient quantities of LDV virions from the primary macrophages, it has been difficult to further characterize LDV envelope proteins. A few expression systems have been reported for structural analysis of the nonglycosylated envelope protein M/VP-2, however, very few studies of the antigenicity of M/VP-2 have been reported. We cloned and expressed the ORF6 gene, which encodes the M/VP-2, as a fusion protein with a polyhistidine metal-binding tag (6 x His-tag) in Autographa californica nuclear polyhedrosis virus (baculovirus) under the control of the polyhedrin promoter. In Western blotting analysis, the expressed protein was similar in size to the native M/VP-2 plus 6 x His-tag. The usefulness of the baculovirus-expressed LDV ORF6 protein for analysis of the immunogenicity of LDV M/VP-2 was discussed.


Assuntos
Vírus Elevador do Lactato Desidrogenase/genética , Proteínas do Envelope Viral/genética , Animais , Antígenos Virais/genética , Antígenos Virais/isolamento & purificação , Infecções por Arterivirus/imunologia , Infecções por Arterivirus/virologia , Sequência de Bases , Linhagem Celular , DNA Viral/genética , Genes Virais , Vírus Elevador do Lactato Desidrogenase/imunologia , Camundongos , Nucleopoliedrovírus/genética , Fases de Leitura Aberta , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/isolamento & purificação , Spodoptera , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/isolamento & purificação
16.
Comp Immunol Microbiol Infect Dis ; 27(1): 47-55, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14656541

RESUMO

The elucidation of the antigenic structure of the envelope proteins of Arteriviridae which includes lactate dehydrogenase-elevating virus (LDV) will provide further understanding of a mechanism of strict host cell specificity. To analyze the linkage between LDV envelope proteins, M/VP-2 and VP-3, which may play an important role in viral infectivity, we generated specific antibody against M/VP-2 that has not been reported in previous studies. A synthetic polypeptide corresponding to the C-terminal region of LDV strain C (LDV-C) ORF6, which encodes M/VP-2, was chemically synthesized and coupled to keyhole limpet hemocyanin (KLH). The peptide was immunogenic in rabbits and induced antibody specific for viral protein. Western blotting and immunofluorescence analysis of virion M/VP-2 in infected macrophages showed that the antibody was able to react specifically with authentic virion protein. The immunoreactive antibody against LDV M/VP-2 described in this study will be useful for further studies of the specific roles of the envelope proteins in arterivirus assembly and infectivity.


Assuntos
Anticorpos Antivirais/imunologia , Infecções por Arterivirus/imunologia , Vírus Elevador do Lactato Desidrogenase/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Antivirais/biossíntese , Western Blotting , Ensaio de Imunoadsorção Enzimática , Soros Imunes/biossíntese , Soros Imunes/imunologia , Vírus Elevador do Lactato Desidrogenase/genética , Camundongos , Fases de Leitura Aberta , Coelhos , Proteínas do Envelope Viral/genética
17.
Virus Res ; 92(1): 83-7, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12606079

RESUMO

Lactate dehydrogenase-elevating virus (LDV) is a macrophage-tropic arterivirus which generally causes a persistent viremic infection in mice. LDV replication in vivo seems to be primarily regulated by the extent and dynamics of a virus-permissive macrophage population. Previous studies have shown that glucocorticoid treatment of chronically LDV-infected mice transiently increases viremia 10-100-fold, apparently by increasing the productive infection of macrophages. We have further investigated this phenomenon by comparing the effect of dexamethasone on the in vivo and in vitro replication of two LDV quasispecies that differ in sensitivity to immune control by the host. The single neutralizing epitope of LDV-P is flanked by two N-glycans that impair its immunogenicity and render LDV-P resistant to antibody neutralization. In contrast, replication of the neuropathogenic mutant LDV-C is suppressed by antibody neutralization because its epitope lacks the two protective N-glycans. Dexamethasone treatment of mice 16 h prior to LDV-P infection, or of chronically LDV-P infected mice, stimulated viremia 10-100-fold, which correlated with an increase of LDV permissive macrophages in the peritoneum and increased LDV infected cells in the spleen, respectively. The increase in viremia occurred in the absence of changes in total anti-LDV and neutralizing antibodies. The results indicate that increased viremia was due to increased availability of LDV permissive macrophages, and that during a chronic LDV-P infection virus replication is strictly limited by the rate of regeneration of permissive macrophages. In contrast, dexamethasone treatment had no significant effect on the level of viremia in chronically LDV-C infected mice, consistent with the view that LDV-C replication is primarily restricted by antibody neutralization and not by a lack of permissive macrophages. beta-Glucan, the receptor of which is induced on macrophages by dexamethasone treatment, had no effect on the LDV permissiveness of macrophages.


Assuntos
Dexametasona/farmacologia , Glucocorticoides/farmacologia , Vírus Elevador do Lactato Desidrogenase/efeitos dos fármacos , Vírus Elevador do Lactato Desidrogenase/fisiologia , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Replicação Viral/efeitos dos fármacos , Animais , Anticorpos Antivirais/biossíntese , Infecções por Arterivirus/imunologia , Infecções por Arterivirus/virologia , Feminino , Vírus Elevador do Lactato Desidrogenase/imunologia , Vírus Elevador do Lactato Desidrogenase/patogenicidade , Camundongos , Testes de Neutralização , Baço/efeitos dos fármacos , Baço/virologia
18.
Scand J Immunol ; 57(2): 144-50, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12588660

RESUMO

An immunization protocol that induces antibodies (Abs) directed to cryptic epitopes of a protein antigen (Ag) reduces the efficacy of vaccines that ideally should induce Abs against native epitopes. We have shown earlier that viral infections concomitant with immunization against a protein tend to shift the Ab specificity toward cryptic epitopes and tend to induce the production of autoantibodies (autoAbs). Here, we show the effects of three adjuvants on the Ab specificity in the absence or presence of a viral infection (lactate dehydrogenase-elevating virus or LDV), with human growth hormone (hGH) being, as before, the protein Ag. Pathogen-free CBA/Ht and BALB/c mice were immunized with hGH in the presence of complete Freund's adjuvant (CFA), monophosphoryl lipid A (MPL) or alum, with the animals being either infected with LDV or not infected with LDV. Conventional and competition enzyme-linked immunosorbent assays (ELISAs) indicated that in noninfected mice, CFA induced higher titres of anti-hGH Ab than did MPL or alum, with the Ab being almost totally directed to cryptic hGH epitopes. Strikingly, CFA plus LDV infection in CBA/Ht mice shifted the specificity of the anti-hGH Ab toward native epitopes, whereas the virus decreased the Ab titre when MPL or alum was used. Our Western blot results showed that 70% of mice immunized with hGH in the presence of any adjuvant produced autoAbs against a variety of tissue Ags. The amount of autoAb and the concentration of Ab to hGH cryptic epitopes did correlate, suggesting a relationship between both kinds of Ab. Significant differences were observed in the various effects of adjuvants and the viral infection between the two mouse strains used in this work.


Assuntos
Adjuvantes Imunológicos/farmacologia , Especificidade de Anticorpos/imunologia , Infecções por Arterivirus/imunologia , Epitopos/imunologia , Hormônio do Crescimento Humano/imunologia , Vírus Elevador do Lactato Desidrogenase/imunologia , Lipídeo A/análogos & derivados , Compostos de Alúmen/farmacologia , Animais , Anticorpos Antivirais/sangue , Autoanticorpos/biossíntese , Western Blotting , Ensaio de Imunoadsorção Enzimática , Epitopos/metabolismo , Feminino , Adjuvante de Freund/farmacologia , Rim/imunologia , Lipídeo A/farmacologia , Fígado/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Músculos/imunologia , Miocárdio/imunologia
19.
Viral Immunol ; 16(4): 511-23, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14733738

RESUMO

Persistent infection of mice with lactate dehydrogenase-elevating virus (LDV) is associated with polyclonal B cell activation, autoimmunity, and circulating hydrophobic IgG-containing immune complexes (ICs), which bind to the surfaces of uncoated ELISA plates in the presence of 0.05% Tween 20. We demonstrate here that hydrophobic IgG-containing ICs also appear naturally in the plasma of autoimmune MRL/lpr mice. These and the similar hydrophobic ICs of LDV-infected mice as well as pigs coincide on ELISA plate surfaces with TGF-beta, apparently in the form of an IgG-TGF-beta complex. Circulating hydrophobic IgG-containing ICs are also susceptible to considerable amplification in vitro by exposure to alkaline conditions. By this latter method, the fraction of in vivo hydrophobic IgG, relative to the maximum in vitro chemically inducible IgG, was found to be about 20% in the plasma of LDV-infected mice, 5% in normal mouse plasma, and less than about 2% in pig plasma. These results indicate the potential for both chemically induced and protein-binding contributions to the generation of hydrophobic IgG-containing molecules, and have implications for immunopathological mechanisms in autoimmunity and persistent virus infections.


Assuntos
Complexo Antígeno-Anticorpo/sangue , Infecções por Arterivirus/imunologia , Autoimunidade , Imunoglobulina G/sangue , Vírus Elevador do Lactato Desidrogenase/imunologia , Fator de Crescimento Transformador beta/sangue , Animais , Infecções por Arterivirus/virologia , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Camundongos Endogâmicos MRL lpr , Suínos
20.
J Gen Virol ; 83(Pt 11): 2709-2716, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12388806

RESUMO

Early after infection, lactate dehydrogenase-elevating virus (LDV) alters the immune system by polyclonally activating B lymphocytes, which leads to IgG2a-restricted hypergammaglobulinaemia, and by suppressing the secretion of Th2 cytokines. Considering that these alterations may involve cells of the innate immune system and cytokines such as interferon-gamma (IFN-gamma), we analysed the effect of LDV on natural killer (NK) cells. Within a few days of infection, a strong and transient NK cell activation, characterized by enhanced IFN-gamma message expression and cytolysis, was observed. LDV triggered a large increase in serum IFN-gamma levels. Because NK cells and IFN-gamma may participate in the defence against virus infection, we analysed their possible role in the control of LDV titres with a new agglutination assay. Our results indicate that neither the activation of NK cells nor the IFN-gamma secretion affect the early and rapid virus replication that follows LDV inoculation.


Assuntos
Infecções por Arterivirus/imunologia , Células Matadoras Naturais/imunologia , Vírus Elevador do Lactato Desidrogenase/imunologia , Animais , Infecções por Arterivirus/sangue , Infecções por Arterivirus/virologia , Biomarcadores , Citotoxicidade Imunológica , Expressão Gênica , Integrina alfa2 , Interferon gama/biossíntese , Interferon gama/sangue , Interferon gama/genética , Células Matadoras Naturais/citologia , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Camundongos Knockout , Camundongos SCID , Peritônio/citologia , Receptores de Interferon/genética , Baço/citologia , Viremia , Receptor de Interferon gama
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...