Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Am J Physiol Lung Cell Mol Physiol ; 311(3): L611-27, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27448666

RESUMO

CCR2-expressing leukocytes are required for the progression of fibrosis in models of induced lung injury as well as models of bone marrow transplant (BMT)-related idiopathic pneumonia syndrome. Infection with murid γ-herpesvirus-68 (γHV-68) results in severe pneumonitis and pulmonary fibrosis following syngeneic BMT; however, the roles that various proinflammatory leukocyte populations play in this process remain unclear. Deletion of CCR2 in both non-BMT and BMT mice increased early lytic viral replication and resulted in a reduction in the numbers of lung-infiltrating GR1+,F4/80+ and CXCR1+ cells, while maintaining robust neutrophil infiltration. Similarly, in γHV-68-infected CCR2(-/-) BMT mice, recruitment of monocytes and lymphocytes were reduced whereas neutrophil recruitment was increased compared with wild-type (WT) BMT mice. Interestingly, levels of profibrotic IL-17 were increased in infected CCR2 BMT mice compared with WT BMT. Furthermore, an increase in lung-associated collagen was detected even though there was an overall decrease in the number of profibrotic CCR2+ fibrocytes detected in the lungs of CCR2(-/-) BMT mice. These data indicate that, contrary to most models of fibrosis, deletion of CCR2 offers no protection from γ-herpesvirus-induced pneumonitis and fibrosis, and, indeed, CCR2+ cells play a suppressive role during the development of pulmonary fibrosis following γ-herpesvirus infection post-BMT by limiting IL-7 and collagen production.


Assuntos
Infecções por Herpesviridae/metabolismo , Pneumonia Viral/metabolismo , Fibrose Pulmonar/metabolismo , Receptores CCR2/fisiologia , Animais , Transplante de Medula Óssea , Células Cultivadas , Gammaherpesvirinae/imunologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos , Pneumonia Viral/imunologia , Pneumonia Viral/virologia , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/virologia , Transdução de Sinais , Replicação Viral
2.
Am J Physiol Lung Cell Mol Physiol ; 310(11): L1111-20, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27059285

RESUMO

The production of prostaglandin E2 (PGE2) increases dramatically during pneumococcal pneumonia, and this lipid mediator impairs alveolar macrophage (AM)-mediated innate immune responses. Microsomal prostaglandin E synthase-1 (mPGES-1) is a key enzyme involved in the synthesis of PGE2, and its expression is enhanced during bacterial infections. Genetic deletion of mPGES-1 in mice results in diminished PGE2 production and elevated levels of other prostaglandins after infection. Since PGE2 plays an important immunoregulatory role during bacterial pneumonia we assessed the impact of mPGES-1 deletion in the host defense against pneumococcal pneumonia in vivo and in AMs in vitro. Wild-type (WT) and mPGES-1 knockout (KO) mice were challenged with Streptococcus pneumoniae via the intratracheal route. Compared with WT animals, we observed reduced survival and increased lung and spleen bacterial burdens in mPGES-1 KO mice 24 and 48 h after S. pneumoniae infection. While we found modest differences between WT and mPGES-1 KO mice in pulmonary cytokines, AMs from mPGES-1 KO mice exhibited defective killing of ingested bacteria in vitro that was associated with diminished inducible nitric oxide synthase expression and reduced nitric oxide (NO) synthesis. Treatment of AMs from mPGES-1 KO mice with an NO donor restored bacterial killing in vitro. These results suggest that mPGES-1 plays a critical role in bacterial pneumonia and that genetic ablation of this enzyme results in diminished pulmonary host defense in vivo and in vitro. These results suggest that specific inhibition of PGE2 synthesis by targeting mPGES-1 may weaken host defense against bacterial infections.


Assuntos
Ciclo-Oxigenase 1/genética , Proteínas de Membrana/genética , Pneumonia Pneumocócica/enzimologia , Streptococcus pneumoniae/imunologia , Animais , Citocinas/biossíntese , Citocinas/sangue , Dinoprostona/biossíntese , Feminino , Imunidade Inata , Pulmão/enzimologia , Pulmão/imunologia , Pulmão/microbiologia , Macrófagos Alveolares/enzimologia , Macrófagos Alveolares/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microssomos/enzimologia , Óxido Nítrico/biossíntese , Pneumonia Pneumocócica/imunologia
3.
J Virol ; 89(1): 468-79, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25320326

RESUMO

UNLABELLED: Adenoviruses are frequent causes of pediatric myocarditis. Little is known about the pathogenesis of adenovirus myocarditis, and the species specificity of human adenoviruses has limited the development of animal models, which is a significant barrier to strategies for prevention or treatment. We have developed a mouse model of myocarditis following mouse adenovirus 1 (MAV-1) infection to study the pathogenic mechanisms of this important cause of pediatric myocarditis. Following intranasal infection of neonatal C57BL/6 mice, we detected viral replication and induction of interferon gamma (IFN-γ) in the hearts of infected mice. MAV-1 caused myocyte necrosis and induced substantial cellular inflammation that was composed predominantly of CD3(+) T lymphocytes. Depletion of IFN-γ during acute infection reduced cardiac inflammation in MAV-1-infected mice without affecting viral replication. We observed decreased contractility during acute infection of neonatal mice, and persistent viral infection in the heart was associated with cardiac remodeling and hypertrophy in adulthood. IFN-γ is a proinflammatory mediator during adenovirus-induced myocarditis, and persistent adenovirus infection may contribute to ongoing cardiac dysfunction. IMPORTANCE: Studying the pathogenesis of myocarditis caused by different viruses is essential in order to characterize both virus-specific and generalized factors that contribute to disease. Very little is known about the pathogenesis of adenovirus myocarditis, which is a significant impediment to the development of treatment or prevention strategies. We used MAV-1 to establish a mouse model of human adenovirus myocarditis, providing the means to study host and pathogen factors contributing to adenovirus-induced cardiac disease during acute and persistent infection. The MAV-1 model will enable fundamental studies of viral myocarditis, including IFN-γ modulation as a therapeutic strategy.


Assuntos
Interferon gama/metabolismo , Mastadenovirus/imunologia , Miocardite/imunologia , Miocardite/patologia , Miocárdio/patologia , Animais , Complexo CD3/análise , Feminino , Masculino , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Linfócitos T/química , Linfócitos T/imunologia , Replicação Viral
4.
J Virol ; 86(8): 4194-203, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22345470

RESUMO

There is an incomplete understanding of the differences between neonatal immune responses that contribute to the increased susceptibility of neonates to some viral infections. We tested the hypothesis that neonates are more susceptible than adults to mouse adenovirus type 1 (MAV-1) respiratory infection and are impaired in the ability to generate a protective immune response against a second infection. Following intranasal infection, lung viral loads were greater in neonates than in adults during the acute phase but the virus was cleared from the lungs of neonates as efficiently as it was from adult lungs. Lung gamma interferon (IFN-γ) responses were blunted and delayed in neonates, and lung viral loads were higher in adult IFN-γ(-/-) mice than in IFN-γ(+/+) controls. However, administration of recombinant IFN-γ to neonates had no effect on lung viral loads. Recruitment of inflammatory cells to the airways was impaired in neonates. CD4 and CD8 T cell responses were similar in the lungs of neonates and adults, although a transient increase in regulatory T cells occurred only in the lungs of infected neonates. Infection of neonates led to protection against reinfection later in life that was associated with increased effector memory CD8 T cells in the lungs. We conclude that neonates are more susceptible than adults to acute MAV-1 respiratory infection but are capable of generating protective immune responses.


Assuntos
Infecções por Adenoviridae/imunologia , Infecções por Adenoviridae/prevenção & controle , Adenoviridae/imunologia , Infecções Respiratórias/imunologia , Infecções Respiratórias/prevenção & controle , Infecções por Adenoviridae/genética , Animais , Linhagem Celular , Citocinas/biossíntese , Interferon gama/deficiência , Interferon gama/genética , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Infecções Respiratórias/genética , Linfócitos T/imunologia , Carga Viral
5.
Microbiol Spectr ; 11(3): e0087323, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37154756

RESUMO

By adulthood, the majority of the population is persistently infected with BK polyomavirus (BKPyV). Only a subset of the population, generally transplant recipients on immunosuppressive drugs, will experience disease from BKPyV, but those who do have few treatment options and, frequently, poor outcomes, because to date there are no effective antivirals to treat or approved vaccines to prevent BKPyV. Most studies of BKPyV have been performed on bulk populations of cells, and the dynamics of infection at single-cell resolution have not been explored. As a result, much of our knowledge is based upon the assumption that all cells within a greater population are behaving the same way with respect to infection. The present study examines BKPyV infection on a single-cell level using high-content microscopy to measure and analyze the viral protein large T antigen (TAg), promyelocytic leukemia protein (PML), DNA, and nuclear morphological features. We observed significant heterogeneity among infected cells, within and across time points. We found that the levels of TAg within individual cells did not necessarily increase with time and that cells with the same TAg levels varied in other ways. Overall, high-content, single-cell microscopy is a novel approach to studying BKPyV that enables experimental insight into the heterogenous nature of the infection. IMPORTANCE BK polyomavirus (BKPyV) is a human pathogen that infects nearly everyone by adulthood and persists throughout a person's life. Only people with significant immune suppression develop disease from the virus, however. Until recently the only practical means of studying many viral infections was to infect a group of cells in the laboratory and measure the outcomes in that group. However, interpreting these bulk population experiments requires the assumption that infection influences all cells within a group similarly. This assumption has not held for multiple viruses tested so far. Our study establishes a novel single-cell microscopy assay for BKPyV infection. Using this assay, we discovered differences among individual infected cells that have not been apparent in bulk population studies. The knowledge gained in this study and the potential for future use demonstrate the power of this assay as a tool for understanding the biology of BKPyV.


Assuntos
Vírus BK , Infecções por Polyomavirus , Infecções Tumorais por Vírus , Humanos , Adulto , Microscopia , Proteínas Virais , Antivirais
6.
Elife ; 62017 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-28598328

RESUMO

Influenza virus' low replicative fidelity contributes to its capacity for rapid evolution. Clonal sequencing and fluctuation tests have suggested that the influenza virus mutation rate is 2.7 × 10-6 - 3.0 × 10-5 substitutions per nucleotide per strand copied (s/n/r). However, sequencing assays are biased toward mutations with minimal fitness impacts and fluctuation tests typically investigate only a subset of all possible single nucleotide mutations. We developed a fluctuation test based on reversion to fluorescence in a set of virally encoded mutant green fluorescent proteins, which allowed us to measure the rates of selectively neutral mutations representative of the twelve different mutation types. We measured an overall mutation rate of 1.8 × 10-4 s/n/r for PR8 (H1N1) and 2.5 × 10-4 s/n/r for Hong Kong 2014 (H3N2) and a transitional bias of 2.7-3.6. Our data suggest that each replicated genome will have an average of 2-3 mutations and highlight the importance of mutational load in influenza virus evolution.


Assuntos
Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H3N2/genética , Biologia Molecular/métodos , Taxa de Mutação , Animais , Cães , Fluorometria , Genes Reporter , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Células Madin Darby de Rim Canino , Virologia/métodos
7.
Virology ; 507: 64-74, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28410483

RESUMO

CD8 T cells are key components of the immune response to viruses, but their roles in the pathogenesis of adenovirus respiratory infection have not been characterized. We used mouse adenovirus type 1 (MAV-1) to define CD8 T cell contributions to the pathogenesis of adenovirus respiratory infection. CD8 T cell deficiency in ß2m-/- mice had no effect on peak viral replication in lungs, but clearance of virus was delayed in ß2m-/- mice. Virus-induced weight loss and increases in bronchoalveolar lavage fluid total protein, IFN-γ, TNF-α, IL-10, CCL2, and CCL5 concentrations were less in ß2m-/- mice than in controls. CD8 T cell depletion had similar effects on virus clearance, weight loss, and inflammation. Deficiency of IFN-γ or perforin had no effect on viral replication or inflammation, but perforin-deficient mice were partially protected from weight loss. CD8 T cells promote MAV-1-induced pulmonary inflammation via a mechanism that is independent of direct antiviral effects.


Assuntos
Infecções por Adenoviridae/veterinária , Linfócitos T CD8-Positivos/imunologia , Pulmão/imunologia , Mastadenovirus/fisiologia , Doenças dos Roedores/imunologia , Infecções por Adenoviridae/genética , Infecções por Adenoviridae/imunologia , Infecções por Adenoviridae/virologia , Animais , Feminino , Interferon gama/genética , Interferon gama/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Pulmão/virologia , Masculino , Mastadenovirus/genética , Mastadenovirus/isolamento & purificação , Camundongos , Camundongos Endogâmicos C57BL , Perforina/genética , Perforina/imunologia , Doenças dos Roedores/genética , Doenças dos Roedores/virologia , Replicação Viral
8.
Virus Res ; 214: 26-32, 2016 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-26795547

RESUMO

Neonatal mice are more susceptible than adults to mouse adenovirus type 1 (MAV1) respiratory infection. In adult mice, MAV-1 respiratory infection induces production of prostaglandin E2 (PGE2), a lipid mediator that exerts suppressive effects on a variety of host immune functions. We tested the hypothesis that exaggerated PGE2 production in neonatal mice contributes to increased susceptibility to MAV-1. PGE2 concentrations were lower in lungs of uninfected neonatal mice than in adults. PGE2 production was induced by both MAV-1 and a nonspecific stimulus to a greater degree in neonatal mice than in adults, but only in adults was PGE2 induced in a virus-specific manner. Lung viral loads were equivalent in PGE2-deficient neonatal mice and wild type controls, as was virus-induced expression of IFN-γ, IL-17A, and CCL5 in the lungs. PGE2 deficiency had minimal effect on production of virus-specific IgG or establishment of protective immunity in neonatal mice. Collectively, our data indicate that lung PGE2 production is exaggerated early in life, but this effect does not mediate increased susceptibility to MAV-1 infection.


Assuntos
Infecções por Adenoviridae/metabolismo , Infecções por Adenoviridae/virologia , Adenoviridae , Dinoprostona/metabolismo , Infecções Respiratórias/metabolismo , Infecções Respiratórias/virologia , Imunidade Adaptativa , Infecções por Adenoviridae/genética , Animais , Animais Recém-Nascidos , Anticorpos Antivirais/imunologia , Especificidade de Anticorpos/imunologia , Linhagem Celular , Citocinas/genética , Citocinas/metabolismo , Expressão Gênica , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Knockout , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/metabolismo , Infecções Respiratórias/genética , Carga Viral
9.
Virology ; 498: 57-68, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27560373

RESUMO

The immunoproteasome is an inducible host mechanism that aids in the clearance of damaged proteins. The immunoproteasome also influences immune function by enhancing peptide presentation by MHC class I and promotes inflammation via IκB degradation and activation of NF-κB. We used mouse adenovirus type 1 (MAV-1) to characterize the role of the immunoproteasome in adenovirus pathogenesis. Following intranasal infection of mice, immunoproteasome activity in the heart and lung was significantly increased in an IFN-γ-dependent manner. Absence of the ß5i immunoproteasome subunit and pharmacological inhibition of ß5i activity had minimal effects on viral replication, virus-induced cellular inflammation, or induction of cytokine expression. Likewise, the establishment of protective immunity following primary infection was not significantly altered by ß5i deficiency. Thus, although immunoproteasome activity is robustly induced during acute infection with MAV-1, our data suggest that other mechanisms are capable of compensating for immunoproteasome activity to maintain antiviral immunity and appropriate inflammatory responses.


Assuntos
Infecções por Adenoviridae/imunologia , Infecções por Adenoviridae/metabolismo , Adenoviridae/fisiologia , Imunomodulação , Interferons/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Infecções por Adenoviridae/mortalidade , Infecções por Adenoviridae/virologia , Animais , Imunomodulação/genética , Imunomodulação/imunologia , Cadeias beta de Integrinas/genética , Cadeias beta de Integrinas/metabolismo , Interferon gama/genética , Interferon gama/metabolismo , Interferons/genética , Camundongos , Camundongos Knockout , Miocardite/genética , Miocardite/imunologia , Miocardite/metabolismo , Miocardite/virologia , Complexo de Endopeptidases do Proteassoma/genética
10.
PLoS One ; 10(9): e0139235, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26407316

RESUMO

Adenovirus infections are important complications of bone marrow transplantation (BMT). We demonstrate delayed clearance of mouse adenovirus type 1 (MAV-1) from lungs of mice following allogeneic BMT. Virus-induced prostaglandin E2 (PGE2) production was greater in BMT mice than in untransplanted controls, but BMT using PGE2-deficient donors or recipients failed to improve viral clearance, and treatment of untransplanted mice with the PGE2 analog misoprostol did not affect virus clearance. Lymphocyte recruitment to the lungs was not significantly affected by BMT. Intracellular cytokine staining of lung lymphocytes demonstrated impaired production of INF-γ and granzyme B by cells from BMT mice, and production of IFN-γ, IL-2, IL-4, and IL-17 following ex vivo stimulation was impaired in lymphocytes obtained from lungs of BMT mice. Viral clearance was not delayed in untransplanted INF-γ-deficient mice, suggesting that delayed viral clearance in BMT mice was not a direct consequence of impaired IFN-γ production. However, lung viral loads were higher in untransplanted CD8-deficient mice than in controls, suggesting that delayed MAV-1 clearance in BMT mice is due to defective CD8 T cell function. We did not detect significant induction of IFN-ß expression in lungs of BMT mice or untransplanted controls, and viral clearance was not delayed in untransplanted type I IFN-unresponsive mice. We conclude that PGE2 overproduction in BMT mice is not directly responsible for delayed viral clearance. PGE2-independent effects on CD8 T cell function likely contribute to the inability of BMT mice to clear MAV-1 from the lungs.


Assuntos
Infecções por Adenoviridae/etiologia , Adenoviridae/fisiologia , Transplante de Medula Óssea/efeitos adversos , Dinoprostona/biossíntese , Linfócitos T/fisiologia , Infecções por Adenoviridae/patologia , Infecções por Adenoviridae/virologia , Animais , Líquido da Lavagem Broncoalveolar/química , Citocinas/fisiologia , Dinoprostona/análise , Pulmão/patologia , Pulmão/fisiopatologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Carga Viral
11.
Virology ; 456-457: 259-67, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24889245

RESUMO

IL-17-producing CD4(+) helper T cells (Th17 cells) promote inflammatory responses to many pathogens. We used mouse adenovirus type 1 (MAV-1) to determine contributions of IL-17 to adenovirus pathogenesis. MAV-1 infection of C57BL/6 mice upregulated lung expression of IL-17 and the Th17-associated factors IL-23 and RORγt. Only CD4(+)T cells were associated with virus-specific IL-17 production. Fewer neutrophils were recruited to airways of IL-17(-/-) mice following MAV-1 infection, but there were no other differences in pulmonary inflammation between IL-17(+/+) and IL-17(-/-) mice. Mice depleted of neutrophils using anti-Gr-1 antibody had greater lung viral loads than controls. Despite impaired neutrophil recruitment, there were no differences between IL-17(+/+) and IL-17(-/-) mice in peak lung viral loads, clearance of virus from the lungs, or establishment of protective immunity. We demonstrate robust Th17 responses during MAV-1 respiratory infection, but these responses are not essential for control of virus infection or for virus-induced pulmonary inflammation.


Assuntos
Infecções por Adenoviridae/imunologia , Adenoviridae/imunologia , Interações Hospedeiro-Patógeno , Interleucina-17/imunologia , Infiltração de Neutrófilos , Infecções Respiratórias/imunologia , Replicação Viral , Adenoviridae/fisiologia , Animais , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
PLoS One ; 8(10): e77628, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24147040

RESUMO

Respiratory viruses cause substantial disease and are a significant healthcare burden. Virus-induced inflammation can be detrimental to the host, causing symptoms during acute infection and leading to damage that contributes to long-term residual lung disease. Prostaglandin E2 (PGE2) is a lipid mediator that is increased in response to many viral infections, and inhibition of PGE2 production during respiratory viral infection often leads to a decreased inflammatory response. We tested the hypothesis that PGE2 promotes inflammatory responses to mouse adenovirus type 1 (MAV-1) respiratory infection. Acute MAV-1 infection increased COX-2 expression and PGE2 production in wild type mice. Deficiency of the E prostanoid 2 receptor had no apparent effect on MAV-1 pathogenesis. Virus-induced induction of PGE2, IFN-γ, CXCL1, and CCL5 was reduced in mice deficient in microsomal PGE synthase-1 (mPGES-1(-/-) mice). However, there were no differences between mPGES-1(+/+) and mPGES-1(-/-) mice in viral replication, recruitment of leukocytes to airways or lung inflammation. Infection of both mPGES­1(+/+) and mPGES-1(-/-) mice led to protection against reinfection. Thus, while PGE2 promotes the expression of a variety of cytokines in response to acute MAV-1 infection, PGE2 synthesis does not appear to be essential for generating pulmonary immunity.


Assuntos
Adenoviridae/patogenicidade , Dinoprostona/metabolismo , Infecções Respiratórias/imunologia , Adenoviridae/imunologia , Animais , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Dinoprostona/genética , Interferon gama/genética , Interferon gama/metabolismo , Masculino , Camundongos , Camundongos Mutantes , Reação em Cadeia da Polimerase em Tempo Real , Infecções Respiratórias/metabolismo , Infecções Respiratórias/virologia , Replicação Viral/genética , Replicação Viral/fisiologia
13.
Virus Res ; 160(1-2): 351-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21816184

RESUMO

Muc1 (MUC1 in humans) is a membrane-tethered mucin that exerts anti-inflammatory effects in the lung during bacterial infection. Muc1 and other mucins are also likely to form a protective barrier in the lung. We used mouse adenovirus type 1 (MAV-1, also known as MAdV-1) to determine the role of Muc1 in the pathogenesis of an adenovirus in its natural host. Following intranasal inoculation of wild type mice, we detected increased TNF-α, a cytokine linked to Muc1 production, but no consistent changes in the production of lung Muc1, Muc5ac or overall lung mucus production. Viral loads were modestly higher in the lungs of Muc1(-/-) mice compared to Muc1(+/+) mice at several early time points but decreased to similar levels by 14 days post infection in both groups. However, cellular inflammation and the expression of CXCL1, CCL5, and CCL2 did not significantly differ between Muc1(-/-) and Muc1(+/+) mice. Our data therefore suggest that Muc1 may contribute to a physical barrier that protects against MAV-1 respiratory infection. However, our data do not reveal an anti-inflammatory effect of Muc1 that contributes to MAV-1 pathogenesis.


Assuntos
Infecções por Adenoviridae/veterinária , Adenoviridae/patogenicidade , Mucina-1/imunologia , Infecções Respiratórias/veterinária , Doenças dos Roedores/imunologia , Doenças dos Roedores/patologia , Adenoviridae/imunologia , Infecções por Adenoviridae/imunologia , Infecções por Adenoviridae/patologia , Infecções por Adenoviridae/virologia , Animais , Citocinas/metabolismo , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções Respiratórias/imunologia , Infecções Respiratórias/patologia , Infecções Respiratórias/virologia , Doenças dos Roedores/virologia , Carga Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA