Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Aust Vet J ; 98(5): 173-180, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32037517

RESUMO

OBJECTIVE: As ownership of brachycephalic dog breeds rises, the surgical correction of components of brachycephalic airway syndrome (BAS) is increasingly recommended by veterinarians. This study's objective was to describe the incidence of, and strategies for the management of post-operative respiratory complications in brachycephalic dogs undergoing surgical correction of one or more components of BAS. METHODS: Medical records of 248 brachycephalic dogs treated surgically for BAS were retrospectively reviewed for demographic information, procedures performed, post-operative complications and treatment implemented, hospitalisation time, and necessity for further surgery. RESULTS: Pugs, Cavalier King Charles Spaniels and British Bulldogs were the most commonly encountered breeds. Dogs which experienced a complication were significantly older (mean was 5.5 years, compared with 4.1 years [P < 0.01]). Fifty-eight dogs (23.4%) had complications which included: dyspnoea managed with supplemental oxygen alone (7.3%, n = 18), dyspnoea requiring anaesthesia and re-intubation (8.9%, n = 22), dyspnoea necessitating treatment with a temporary tracheostomy (8.9%, n = 22), aspiration pneumonia (4%, n = 10), and respiratory or cardiac arrest (2.4%, n = 6). Five of the 22 dogs requiring anaesthesia and re-intubation deteriorated 12 or more hours after post-surgical anaesthetic recovery. The overall mortality rate in this study was 2.4% (n = 6). Age, concurrent airway pathology, and emergency presentation significantly predicted post-operative complications. CONCLUSION: Our data show the importance of close monitoring for a minimum of 24 h following surgery by an experienced veterinarian or veterinary technician. Surgical intervention for BAS symptomatic dogs should be considered at an earlier age as an elective procedure, to reduce the risk of post-operative complications.


Assuntos
Obstrução das Vias Respiratórias/veterinária , Craniossinostoses/veterinária , Doenças do Cão , Animais , Cães , Humanos , Complicações Pós-Operatórias/veterinária , Período Pós-Operatório , Estudos Retrospectivos
2.
mSphere ; 1(3)2016.
Artigo em Inglês | MEDLINE | ID: mdl-27303748

RESUMO

The gp41 membrane-proximal external region (MPER) is a target for broadly neutralizing antibody responses against human immunodeficiency virus type 1 (HIV-1). However, replication-defective virus vaccines currently under evaluation in clinical trials do not efficiently elicit MPER-specific antibodies. Structural modeling suggests that the MPER forms an α-helical coiled coil that is required for function and immunogenicity. To maintain the native MPER conformation, we used reverse genetics to engineer replication-competent reovirus vectors that displayed MPER sequences in the α-helical coiled-coil tail domain of viral attachment protein σ1. Sequences in reovirus strain type 1 Lang (T1L) σ1 were exchanged with sequences encoding HIV-1 strain Ba-L MPER epitope 2F5 or the entire MPER. Individual 2F5 or MPER substitutions were introduced at virion-proximal or virion-distal sites in the σ1 tail. Recombinant reoviruses containing heterologous HIV-1 sequences were viable and produced progeny yields comparable to those with wild-type virus. HIV-1 sequences were retained following 10 serial passages in cell culture, indicating that the substitutions were genetically stable. Recombinant viruses engineered to display the 2F5 epitope or full-length MPER in σ1 were recognized by purified 2F5 antibody. Inoculation of mice with 2F5-containing vectors or rabbits with 2F5- or MPER-containing vectors elicited anti-reovirus antibodies, but HIV-1-specific antibodies were not detected. Together, these findings indicate that heterologous sequences that form α-helices can functionally replace native sequences in the α-helical tail domain of reovirus attachment protein σ1. However, although these vectors retain native antigenicity, they were not immunogenic, illustrating the difficulty of experimentally inducing immune responses to this essential region of HIV-1. IMPORTANCE Vaccines to protect against HIV-1, the causative agent of AIDS, are not approved for use. Antibodies that neutralize genetically diverse strains of HIV-1 bind to discrete regions of the envelope glycoproteins, including the gp41 MPER. We engineered recombinant reoviruses that displayed MPER epitopes in attachment protein σ1 (REO-MPER vectors). The REO-MPER vectors replicated with wild-type efficiency, were genetically stable, and retained native antigenicity. However, we did not detect HIV-1-specific immune responses following inoculation of the REO-MPER vectors into small animals. This work provides proof of principle for engineering reovirus to express antigenic epitopes and illustrates the difficulty in eliciting MPER-specific immune responses.

3.
J Relig Health ; 54(2): 664-75, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25119628

RESUMO

Positive health outcomes are related to adults' religious congregational participation. For parents of children with chronic disease, structured daily care routines and/or strict infection control precautions may limit participation. For this exploratory study, we examined the relationship between congregational support and religious coping by parents of children with cystic fibrosis (CF) compared to parents for whom child health issues were not significant stressors. CF parents reported higher levels of emotional support from congregation members and use of religious coping. Within-group differences were found for CF parents by denominational affiliation. Congregational support for parents dealing with child chronic disease is important.


Assuntos
Adaptação Psicológica , Atitude Frente a Saúde , Fibrose Cística/psicologia , Pais/psicologia , Religião e Psicologia , Apoio Social , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino
4.
BMJ Qual Saf ; 23 Suppl 1: i56-i63, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24608552

RESUMO

BACKGROUND: In 2001, Cincinnati Children's Hospital embarked on a journey to improve healthcare delivery to patients with cystic fibrosis (CF). Data from the Cystic Fibrosis Foundation National Patient Registry revealed our below-average clinical outcomes, prompting us to initiate improvement interventions. OBJECTIVE: To improve clinical outcomes for patients with CF through a comprehensive quality-improvement approach directed at increasing patient centredness and improving healthcare delivery. INTERVENTIONS: In 2001, we shared our below-average outcomes with patients, families and care providers. We instituted a quality-improvement steering committee with parental and hospital leadership, and our data-management support was restructured to provide real-time clinical data to monitor our progress. In 2002, our weekly chart conference changed to a prospective planning session and individualised daily schedules were created for inpatients. In 2003, an influenza vaccination campaign was initiated and our infection-control practices were redesigned. In 2005, best-practice guidelines were developed for airway-clearance therapy. In 2007, evidence-based clinical algorithms were designed and implemented and key care-team members were added. MEASUREMENTS: Primary outcome measures were median forced expiratory volume in 1 s per cent predicted (age range 6-17 years) and median body mass index percentile (age range 2-20 years). RESULTS: From 2000 to 2010, median forced expiratory volume in 1 s increased from 81.7% to 100.1% predicted and median body mass index increased from the 35th to the 55th centile. DISCUSSION: By focusing on specific outcomes, empowering families and patients, effectively using data, and standardising care processes, we transformed the culture and delivery of care for our patients with CF and learned valuable lessons potentially translatable to other chronic-care providers.


Assuntos
Fibrose Cística/terapia , Atenção à Saúde/organização & administração , Equipe de Assistência ao Paciente/organização & administração , Garantia da Qualidade dos Cuidados de Saúde , Adolescente , Criança , Fibrose Cística/fisiopatologia , Feminino , Pesquisas sobre Atenção à Saúde , Hospitais Pediátricos , Humanos , Masculino , Ohio , Assistência ao Paciente/métodos , Avaliação de Programas e Projetos de Saúde , Melhoria de Qualidade , Fatores de Tempo , Adulto Jovem
5.
J Gen Virol ; 94(Pt 6): 1296-1300, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23426355

RESUMO

Infectious entry of the nonenveloped rotavirus virion requires proteolysis of the spike protein VP4 to mediate conformational changes associated with membrane penetration. We sequenced and characterized an isolate that was cultured in the absence of trypsin and found that it is more resistant to proteolysis than WT virus. A substitution mutation abrogates one of the defined trypsin-cleavage sites, suggesting that blocking proteolysis at this site reduces the overall kinetics of proteolysis. Kinetic analysis of the membrane penetration-associated conformational change indicated that the 'fold-back' of the mutant spike protein is slower than that of WT. Despite these apparent biochemical defects, the mutant virus replicates in an identical manner to the WT virus. These findings enhance an understanding of VP4 functions and establish new strategies to interrogate rotavirus cell entry.


Assuntos
Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Mutação , Infecções por Rotavirus/enzimologia , Infecções por Rotavirus/virologia , Rotavirus/fisiologia , Tripsina/metabolismo , Sequência de Aminoácidos , Animais , Interações Hospedeiro-Patógeno , Humanos , Macaca mulatta , Dados de Sequência Molecular , Processamento de Proteína Pós-Traducional , Rotavirus/química , Rotavirus/genética , Alinhamento de Sequência , Internalização do Vírus , Replicação Viral
6.
Virology ; 433(2): 489-97, 2012 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-22999096

RESUMO

Human milk contains many bioactive components, including secretory IgA, oligosaccharides, and milk-associated proteins. We assessed the antiviral effects of several components of milk against mammalian reoviruses. We found that glucocerebroside (GCB) inhibited the infectivity of reovirus strain type 1 Lang (T1L), whereas gangliosides GD3 and GM3 and 3'-sialyllactose (3SL) inhibited the infectivity of reovirus strain type 3 Dearing (T3D). Agglutination of erythrocytes mediated by T1L and T3D was inhibited by GD3, GM3, and bovine lactoferrin. Additionally, α-sialic acid, 3SL, 6'-sialyllactose, sialic acid, human lactoferrin, osteopontin, and α-lactalbumin inhibited hemagglutination mediated by T3D. Using single-gene reassortant viruses, we found that serotype-specific differences segregate with the gene encoding the viral attachment protein. Furthermore, GD3, GM3, and 3SL inhibit T3D infectivity by blocking binding to host cells, whereas GCB inhibits T1L infectivity post-attachment. These results enhance an understanding of reovirus cell attachment and define a mechanism for the antimicrobial activity of human milk.


Assuntos
Proteínas do Capsídeo/imunologia , Orthoreovirus Mamífero 3/imunologia , Orthoreovirus Mamífero 3/patogenicidade , Leite Humano/imunologia , Orthoreovirus de Mamíferos/imunologia , Orthoreovirus de Mamíferos/patogenicidade , Polissacarídeos/imunologia , Animais , Proteínas do Capsídeo/genética , Bovinos , Feminino , Gangliosídeo G(M3)/imunologia , Gangliosídeos/imunologia , Genes Virais , Células HeLa , Testes de Inibição da Hemaglutinação , Interações Hospedeiro-Patógeno/imunologia , Humanos , Células L , Orthoreovirus Mamífero 3/classificação , Orthoreovirus Mamífero 3/genética , Camundongos , Leite Humano/virologia , Oligossacarídeos/imunologia , Orthoreovirus de Mamíferos/classificação , Orthoreovirus de Mamíferos/genética , Infecções por Reoviridae/imunologia , Infecções por Reoviridae/prevenção & controle , Infecções por Reoviridae/virologia , Sorotipagem , Especificidade da Espécie , Ligação Viral
7.
J Virol Methods ; 184(1-2): 1-7, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22465252

RESUMO

Current microscopy-based approaches for immunofluorescence detection of viral infectivity are time consuming and labor intensive and can yield variable results subject to observer bias. To circumvent these problems, we developed a rapid and automated infrared immunofluorescence imager-based infectivity assay for both rotavirus and reovirus that can be used to quantify viral infectivity and infectivity inhibition. For rotavirus, monolayers of MA104 cells were infected with simian strain SA-11 or SA-11 preincubated with rotavirus-specific human IgA. For reovirus, monolayers of either HeLa S3 cells or L929 cells were infected with strains type 1 Lang (T1L), type 3 Dearing (T3D), or either virus preincubated with a serotype-specific neutralizing monoclonal antibody (mAb). Infected cells were fixed and incubated with virus-specific polyclonal antiserum, followed by an infrared fluorescence-conjugated secondary antibody. Well-to-well variation in cell number was normalized using fluorescent reagents that stain fixed cells. Virus-infected cells were detected by scanning plates using an infrared imager, and results were obtained as a percent response of fluorescence intensity relative to a virus-specific standard. An expected dose-dependent inhibition of both SA-11 infectivity with rotavirus-specific human IgA and reovirus infectivity with T1L-specific mAb 5C6 and T3D-specific mAb 9BG5 was observed, confirming the utility of this assay for quantification of viral infectivity and infectivity blockade. The imager-based viral infectivity assay fully automates data collection and provides an important advance in technology for applications such as screening for novel modulators of viral infectivity. This basic platform can be adapted for use with multiple viruses and cell types.


Assuntos
Automação/métodos , Imunofluorescência/métodos , Processamento de Imagem Assistida por Computador/métodos , Reoviridae/patogenicidade , Rotavirus/patogenicidade , Virologia/métodos , Linhagem Celular , Humanos
8.
J Biol Chem ; 287(11): 8029-38, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22253447

RESUMO

Following attachment and internalization, mammalian reoviruses undergo intracellular proteolytic disassembly followed by viral penetration into the cytoplasm. The initiating event in reovirus disassembly is the cathepsin-mediated proteolytic degradation of viral outer capsid protein σ3. A single tyrosine-to-histidine mutation at amino acid 354 (Y354H) of strain type 3 Dearing (T3D) σ3 enhances reovirus disassembly and confers resistance to protease inhibitors such as E64. The σ3 amino acid sequence of strain type 3 Abney (T3A) differs from that of T3D at eight positions including Y354H. However, T3A displays disassembly kinetics and protease sensitivity comparable with T3D. We hypothesize that one or more additional σ3 polymorphisms suppress the Y354H phenotype and restore T3D disassembly characteristics. To test this hypothesis, we engineered a panel of reovirus variants with T3A σ3 polymorphisms introduced individually into T3D-σ3Y354H. We evaluated E64 resistance and in vitro cathepsin L susceptibility of these viruses and found that one containing a glycine-to-glutamate substitution at position 198 (G198E) displayed disassembly kinetics and E64 sensitivity similar to those properties of T3A and T3D. Additionally, viruses containing changes at positions 233 and 347 (S233L and I347T) developed de novo compensatory mutations at position 198, strengthening the conclusion that residue 198 is a key determinant of σ3 proteolytic susceptibility. Variants with Y354H in σ3 lost infectivity more rapidly than T3A or T3D following heat treatment, an effect abrogated by G198E. These results identify a regulatory network of residues that control σ3 cleavage and capsid stability, thus providing insight into the regulation of nonenveloped virus disassembly.


Assuntos
Proteínas do Capsídeo/metabolismo , Capsídeo/metabolismo , Orthoreovirus de Mamíferos/metabolismo , Proteólise , Substituição de Aminoácidos , Animais , Proteínas do Capsídeo/genética , Catepsina L/genética , Catepsina L/metabolismo , Linhagem Celular , Camundongos , Mutação de Sentido Incorreto , Orthoreovirus de Mamíferos/genética , Infecções por Reoviridae/genética , Infecções por Reoviridae/metabolismo
9.
J Virol ; 83(19): 9630-40, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19640986

RESUMO

The cathepsin family of endosomal proteases is required for proteolytic processing of several viruses during entry into host cells. Mammalian reoviruses utilize cathepsins B (Ctsb), L (Ctsl), and S (Ctss) for disassembly of the virus outer capsid and activation of the membrane penetration machinery. To determine whether cathepsins contribute to reovirus tropism, spread, and disease outcome, we infected 3-day-old wild-type (wt), Ctsb(-/-), Ctsl(-/-), and Ctss(-/-) mice with the virulent reovirus strain T3SA+. The survival rate of Ctsb(-/-) mice was enhanced in comparison to that of wt mice, whereas the survival rates of Ctsl(-/-) and Ctss(-/-) mice were diminished. Peak titers at sites of secondary replication in all strains of cathepsin-deficient mice were lower than those in wt mice. Clearance of the virus was delayed in Ctsl(-/-) and Ctss(-/-) mice in comparison to the levels for wt and Ctsb(-/-) mice, consistent with a defect in cell-mediated immunity in mice lacking cathepsin L or S. Cathepsin expression was dispensable for establishment of viremia, but cathepsin L was required for maximal reovirus growth in the brain. Treatment of wt mice with an inhibitor of cathepsin L led to amelioration of reovirus infection. Collectively, these data indicate that cathepsins B, L, and S influence reovirus pathogenesis and suggest that pharmacologic modulation of cathepsin activity diminishes reovirus disease severity.


Assuntos
Catepsina B/genética , Catepsinas/genética , Cisteína Endopeptidases/genética , Regulação da Expressão Gênica , Infecções por Reoviridae/genética , Reoviridae/metabolismo , Animais , Encéfalo/virologia , Catepsina B/metabolismo , Catepsina L , Catepsinas/metabolismo , Linhagem Celular , Cisteína Endopeptidases/metabolismo , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Fatores de Tempo
10.
Neuroscience ; 154(3): 1042-53, 2008 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-18511209

RESUMO

Dopamine increases in the nucleus accumbens after ethanol administration in rats, but the contributions of the core and shell subregions to this response are unclear. The goal of this study was to determine the effect of various doses of i.v. ethanol infusions on dopamine in these two subregions of the nucleus accumbens. Male Long-Evans rats were infused with either acute i.v. ethanol (0.5, 1.0, 1.5 g/kg), repeated i.v. ethanol (four 1.0 g/kg infusions resulting in a cumulative dose of 4.0 g/kg), or saline as a control for each condition. Dopamine and ethanol were measured in dialysate samples from each experiment. The in vivo extraction fraction for ethanol of probes was determined using i.v. 4-methylpyrazole, and was used to estimate peak brain ethanol concentrations after the infusions. The peak brain ethanol concentrations after the 0.5, 1.0 and 1.5 g/kg ethanol infusions were estimated to be 20, 49 and 57 mM, respectively. A significant dopamine increase was observed for the 0.5 g/kg ethanol group when collapsed across subregions. However, both the 1.0 g/kg and 1.5 g/kg ethanol infusions produced significant increases in dopamine levels in the shell that were significantly higher than those in the core. An ethanol dose-response effect on dopamine in the shell was observed when saline controls, 0.5, 1.0, and 1.5 g/kg groups were compared. For the cumulative-dosing study, the first, second, and fourth infusions resulted in significant increases in dopamine in the shell. However, these responses were not significantly different from one another. The results of this study show that the shell has a stronger response than the core to i.v. ethanol, that dopamine in the shell increases in a dose-dependent manner between 0.5-1.0 g/kg doses, but that the response to higher ethanol doses reaches a plateau.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Dopamina/metabolismo , Etanol/farmacologia , Núcleo Accumbens/metabolismo , Álcool Desidrogenase/antagonistas & inibidores , Animais , Comportamento Animal/efeitos dos fármacos , Depressores do Sistema Nervoso Central/administração & dosagem , Depressores do Sistema Nervoso Central/farmacocinética , Cromatografia Gasosa , Cromatografia Líquida de Alta Pressão , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Etanol/administração & dosagem , Etanol/farmacocinética , Fomepizol , Infusões Intravenosas , Masculino , Microdiálise , Núcleo Accumbens/anatomia & histologia , Núcleo Accumbens/efeitos dos fármacos , Pirazóis/farmacologia , Ratos , Ratos Long-Evans
11.
J Infect Dis ; 197(8): 1189-97, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18419529

RESUMO

Mammalian reoviruses infect respiratory and gastrointestinal epithelia and cause disease in neonates. Junctional adhesion molecule-A (JAM-A) is a serotype-independent receptor for reovirus. JAM-A localizes to tight junctions and contributes to paracellular permeability in polarized epithelia. To investigate the mechanisms of reovirus infection of polarized epithelial cells, we assessed reovirus replication, release, and spread after apical and basolateral adsorption to primary human airway epithelial cultures. Reovirus infection of human airway epithelia was more efficient after adsorption to the basolateral surface than after adsorption to the apical surface, and it was dependent on JAM-A. Reovirus binding to carbohydrate coreceptor sialic acid inhibited apical infection, which was partially ameliorated by treatment of the cultures with neuraminidase. Despite the preference for basolateral infection, reovirus was released from the apical surface of respiratory epithelia and did not disrupt tight junctions. These results establish the existence of an infectious circuit for reovirus in polarized human respiratory epithelial cells.


Assuntos
Orthoreovirus de Mamíferos/patogenicidade , Infecções por Reoviridae/virologia , Mucosa Respiratória/virologia , Infecções Respiratórias/virologia , Animais , Moléculas de Adesão Celular/metabolismo , Polaridade Celular/fisiologia , Impedância Elétrica , Humanos , Imunoglobulinas/metabolismo , Imuno-Histoquímica , Células L , Camundongos , Microscopia Confocal , Neuraminidase/farmacologia , Orthoreovirus de Mamíferos/metabolismo , Vírus Reordenados/patogenicidade , Receptores de Superfície Celular , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo , Junções Íntimas/metabolismo , Junções Íntimas/virologia , Eliminação de Partículas Virais
12.
Cell Host Microbe ; 1(2): 147-57, 2007 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-18005692

RESUMO

Mammalian orthoreoviruses (reoviruses) are highly tractable experimental models for studies of double-stranded (ds) RNA virus replication and pathogenesis. Reoviruses infect respiratory and intestinal epithelium and disseminate systemically in newborn animals. Until now, a strategy to rescue infectious virus from cloned cDNA has not been available for any member of the Reoviridae family of dsRNA viruses. We report the generation of viable reovirus following plasmid transfection of murine L929 (L) cells using a strategy free of helper virus and independent of selection. We used the reovirus reverse genetics system to introduce mutations into viral capsid proteins sigma1 and sigma3 and to rescue a virus that expresses a green fluorescent protein (GFP) transgene, thus demonstrating the tractability of this technology. The plasmid-based reverse genetics approach described here can be exploited for studies of reovirus replication and pathogenesis and used to develop reovirus as a vaccine vector.


Assuntos
Vírus de RNA/genética , RNA de Cadeia Dupla/genética , Infecções por Reoviridae/genética , Vacinas Sintéticas , Vacinas Virais , Animais , Modelos Animais de Doenças , Genoma Viral , Mamíferos , Plasmídeos , Vírus de RNA/imunologia , RNA de Cadeia Dupla/imunologia , Recombinação Genética , Reoviridae/genética , Reoviridae/imunologia , Infecções por Reoviridae/imunologia , Transfecção
13.
J Exp Med ; 204(6): 1349-58, 2007 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-17502662

RESUMO

We defined the function of type I interferons (IFNs) in defense against reovirus strain type 1 Lang (T1L), which is a double-stranded RNA virus that infects Peyer's patches (PPs) after peroral inoculation of mice. T1L induced expression of mRNA for IFN-alpha, IFN-beta, and Mx-1 in PPs and caused localized intestinal infection that was cleared in 10 d. In contrast, T1L produced fatal systemic infection in IFNalphaR1 knockout (KO) mice with extensive cell loss in lymphoid tissues and necrosis of the intestinal mucosa. Studies of bone-marrow chimeric mice indicated an essential role for hematopoietic cells in IFN-dependent viral clearance. Dendritic cells (DCs), including conventional DCs (cDCs), were the major source of type I IFNs in PPs of reovirus-infected mice, whereas all cell types expressed the antiviral protein Mx-1. Neither NK cells nor signaling via Toll-like receptor 3 or MyD88 were essential for viral clearance. These data demonstrate a requirement for type I IFNs in the control of an intestinal viral infection and indicate that cDCs are a significant source of type I IFN production in vivo. Therefore, innate immunity in PPs is an essential component of host defense that limits systemic spread of pathogens that infect the intestinal mucosa.


Assuntos
Células da Medula Óssea/imunologia , Células Dendríticas/imunologia , Interferon Tipo I/imunologia , Orthoreovirus de Mamíferos/imunologia , Nódulos Linfáticos Agregados/imunologia , Infecções por Reoviridae/prevenção & controle , Animais , Imuno-Histoquímica , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nódulos Linfáticos Agregados/virologia , Infecções por Reoviridae/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sobrevida
14.
Ann Biomed Eng ; 34(11): 1778-85, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17031592

RESUMO

We previously described a filament-based antibody recognition assay (FARA) that generates ELISA-like sandwich structures immobilized on a filament. FARA allows the coupling of antibodies to precise locations along a filament, on-line fluorescence detection of captured pathogen, and feedback-directed filament motion. These properties suggest that this approach might be useful as an automated means to rapidly classify unknown pathogens. In this report, we describe validation of the novel decision tree aspect of this technology using mammalian reovirus. Based on available antibodies, we developed a decision tree algorithm to detect virus with increasing specificity at each level of the tree. Using three strains of reovirus and a bacteriophage control, our system correctly classified the reovirus strains at a concentration of 2 x 10(12) virions ml(-1) and M13K07 phage at 3 x 10(11) virions ml(-1). Classification of reovirus strain type 3 Dearing (T3D) required three levels of testing: general reovirus classification in level 1, serotype 3 classification in level 2, and final T3D strain classification in level 3. Strain T3SA + also required three levels of testing before a final classification was returned in level 3. Classification of strain type 1 Lang (T1L) required two levels of testing. M13K07 phage detection required only one level of testing for classification. These results indicate that automated pathogen classification using FARA is feasible. Furthermore, the simplicity of the design could be exploited for development of more complex sub-classification networks with additional levels and branches.


Assuntos
Anticorpos Antivirais/imunologia , Técnicas Biossensoriais/instrumentação , Fluorimunoensaio/instrumentação , Reoviridae/imunologia , Reoviridae/isolamento & purificação , Técnicas Biossensoriais/métodos , Desenho de Equipamento , Análise de Falha de Equipamento , Fluorimunoensaio/métodos , Reoviridae/classificação
15.
Viral Immunol ; 19(3): 546-57, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16987072

RESUMO

Mammalian Orthoreoviruses are important models for studies of viral pathogenesis. In the rat lung, Reovirus strain type 3 Dearing (T3D) induces substantially more inflammation than does strain type 1 Lang (T1L). To better understand mechanisms underlying differences in the host inflammatory response elicited by T1L and T3D, we characterized cytokine expression patterns induced by those strains after infection of THP-1 monocyte cells. THP-1 cells were adsorbed with either viable or ultraviolet- inactivated T1L and T3D and assayed for mRNA and protein production of growth-regulated oncogene-alpha (GRO-alpha), interleukin-8 (IL-8), or tumor necrosis factor-alpha (TNF-alpha). T3D stimulated mRNA and protein production of all three cytokines, whereas T1L stimulated mRNA and protein production of IL-8 and TNF-alpha but not GRO-alpha. In each case, T3D induced greater cytokine mRNA and protein expression than did T1L. Nonviable virus did not stimulate detectable cytokine secretion, suggesting a requirement for viral RNA synthesis in cytokine induction by THP-1 cells. A greater percentage of THP-1 cells was infected with T1L than T3D as assessed by infectious center assay, and T1L achieved higher yields of infectious progeny than did T3D in infected THP-1 cells as determined by plaque assay. These strain-dependent differences in cytokine responses and corresponding replication patterns in monocyte cells parallel findings made in studies of rat models of pneumonia and provide clues about how Reovirus interfaces with the host innate immune response to produce pulmonary disease.


Assuntos
Citocinas/metabolismo , Inflamação/virologia , Orthoreovirus Mamífero 3/imunologia , Monócitos/virologia , Orthoreovirus de Mamíferos/imunologia , Linhagem Celular , Citocinas/genética , Humanos , Inflamação/imunologia , Orthoreovirus Mamífero 3/fisiologia , Orthoreovirus de Mamíferos/fisiologia , Especificidade da Espécie , Replicação Viral
16.
J Virol ; 80(6): 3078-82, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16501117

RESUMO

Mice infected with reovirus develop abnormalities in glucose homeostasis. Reovirus strain type 3 Abney (T3A) was capable of systemic infection of nonobese diabetic (NOD) mice, an experimental model of autoimmune diabetes. Reovirus antigen was detected in pancreatic islets of T3A-infected mice, and primary cultures of pancreatic islets from NOD mice supported T3A growth. Significantly fewer T3A-infected animals compared to uninfected controls developed diabetes. However, despite the alteration in diabetes penetrance, insulitis was evident in T3A-infected mice. These results suggest that viral infection of NOD mice alters autoimmune responses to beta-cell antigens and thereby delays development of diabetes.


Assuntos
Diabetes Mellitus/imunologia , Diabetes Mellitus/fisiopatologia , Orthoreovirus Mamífero 3/patogenicidade , Pancreatopatias/fisiopatologia , Infecções por Reoviridae/complicações , Infecções por Reoviridae/imunologia , Animais , Animais Recém-Nascidos , Doenças Autoimunes/imunologia , Doenças Autoimunes/fisiopatologia , Feminino , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/fisiopatologia , Ilhotas Pancreáticas/virologia , Camundongos , Camundongos Endogâmicos NOD/virologia , Pancreatopatias/imunologia , Pancreatopatias/virologia , Infecções por Reoviridae/virologia
17.
J Virol ; 80(2): 671-81, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16378970

RESUMO

Mammalian reoviruses are internalized into cells by receptor-mediated endocytosis. Within the endocytic compartment, the viral outer capsid undergoes acid-dependent proteolysis resulting in removal of the sigma3 protein and proteolytic cleavage of the mu1/mu1C protein. Ammonium chloride (AC) is a weak base that blocks disassembly of reovirus virions by inhibiting acidification of intracellular vacuoles. To identify domains in reovirus proteins that influence pH-sensitive steps in viral disassembly, we adapted strain type 3 Dearing (T3D) to growth in murine L929 cells treated with AC. In comparison to wild-type (wt) T3D, AC-adapted (ACA-D) variant viruses exhibited increased yields in AC-treated cells. AC resistance of reassortant viruses generated from a cross of wt type 1 Lang and ACA-D variant ACA-D1 segregated with the sigma3-encoding S4 gene. The deduced sigma3 amino acid sequences of six independently derived ACA-D variants contain one or two mutations each, affecting a total of six residues. Four of these mutations, I180T, A246G, I347S, and Y354H, cluster in the virion-distal lobe of sigma3. Linkage of these mutations to AC resistance was confirmed in experiments using reovirus disassembly intermediates recoated with wt or mutant sigma3 proteins. In comparison to wt virions, ACA-D viruses displayed enhanced susceptibility to proteolysis by endocytic protease cathepsin L. Image reconstructions of cryoelectron micrographs of three ACA-D viruses that each contain a single mutation in the virion-distal lobe of sigma3 demonstrated native capsid protein organization and minimal alterations in sigma3 structure. These results suggest that mutations in sigma3 that confer resistance to inhibitors of vacuolar acidification identify a specific domain that regulates proteolytic disassembly.


Assuntos
Cloreto de Amônio/farmacologia , Proteínas do Capsídeo/genética , Reoviridae/fisiologia , Proteínas Virais/genética , Adaptação Fisiológica , Animais , Proteínas do Capsídeo/química , Catepsina L , Catepsinas/metabolismo , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Farmacorresistência Viral/genética , Células L/efeitos dos fármacos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Reoviridae/química , Reoviridae/efeitos dos fármacos , Inoculações Seriadas , Proteínas Virais/química , Proteínas Virais Reguladoras e Acessórias , Montagem de Vírus
18.
J Clin Invest ; 115(9): 2341-50, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16100570

RESUMO

Reovirus induces apoptosis in cultured cells and in vivo. In cell culture models, apoptosis is contingent upon a mechanism involving reovirus-induced activation of transcription factor NF-kappaB complexes containing p50 and p65/RelA subunits. To explore the in vivo role of NF-kappaB in this process, we tested the capacity of reovirus to induce apoptosis in mice lacking a functional nfkb1/p50 gene. The genetic defect had no apparent effect on reovirus replication in the intestine or dissemination to secondary sites of infection. In comparison to what was observed in wild-type controls, apoptosis was significantly diminished in the CNS of p50-null mice following reovirus infection. In sharp contrast, the loss of p50 was associated with massive reovirus-induced apoptosis and uncontrolled reovirus replication in the heart. Levels of IFN-beta mRNA were markedly increased in the hearts of wild-type animals but not p50-null animals infected with reovirus. Treatment of p50-null mice with IFN-beta substantially diminished reovirus replication and apoptosis, which suggests that IFN-beta induction by NF-kappaB protects against reovirus-induced myocarditis. These findings reveal an organ-specific role for NF-kappaB in the regulation of reovirus-induced apoptosis, which modulates encephalitis and myocarditis associated with reovirus infection.


Assuntos
Apoptose/fisiologia , Subunidade p50 de NF-kappa B/metabolismo , Infecções por Reoviridae , Reoviridae/fisiologia , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Encéfalo/metabolismo , Encéfalo/virologia , Linhagem Celular , Coração/virologia , Marcação In Situ das Extremidades Cortadas , Interferon beta/genética , Interferon beta/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/citologia , Intestinos/virologia , Camundongos , Camundongos Knockout , Miocardite/patologia , Miocardite/virologia , Miocárdio/citologia , Miocárdio/metabolismo , Miocárdio/patologia , Subunidade p50 de NF-kappa B/genética , Reoviridae/genética , Infecções por Reoviridae/metabolismo , Infecções por Reoviridae/patologia , Replicação Viral
19.
J Virol ; 79(13): 7967-78, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15956543

RESUMO

Reovirus infections are initiated by the binding of viral attachment protein sigma1 to receptors on the surface of host cells. The sigma1 protein is an elongated fiber comprised of an N-terminal tail that inserts into the virion and a C-terminal head that extends from the virion surface. The prototype reovirus strains type 1 Lang/53 (T1L/53) and type 3 Dearing/55 (T3D/55) use junctional adhesion molecule A (JAM-A) as a receptor. The C-terminal half of the T3D/55 sigma1 protein interacts directly with JAM-A, but the determinants of receptor-binding specificity have not been identified. In this study, we investigated whether JAM-A also mediates the attachment of the prototype reovirus strain type 2 Jones/55 (T2J/55) and a panel of field-isolate strains representing each of the three serotypes. Antibodies specific for JAM-A were capable of inhibiting infections of HeLa cells by T1L/53, T2J/55, and T3D/55, demonstrating that strains of all three serotypes use JAM-A as a receptor. To corroborate these findings, we introduced JAM-A or the structurally related JAM family members JAM-B and JAM-C into Chinese hamster ovary cells, which are poorly permissive for reovirus infection. Both prototype and field-isolate reovirus strains were capable of infecting cells transfected with JAM-A but not those transfected with JAM-B or JAM-C. A sequence analysis of the sigma1-encoding S1 gene segment of the strains chosen for study revealed little conservation in the deduced sigma1 amino acid sequences among the three serotypes. This contrasts markedly with the observed sequence variability within each serotype, which is confined to a small number of amino acids. Mapping of these residues onto the crystal structure of sigma1 identified regions of conservation and variability, suggesting a likely mode of JAM-A binding via a conserved surface at the base of the sigma1 head domain.


Assuntos
Moléculas de Adesão Celular/fisiologia , Receptores Virais/fisiologia , Reoviridae/isolamento & purificação , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Cricetinae , Primers do DNA , Genes Virais , Células HeLa , Humanos , Moléculas de Adesão Juncional , Células L , Mamíferos , Camundongos , Dados de Sequência Molecular , Reoviridae/classificação , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Junções Íntimas/virologia
20.
J Exp Med ; 200(2): 235-45, 2004 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-15263030

RESUMO

We explored the role of Peyer's patch (PP) dendritic cell (DC) populations in the induction of immune responses to reovirus strain type 1 Lang (T1L). Immunofluorescence staining revealed the presence of T1L structural (sigma1) and nonstructural (sigmaNS) proteins in PPs of T1L-infected mice. Cells in the follicle-associated epithelium contained both sigma1 and sigmaNS, indicating productive viral replication. In contrast, sigma1, but not sigmaNS, was detected in the subepithelial dome (SED) in association with CD11c(+)/CD8alpha(-)/CD11b(lo) DCs, suggesting antigen uptake by these DCs in the absence of infection. Consistent with this possibility, PP DCs purified from infected mice contained sigma1, but not sigmaNS, and PP DCs from uninfected mice could not be productively infected in vitro. Furthermore, sigma1 protein in the SED was associated with fragmented DNA by terminal deoxy-UTP nick-end labeling staining, activated caspase-3, and the epithelial cell protein cytokeratin, suggesting that DCs capture T1L antigen from infected apoptotic epithelial cells. Finally, PP DCs from infected mice activated T1L-primed CD4(+) T cells in vitro. These studies show that CD8alpha(-)/CD11b(lo) DCs in the PP SED process T1L antigen from infected apoptotic epithelial cells for presentation to CD4(+) T cells, and therefore demonstrate the cross-presentation of virally infected cells by DCs in vivo during a natural viral infection.


Assuntos
Antígenos Virais/metabolismo , Apoptose , Células Dendríticas/virologia , Células Epiteliais/virologia , Intestinos/virologia , Orthoreovirus de Mamíferos/metabolismo , Nódulos Linfáticos Agregados/virologia , Animais , Antígeno CD11b/biossíntese , Antígeno CD11c/biossíntese , Linfócitos T CD4-Positivos/metabolismo , Antígenos CD8/biossíntese , Divisão Celular , Linhagem Celular , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Células Epiteliais/metabolismo , Feminino , Imunidade nas Mucosas , Mucosa Intestinal/metabolismo , Queratinas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Microscopia de Fluorescência , Orthoreovirus de Mamíferos/genética , Nódulos Linfáticos Agregados/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...