Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Stem Cell Res Ther ; 9(1): 17, 2018 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-29378639

RESUMO

BACKGROUND: Mesenchymal stem (stromal) cells (MSCs) mediate their immunoregulatory and tissue repair functions by secreting paracrine factors, including extracellular vesicles (EVs). In several animal models of human diseases, MSC-EVs mimic the beneficial effects of MSCs. Influenza viruses cause annual outbreaks of acute respiratory illness resulting in significant mortality and morbidity. Influenza viruses constantly evolve, thus generating drug-resistant strains and rendering current vaccines less effective against the newly generated strains. Therefore, new therapies that can control virus replication and the inflammatory response of the host are needed. The objective of this study was to examine if MSC-EV treatment can attenuate influenza virus-induced acute lung injury in a preclinical model. METHODS: We isolated EVs from swine bone marrow-derived MSCs. Morphology of MSC-EVs was determined by electron microscopy and expression of mesenchymal markers was examined by flow cytometry. Next, we examined the anti-influenza activity of MSC-EVs in vitro in lung epithelial cells and anti-viral and immunomodulatory properties in vivo in a pig model of influenza virus. RESULTS: MSC-EVs were isolated from MSC-conditioned medium by ultracentrifugation. MSC-EVs were round-shaped and, similarly to MSCs, expressed mesenchymal markers and lacked the expression of swine leukocyte antigens I and II. Incubation of PKH-26-labeled EVs with lung epithelial cells revealed that MSC-EVs incorporated into the epithelial cells. Next, we examined the anti-influenza and anti-inflammatory properties of MSC-EVs. MSC-EVs inhibited the hemagglutination activity of avian, swine, and human influenza viruses at concentrations of 1.25-5 µg/ml. MSC-EVs inhibited influenza virus replication and virus-induced apoptosis in lung epithelial cells. The anti-influenza activity of MSC-EVs was due to transfer of RNAs from EVs to epithelial cells since pre-incubation of MSC-EVs with RNase enzyme abrogated the anti-influenza activity of MSC-EVs. In a pig model of influenza virus, intratracheal administration of MSC-EVs 12 h after influenza virus infection significantly reduced virus shedding in the nasal swabs, influenza virus replication in the lungs, and virus-induced production of proinflammatory cytokines in the lungs of influenza-infected pigs. The histopathological findings revealed that MSC-EVs alleviated influenza virus-induced lung lesions in pigs. CONCLUSIONS: Our data demonstrated in a relevant preclinical large animal model of influenza virus that MSC-EVs possessed anti-influenza and anti-inflammatory properties and that EVs may be used as cell-free therapy for influenza in humans.


Assuntos
Lesão Pulmonar Aguda/terapia , Vesículas Extracelulares/transplante , Células-Tronco Mesenquimais/metabolismo , Infecções por Orthomyxoviridae/patologia , Mucosa Respiratória/patologia , Lesão Pulmonar Aguda/virologia , Animais , Anti-Inflamatórios/metabolismo , Apoptose , Terapia Baseada em Transplante de Células e Tecidos/métodos , Citocinas/metabolismo , Modelos Animais de Doenças , Células Epiteliais/virologia , Alphainfluenzavirus/metabolismo , Infecções por Orthomyxoviridae/virologia , RNA Viral/genética , Mucosa Respiratória/citologia , Mucosa Respiratória/virologia , Suínos , Replicação Viral
2.
J Cell Physiol ; 233(7): 5447-5457, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29231967

RESUMO

In this study, we isolated mesenchymal stromal (stem) cells (MSCs) from broncho-alveolar lavage fluid (BAL) of 2-6-week-old commercial pigs. BAL-MSCs displayed fibroblastic morphology and possessed self-renewal properties. Similar to bone-marrow MSCs, BAL-MSCs expressed mesenchymal markers and both cell types lacked the expression of hematopoetic markers. BAL-MSCs, when cultured in differentiation induction media, differentiated into adipocytes, osteocytes, and chondrocytes. Next, we examined if BAL-MSCs have the ability to treat lipopolysaccharide (LPS)-induced acute lung injury (ALI) in a pig model. Five-week-old commercial pigs were inoculated intra-tracheally with E. coli LPS (1 mg/kg body weight [b.wt.]). Twelve hours after the LPS inoculation, groups of pigs were inoculated intra-tracheally with BM-MSCs or BAL-MSCs (2 × 106 cells/kg b.wt.). Forty eight hours after the cells administration pigs were euthanized and neutrophils in BAL, lung lesions, and cytokines in lung lysates, and engraftment of MSCs in lungs were examined. Engraftment of BAL-MSCs in injured lungs was significantly higher than the BM-MSCs, however, both cell types were equally effective in attenuating LPS-induced ALI as evidenced by decreased inflammation, lung lesions, and proinflammatory cytokines in the lungs of pigs treated with BAL- or BM-MSCs. These data in a preclinical large animal model suggest that BAL-MSCs may be used in clinical settings to treat ALI in humans.


Assuntos
Lesão Pulmonar Aguda/terapia , Líquido da Lavagem Broncoalveolar/citologia , Inflamação/terapia , Transplante de Células-Tronco Mesenquimais , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/patologia , Animais , Líquido da Lavagem Broncoalveolar/química , Diferenciação Celular/genética , Autorrenovação Celular/efeitos dos fármacos , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Neutrófilos/citologia , Suínos
3.
Vet Microbiol ; 186: 157-63, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-27016770

RESUMO

Pigs are considered as the source of some of the emerging human flu viruses. Inactivated swine influenza virus (SwIV) vaccine has been in use in the US swine herds, but it failed to control the flu outbreaks. The main reason has been attributed to lack of induction of strong local mucosal immunity in the respiratory tract. Invariant natural killer T (iNKT) cell is a unique T cell subset, and activation of iNKT cell using its ligand α-Galactosylceramide (α-GalCer) has been shown to potentiate the cross-protective immunity to inactivated influenza virus vaccine candidates in mice. Recently, we discovered iNKT cell in pig and demonstrated its activation using α-GalCer. In this study, we evaluated the efficacy of an inactivated H1N1 SwIV coadministered with α-GalCer intranasally against a homologous viral challenge. Our results demonstrated the potent adjuvant effects of α-GalCer in potentiating both innate and adaptive immune responses to SwIV Ags in the lungs of pigs, which resulted in reduction in the lung viral load by 3 logs compared to without adjuvant. Immunologically, in the lungs of pigs vaccinated with α-GalCer an increased virus specific IgA response, IFN-α secretion and NK cell-cytotoxicity was observed. In addition, iNKT cell-stimulation enhanced the secretion of Th1 cytokines (IFN-γ and IL-12) and reduced the production of immunosuppressive cytokines (IL-10 and TGF-ß) in the lungs of pigs⋅ In conclusion, we demonstrated for the first time iNKT cell adjuvant effects in pigs to SwIV Ags through augmenting the innate and adaptive immune responses in the respiratory tract.


Assuntos
Imunidade Adaptativa/efeitos dos fármacos , Galactosilceramidas/farmacologia , Imunidade Inata/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/veterinária , Doenças dos Suínos/imunologia , Imunidade Adaptativa/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Citocinas/imunologia , Imunidade Inata/imunologia , Imunoglobulina A/sangue , Pulmão/virologia , Células T Matadoras Naturais/química , Células T Matadoras Naturais/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/fisiopatologia , Distribuição Aleatória , Suínos , Doenças dos Suínos/fisiopatologia , Vacinas de Produtos Inativados/imunologia , Carga Viral , Aumento de Peso/efeitos dos fármacos
4.
Stem Cell Res Ther ; 6: 222, 2015 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-26560714

RESUMO

INTRODUCTION: Mesenchymal stem (stromal) cells (MSCs) possess self-renewal, differentiation and immunoregulatory properties, and therefore are being evaluated as cellular therapy for inflammatory and autoimmune diseases, and for tissue repair. MSCs isolated from bone marrow are extensively studied. Besides bone marrow, MSCs have been identified in almost all organs of the body including the lungs. Lung-derived MSCs may be more effective as therapy for lung diseases as compared to bone marrow-derived MSCs. Pigs are similar to humans in anatomy, physiology and immunological responses, and thus may serve as a useful large animal preclinical model to study potential cellular therapy for human diseases. METHODS: We isolated MSCs from the lungs (L-MSCs) of 4-6-week-old germ-free pigs. We determined the self-renewal, proliferation and differentiation potential of L-MSCs. We also examined the mechanisms of immunoregulation by porcine L-MSCs. RESULTS: MSCs isolated from porcine lungs showed spindle-shaped morphology and proliferated actively in culture. Porcine L-MSCs expressed mesenchymal markers CD29, CD44, CD90 and CD105 and lacked the expression of hematopoietic markers CD34 and CD45. These cells were multipotent and differentiated into adipocytes, osteocytes and epithelial cells. Like human MSCs, L-MSCs possessed immunoregulatory properties and inhibited proliferation of T cells and interferon-γ and tumor necrosis factor-α production by T cells and dendritic cells, respectively, and increased the production of T-helper 2 cytokines interleukin (IL)-4 and IL-13 by T cells. L-MSCs induced the production of prostaglandin E2 (PGE2) in MSC-T cell co-cultures and inhibition of PGE2 significantly restored (not completely) the immune modulatory effects of L-MSCs. CONCLUSIONS: Here, we demonstrate that MSCs can be isolated from porcine lung and that these cells, similar to human lung MSCs, possess in vitro proliferation, differentiation and immunomodulatory functions. Thus, these cells may serve as a model system to evaluate the contribution of lung MSCs in modulating the immune response, interactions with resident epithelial cells and tissue repair in a pig model of human lung diseases.


Assuntos
Diferenciação Celular , Imunomodulação , Pulmão/citologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Animais , Antígenos de Diferenciação/biossíntese , Proliferação de Células , Separação Celular , Células Dendríticas/metabolismo , Dinoprostona/imunologia , Feminino , Leucócitos Mononucleares/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Pluripotentes/metabolismo , Suínos
6.
Virulence ; 6(1): 40-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25517546

RESUMO

In this study, we describe the isolation and characterization of epithelial stem-like cells from the swine umbilical cord and their susceptibility to influenza virus infection. Swine umbilical cord epithelial stem cells (SUCECs) expressed stem cell and pluripotency associated markers such as SSEA-1, SSEA-4, TRA 1-60 and TRA 1-81 and Oct4. Morphologically, cells displayed polygonal morphology and were found to express epithelial markers; pancytokeratin, cytokeratin-18 and occludin; mesenchymal cell markers CD44, CD90 and haematopoietic cell marker CD45 were not detected on these cells. The cells had extensive proliferation and self- renewal properties. The cells also possessed immunomodulatory activity and inhibited the proliferation of T cells. Also, higher levels of anti-inflammatory cytokine IL-10 were detected in SUCEC-T cell co-cultures. The cells were multipotent and differentiated into lung epithelial cells when cultured in epithelial differentiation media. We also examined if SUCECs are susceptible to infection with influenza virus. SUCECs expressed sialic acid receptors, used by influenza virus for binding to cells. The 2009 pandemic influenza virus and swine influenza virus replicated in these cells. SUCECs due to their differentiation and immunoregulatory properties will be useful as cellular therapy in a pig model for human diseases. Additionally, our data indicate that influenza virus can infect SUCECs and may transmit influenza virus from mother to fetus through umbilical cord and transplantation of influenza virus-infected stem cells may transmit infection to recipients. Therefore, we propose that umbilical cord cells, in addition to other agents, should also be tested for influenza virus before cryopreservation for future use as a cell therapy for disease conditions.


Assuntos
Células Epiteliais/virologia , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Células-Tronco/virologia , Cordão Umbilical/citologia , Animais , Antígenos de Superfície/metabolismo , Biomarcadores/metabolismo , Biomarcadores Tumorais/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Células Epiteliais/citologia , Interleucina-10/metabolismo , Antígenos CD15/metabolismo , Infecções por Orthomyxoviridae , Mucosa Respiratória/citologia , Antígenos Embrionários Estágio-Específicos/metabolismo , Células-Tronco/citologia , Suínos , Cordão Umbilical/virologia
7.
Vaccine ; 33(4): 542-8, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25437101

RESUMO

Swine influenza is widely prevalent in swine herds in North America and Europe causing enormous economic losses and a public health threat. Pigs can be infected by both avian and mammalian influenza viruses and are sources of generation of reassortant influenza viruses capable of causing pandemics in humans. Current commercial vaccines provide satisfactory immunity against homologous viruses; however, protection against heterologous viruses is not adequate. In this study, we evaluated the protective efficacy of an intranasal Poly I:C adjuvanted UV inactivated bivalent swine influenza vaccine consisting of Swine/OH/24366/07 H1N1 and Swine/CO/99 H3N2, referred as PAV, in maternal antibody positive pigs against an antigenic variant and a heterologous swine influenza virus challenge. Groups of three-week-old commercial-grade pigs were immunized intranasally with PAV or a commercial vaccine (CV) twice at 2 weeks intervals. Three weeks after the second immunization, pigs were challenged with the antigenic variant Swine/MN/08 H1N1 (MN08) and the heterologous Swine/NC/10 H1N2 (NC10) influenza virus. Antibodies in serum and respiratory tract, lung lesions, virus shedding in nasal secretions and virus load in lungs were assessed. Intranasal administration of PAV induced challenge viruses specific-hemagglutination inhibition- and IgG antibodies in the serum and IgA and IgG antibodies in the respiratory tract. Importantly, intranasal administration of PAV provided protection against the antigenic variant MN08 and the heterologous NC10 swine influenza viruses as evidenced by significant reductions in lung virus load, gross lung lesions and significantly reduced shedding of challenge viruses in nasal secretions. These results indicate that Poly I:C or its homologues may be effective as vaccine adjuvants capable of generating cross-protective immunity against antigenic variants/heterologous swine influenza viruses in pigs.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Poli I-C/administração & dosagem , Doenças dos Suínos/prevenção & controle , Vacinação/métodos , Administração Intranasal , Animais , Anticorpos Antivirais/análise , Anticorpos Antivirais/sangue , Líquidos Corporais/imunologia , Europa (Continente) , Imunoglobulina A/análise , Imunoglobulina G/análise , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H2N2/imunologia , Pulmão/patologia , Pulmão/virologia , América do Norte , Sistema Respiratório/imunologia , Suínos , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia , Carga Viral , Eliminação de Partículas Virais
8.
PLoS One ; 9(3): e90066, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24594847

RESUMO

Influenza A virus (IAV) and Streptococcus pneumoniae (pneumococcus) are two major upper respiratory tract pathogens responsible for exacerbated disease in coinfected individuals. Despite several studies showing increased susceptibility to secondary bacterial infections following IAV infection, information on the direct effect of S. pneumoniae on IAV in vitro is unknown. This is an important area of investigation as S. pneumoniae is a common commensal of the human upper respiratory tract, present as an important coinfecting pathogen with IAV infection. A recent study showed that S. pneumoniae enhances human metapneumovirus infection in polarized bronchial epithelial cells in vitro. The aim of the current study was to determine whether treatment of epithelial cells with S. pneumoniae affects IAV replication using a standard immunofluorescence assay (IFA). For this study we used four IAV permissive epithelial cell lines including two human-derived cell lines, 12 pneumococcal strains including recent human clinical isolates which represent different genetic backgrounds and serotypes, and six IAV strains of varying genetic nature and pathogenic potential including the pandemic 2009 H1N1 virus. Our results suggested that pretreatment of MDCK cells with 7.5×10(6) colony-forming units (CFUs) of live S. pneumoniae resulted in gradual cell-death in a time-dependent manner (0.5 to 4 hr). But, pretreatment of cell lines with 7.5×10(5) and lower CFUs of S. pneumoniae had no detectable effect on either the morphology of cells or on the IAV replication. However, unlike in epithelial cell lines, due to influence of secreted host factors the effect of pneumococci on IAV replication may be different during coinfections in vivo in the human upper respiratory tract, and in vitro with primary human polarized bronchial epithelial cells.


Assuntos
Células Epiteliais/microbiologia , Vírus da Influenza A/fisiologia , Streptococcus pneumoniae/fisiologia , Replicação Viral , Animais , Calibragem , Cães , Técnicas In Vitro , Células Madin Darby de Rim Canino , Microscopia de Fluorescência , Streptococcus pneumoniae/classificação
9.
Microbiologyopen ; 3(2): 168-81, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24515965

RESUMO

The methylmenaquinol:fumarate reductase (Mfr) of Campylobacter jejuni is a periplasmic respiratory (redox) protein that contributes to the metabolism of fumarate and displays homology to succinate dehydrogenase (Sdh). Since chemically oxidized redox-enzymes, including fumarate reductase and Sdh, contribute to the generation of oxidative stress in Escherichia coli, we assessed the role of Mfr in C. jejuni after exposure to hydrogen peroxide (H2 O2 ). Our results show that a Mfr mutant (∆mfrA) strain was less susceptible to H2 O2 as compared to the wildtype (WT). Furthermore, the H2 O2 concentration in the ∆mfrA cultures was significantly higher than that of WT after exposure to the oxidant. In the presence of H2 O2 , catalase (KatA) activity and katA expression were significantly lower in the ∆mfrA strain as compared to the WT. Exposure to H2 O2 resulted in a significant decrease in total intracellular iron in the ∆mfrA strain as compared to WT, while the addition of iron to the growth medium mitigated H2 O2 susceptibility and accumulation in the mutant. The ∆mfrA strain was significantly more persistent in RAW macrophages as compared to the WT. Scanning electron microscopy showed that infection with the ∆mfrA strain caused prolonged changes to the macrophages' morphology, mainly resulting in spherical-shaped cells replete with budding structures and craters. Collectively, our results suggest a role for Mfr in maintaining iron homeostasis in H2 O2 stressed C. jejuni, probably via affecting the concentrations of intracellular iron.


Assuntos
Campylobacter jejuni/efeitos dos fármacos , Campylobacter jejuni/enzimologia , Peróxido de Hidrogênio/toxicidade , Ferro/metabolismo , Succinato Desidrogenase/metabolismo , Animais , Campylobacter jejuni/genética , Linhagem Celular , Deleção de Genes , Macrófagos/citologia , Macrófagos/microbiologia , Camundongos , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Microscopia Eletrônica de Varredura , Succinato Desidrogenase/genética
10.
Cell Transplant ; 22(3): 461-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23006541

RESUMO

Mesenchymal stromal cells (MSCs) have differentiation, immunomodulatory, and self-renewal properties and are, therefore, an attractive tool for regenerative medicine and autoimmune diseases. MSCs may be of great value to treat graft-versus-host disease. Influenza virus causes highly contagious seasonal infection and occasional pandemics. The infection is severe in children, elderly, and immunocompromised hosts including hematopoietic stem cell transplant patients. The objective of this study was to determine if MSCs are permissive to influenza virus replication. We isolated MSCs from the bone marrow of 4- to 6-week-old germ-free pigs. Swine and human influenza virus strains were used to infect MSCs in vitro. MSCs expressed known influenza virus α-2,3 and α-2,6 sialic acid receptors and supported replication of swine and human influenza viruses. Viral infection of MSCs resulted in cell lysis and proinflammatory cytokine production. These findings demonstrate that bone marrow-derived MSCs are susceptible to influenza virus. The data also suggest that transplantation of bone marrow MSCs from influenza virus-infected donors may transmit infection to recipients. Also, MSCs may get infected if infused into a patient with an ongoing influenza virus infection.


Assuntos
Células da Medula Óssea/citologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Células-Tronco Mesenquimais/citologia , Animais , Diferenciação Celular , Células Cultivadas , Humanos , Interleucina-6/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/virologia , Receptores de Superfície Celular/metabolismo , Suínos , Fator de Necrose Tumoral alfa/metabolismo
11.
BMC Microbiol ; 12: 258, 2012 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-23148765

RESUMO

BACKGROUND: The genetic features that facilitate Campylobacter jejuni's adaptation to a wide range of environments are not completely defined. However, whole genome expression studies showed that respiratory proteins (RPs) were differentially expressed under varying conditions and stresses, suggesting further unidentified roles for RPs in C. jejuni's adaptation. Therefore, our objectives were to characterize the contributions of selected RPs to C. jejuni's i- key survival phenotypes under different temperature (37°C vs. 42°C) and oxygen (microaerobic, ambient, and oxygen-limited/anaerobic) conditions and ii- its interactions with intestinal epithelial cells from disparate hosts (human vs. chickens). RESULTS: C. jejuni mutant strains with individual deletions that targeted five RPs; nitrate reductase (ΔnapA), nitrite reductase (ΔnrfA), formate dehydrogenase (ΔfdhA), hydrogenase (ΔhydB), and methylmenaquinol:fumarate reductase (ΔmfrA) were used in this study. We show that only the ΔfdhA exhibited a decrease in motility; however, incubation at 42°C significantly reduced the deficiency in the ΔfdhA's motility as compared to 37°C. Under all tested conditions, the ΔmfrA showed a decreased susceptibility to hydrogen peroxide (H(2)O(2)), while the ΔnapA and the ΔfdhA showed significantly increased susceptibility to the oxidant as compared to the wildtype. Further, the susceptibility of the ΔnapA to H(2)O(2) was significantly more pronounced at 37°C. The biofilm formation capability of individual RP mutants varied as compared to the wildtype. However, the impact of the deletion of certain RPs affected biofilm formation in a manner that was dependent on temperature and/or oxygen concentration. For example, the ΔmfrA displayed significantly deficient and increased biofilm formation under microaerobic conditions at 37°C and 42°C, respectively. However, under anaerobic conditions, the ΔmfrA was only significantly impaired in biofilm formation at 42°C. Additionally, the RPs mutants showed differential ability for infecting and surviving in human intestinal cell lines (INT-407) and primary chicken intestinal epithelial cells, respectively. Notably, the ΔfdhA and the ΔhydB were deficient in interacting with both cell types, while the ΔmfrA displayed impairments only in adherence to and invasion of INT-407. Scanning electron microscopy showed that the ΔhydB and the ΔfdhA exhibited filamentous and bulging (almost spherical) cell shapes, respectively, which might be indicative of defects in cell division. CONCLUSIONS: We conclude that the RPs contribute to C. jejuni's motility, H(2)O(2) resistance, biofilm formation, and in vitro interactions with hosts' intestinal cells. Further, the impact of certain RPs varied in response to incubation temperature and/or oxygen concentration. Therefore, RPs may facilitate the prevalence of C. jejuni in a variety of niches, contributing to the pathogen's remarkable potential for adaptation.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Campylobacter jejuni/fisiologia , Células Epiteliais/microbiologia , Viabilidade Microbiana , Oxirredutases/metabolismo , Fatores de Virulência/metabolismo , Animais , Biofilmes/crescimento & desenvolvimento , Campylobacter jejuni/efeitos dos fármacos , Campylobacter jejuni/patogenicidade , Campylobacter jejuni/efeitos da radiação , Galinhas , Deleção de Genes , Humanos , Peróxido de Hidrogênio/toxicidade , Locomoção , Microscopia Eletrônica de Varredura , Oxirredutases/genética , Temperatura
12.
J Virol ; 86(12): 6427-33, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22491467

RESUMO

We isolated stem/progenitor epithelial cells from the lungs of 4- to 6-week-old pigs. The epithelial progenitor colony cells were surrounded by mesenchymal stromal cells. The progenitor epithelial colony cells expressed stem cell markers such as octamer binding transcription factor 4 (Oct4) and stage-specific embryonic antigen 1 (SSEA-1), as well as the epithelial markers pancytokeratin, cytokeratin-18, and occludin, but not mesenchymal (CD44, CD29, and CD90) and hematopoietic (CD45) markers. The colony cells had extensive self-renewal potential and had the capacity to undergo differentiation to alveolar type I- and type II-like pneumocytes. Additionally, these cells expressed sialic acid receptors and supported the active replication of influenza virus, which was accompanied by cell lysis. The lysis of progenitor epithelial cells by influenza virus may cause a marked reduction in the potential of progenitor cells for self renewal and for their ability to differentiate into specialized cells of the lung. These observations suggest the possible involvement of lung stem/progenitor cells in influenza virus infection.


Assuntos
Células Epiteliais/metabolismo , Vírus da Influenza A/fisiologia , Influenza Humana/virologia , Pulmão/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco/metabolismo , Replicação Viral , Animais , Diferenciação Celular , Células Epiteliais/citologia , Células Epiteliais/virologia , Humanos , Vírus da Influenza A/genética , Influenza Humana/genética , Influenza Humana/metabolismo , Pulmão/citologia , Pulmão/virologia , Fator 3 de Transcrição de Octâmero/genética , Células-Tronco/citologia , Células-Tronco/virologia , Suínos
13.
Vet Microbiol ; 158(1-2): 60-8, 2012 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-22397932

RESUMO

In October and November 2010, novel H1N2 reassortant influenza viruses were identified from pigs showing mild respiratory signs that included cough and depression. Sequence and phylogenetic analysis showed that the novel H1N2 reassortants possesses HA and NA genes derived from recent H1N2 swine isolates similar to those isolated from Midwest. Compared to the majority of reported reassortants, both viruses preserved human-like host restrictive and putative antigenic sites in their HA and NA genes. The four internal genes, PB2, PB1, PA, and NS were similar to the contemporary swine triple reassortant viruses' internal genes (TRIG). Interestingly, NP and M genes of the novel reassortants were derived from the 2009 pandemic H1N1. The NP and M proteins of the two isolates demonstrated one (E16G) and four (G34A, D53E, I109T, and V313I) amino acid changes in the M2 and NP proteins, respectively. Similar amino acid changes were also noticed upon incorporation of the 2009 pandemic H1N1 NP in other reassortant viruses reported in the U.S. Thus the role of those amino acids in relation to host adaptation need to be further investigated. The reassortments of pandemic H1N1 with swine influenza viruses and the potential of interspecies transmission of these reassortants from swine to other species including human indicate the importance of systematic surveillance of swine population to determine the origin, the prevalence of similar reassortants in the U.S. and their impact on both swine production and public health.


Assuntos
Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Vírus da Influenza A Subtipo H1N2/isolamento & purificação , Infecções por Orthomyxoviridae/veterinária , Vírus Reordenados/isolamento & purificação , Doenças dos Suínos/virologia , Animais , Cães , Humanos , Vírus da Influenza A Subtipo H1N1/classificação , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N2/classificação , Vírus da Influenza A Subtipo H1N2/genética , Influenza Humana/transmissão , Influenza Humana/virologia , Células Madin Darby de Rim Canino , Ohio/epidemiologia , Infecções por Orthomyxoviridae/epidemiologia , Infecções por Orthomyxoviridae/virologia , Filogenia , Vírus Reordenados/classificação , Vírus Reordenados/genética , Sus scrofa , Suínos , Doenças dos Suínos/epidemiologia
14.
Results Immunol ; 2: 112-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-24371574

RESUMO

Infectious bursal disease (IBD) is a highly contagious disease of chickens which leads to immunosuppression. In our previous study it was demonstrated that, possibly, CD4(+) and CD8(+) T cells may employ perforin and granzyme-A pathway for the clearance of IBDV-infected bursal cells. In this study, we evaluated the cytotoxic T cell responses involving two independently functioning but complementary mechanisms: Fas-Fas ligand and perforin-granzyme pathways in IBDV-infected chickens. As demonstrated previously, infection of chickens with IBDV was accompanied by influx of CD8(+) T cells in the bursa and spleen. There was an upregulation in the gene expression of cytolytic molecules: Fas and Fas ligand (FasL), perforin (PFN) and granzyme-A (Gzm-A) in bursal and in the splenic tissues of IBDV inoculated chickens. Additionally, for the first time, we detected Fas, Fas ligand, Caspase-3 and PFN producing CD8(+) T cells in the bursa and spleen of IBDV-infected chickens. The infiltration and activation of CD8(+) T cells was substantiated by the detection of Th1 cytokine, IFN-γ. These data suggest that T cells may be involved in the clearance of virus from the target organ bursa and peripheral tissues such as spleen. The findings of these studies provide new insights into the pathogenesis of IBD and provide mechanistic evidence that the cytotoxic T cells may act through both Fas-FasL and perforin-granzyme pathways in mediating the clearance of virus-infected cells.

15.
Vet Res ; 42: 85, 2011 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-21749706

RESUMO

Infectious bursal disease (IBD) is an important immunosuppressive disease of chickens. The causative agent, infectious bursal disease virus (IBDV), consists of two serotypes, 1 and 2. Serotype 1 consists of classic IBDV (cIBDV) and variant IBDV (vIBDV). Both of these strains vary in antigenicity and pathogenesis. The goal of this study was to compare the immunopathogenesis of cIBDV and vIBDV. Three-week-old specific pathogen free chickens were inoculated intraocularly with standard challenge strain (STC) (cIBDV) and a variant strain Indiana (IN) (vIBDV). The cIBDV produced more pronounced bursal damage, inflammatory response and infiltration of T cells as compared to vIBDV. There were significant differences in the expression of innate (IFN-α and IFN-ß), proinflammatory cytokine and mediator (IL-6 and iNOS) in cIBDV- and vIBDV-infected bursas. The expression of chemokines genes, IL-8 and MIP-α was also higher in cIBDV-infected chickens during the early phase of infection. The expression of Toll like receptor 3 (TLR3) was downregulated at post inoculation days (PIDs) 3, 5, and 7 in the bursas of vIBDV-infected chickens whereas TLR3 was upregulated at PIDs 3 and 5 in cIBDV-infected bursas. In vIBDV-infected bursa, TLR7 expression was downregulated at PIDs 3 and 5 and upregulated at PID 7. However, TLR7 was upregulated at PIDs 3 and 7 in cIBDV-infected bursas. The expression of MyD88 was downregulated whereas TRIF gene expression was upregulated in cIBDV- and vIBDV-infected bursa. These findings demonstrate the critical differences in bursal lesions, infiltration of T cells, expression of cytokines, chemokines and TLRs in the bursa of cIBDV-and vIBDV-infected chickens.


Assuntos
Proteínas Aviárias/genética , Infecções por Birnaviridae/veterinária , Galinhas , Citocinas/genética , Regulação da Expressão Gênica , Vírus da Doença Infecciosa da Bursa/fisiologia , Doenças das Aves Domésticas/imunologia , Receptores Toll-Like/genética , Animais , Antígenos Virais/metabolismo , Proteínas Aviárias/metabolismo , Infecções por Birnaviridae/genética , Infecções por Birnaviridae/imunologia , Infecções por Birnaviridae/virologia , Bolsa de Fabricius/imunologia , Bolsa de Fabricius/patologia , Bolsa de Fabricius/virologia , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/metabolismo , Vírus da Doença Infecciosa da Bursa/genética , Vírus da Doença Infecciosa da Bursa/imunologia , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/virologia , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Receptores Toll-Like/metabolismo
16.
Influenza Other Respir Viruses ; 5(4): 268-75, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21651737

RESUMO

BACKGROUND: European starlings (Sturnus vulgaris) are common, widely distributed birds in North America that frequently come into contact with agricultural operations. However, starlings have been one of the neglected land-based wild bird species for influenza surveillance. OBJECTIVES: To study the potential role of starlings in the ecology and epidemiology of influenza virus. METHODS: We collected 328 digestive and 156 tracheal samples from starlings in Ohio in years 2007 (July) to 2008 (August) and screened for the presence of influenza virus by real-time RT-PCR, standard RT-PCR and virus isolation using embryonated chicken eggs. In addition, we conducted an experimental infection study to evaluate the replication and induction of antibody response by two low pathogenic avian influenza (AI) viruses in starlings. RESULTS: Although virus isolation was negative, we confirmed 21 influenza positive digestive and tracheal samples by real-time and standard RT-PCR tests. Phylogenetic analysis revealed that five NS genes recovered from Starlings belonged to NS subtype A and were most similar to the NS genes from a wild aquatic bird origin isolate from Ohio. Experimental infection studies using two low pathogenic AI strains showed that starlings could be infected, shed virus, and seroconvert. CONCLUSIONS: This study shows that starlings can carry influenza virus that is genetically similar to wild aquatic bird origin strains and may serve as a carrier of influenza virus to domestic animals.


Assuntos
Genes Virais , Vírus da Influenza A/isolamento & purificação , Influenza Aviária/virologia , RNA Viral/isolamento & purificação , Estorninhos/virologia , Animais , Embrião de Galinha , Análise por Conglomerados , Sistema Digestório/virologia , Vírus da Influenza A/genética , Dados de Sequência Molecular , Ohio , Filogenia , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Traqueia/virologia , Proteínas não Estruturais Virais/genética , Cultura de Vírus
17.
Dev Comp Immunol ; 35(5): 620-7, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21241730

RESUMO

Infectious bursal disease (IBD) is an economically important immunosuppressive disease of chickens. The IBD virus (IBDV) actively replicates in B cells and causes severe bursal damage. Generally, T cells are refractory to infection with IBDV but are known to promote virus clearance. However, the mechanisms of T cell mediated viral clearance are not well understood. In this study, we evaluated the molecular mechanisms of cytotoxic T cell responses in the pathogenesis of IBD in chickens. Infection of chickens with IBDV was accompanied by the infiltration of CD4(+) and CD8(+) T cells in the bursa. There was an upregulation in the gene expression of important cytolytic molecules; perforin (PFN), granzyme-A (Gzm-A), DNA repair and apoptotic proteins; high mobility proteins group (HMG) and poly (ADP-ribose) polymerase (PARP) in the bursa of Fabricius (BF) whereas expression of NK (natural killer) lysin was downregulated. Importantly, PFN producing CD4(+) and CD8(+) T cells were also detected in the bursa of IBDV-infected chickens by immunohistochemistry. The Th1 cytokines, IL-2 and IFN-γ expression was also strongly upregulated, suggesting the activation of T cells. The findings of this study highlight the mechanisms of IBD pathogenesis and the role of cytotoxic T cells in the clearance of virus-infected cells.


Assuntos
Infecções por Birnaviridae/veterinária , Bolsa de Fabricius/metabolismo , Galinhas/imunologia , Granzimas/imunologia , Vírus da Doença Infecciosa da Bursa , Perforina/imunologia , Doenças das Aves Domésticas/imunologia , Animais , Apoptose , Infecções por Birnaviridae/imunologia , Galinhas/genética , Galinhas/virologia , Reparo do DNA , Regulação da Expressão Gênica , Granzimas/genética , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Perforina/genética , Linfócitos T/citologia , Linfócitos T/imunologia
18.
Vet Microbiol ; 148(2-4): 175-82, 2011 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-20965669

RESUMO

The H3N2 triple reassortant (TR) influenza viruses emerged in swine in 1998 and then in turkeys in 2003. It was then hypothesized that these viruses crossed the species barrier and transmitted from pigs to turkeys. In previous work we identified viruses with different transmission behavior between the two species, of which A/turkey/Ohio/313053/04 (TK04) transmitted both ways between swine and turkeys, and A/swine/North Carolina/03 (SW03) did not transmit either way between the two species. Utilizing the 12-plasmid reverse genetics (RG) system, we rescued two viruses (TK04 and SW03) with potentially different transmission behavior between pigs and turkeys. Single gene reassortants (SGR) were generated by switching the hemagglutinin (HA) or the neuraminidase (NA) genes between both viruses, and were evaluated for replication in vitro (pig and turkey tracheal/bronchial epithelial cells) and in vivo (pigs and turkeys). RG-created TK04 replicated more efficiently than SW03 in vitro and in vivo. Additionally, TK04 exhibited better binding affinity to plasma membrane preparations (PMP) from pig and turkey tracheal/bronchial epithelial cells compared to SW03. In study with SGR viruses, the HA protein was found to be essential for TK04 virus transmission amongst turkeys, but not sole factor contributing to the efficient replication of virus in turkeys and pigs. Such findings further highlight the polygenic nature of influenza virus pathogenesis.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Influenza A Subtipo H3N2/fisiologia , Vírus Reordenados/genética , Suínos/virologia , Perus/virologia , Replicação Viral , Animais , Dissacarídeos , Eletrólitos , Células Epiteliais/virologia , Glutamatos , Glutationa , Histidina , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A Subtipo H3N2/patogenicidade , Influenza Aviária/transmissão , Influenza Aviária/virologia , Manitol , Neuraminidase/genética , Infecções por Orthomyxoviridae/transmissão , Infecções por Orthomyxoviridae/virologia , RNA Viral/genética , Doenças dos Suínos/transmissão , Doenças dos Suínos/virologia
19.
Vet Immunol Immunopathol ; 140(1-2): 30-6, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21146877

RESUMO

Commercial turkey eggs, free of antibodies to avian metapneumovirus subtype C (aMPV/C), were inoculated with aMPV/C at embryonation day (ED) 24. There was no detectable effect of virus inoculation on the hatchability of eggs. At 4 days post inoculation (DPI) (the day of hatch (ED 28)) and 9 DPI (5 days after hatch), virus replication was detected by quantitative RT-PCR in the turbinate, trachea and lung but not in the thymus or spleen. Mild histological lesions characterized by lymphoid cell infiltration were evident in the turbinate mucosa. Virus exposure inhibited the mitogenic response of splenocytes and thymocytes and upregulated gene expression of IFN-γ and IL-10 in the turbinate tissue. Turkeys hatching from virus-exposed eggs had aMPV/C-specific IgG in the serum and the lachrymal fluid. At 3 week of age, in ovo immunized turkeys were protected against a challenge with pathogenic aMPV/C.


Assuntos
Metapneumovirus/imunologia , Infecções por Paramyxoviridae/veterinária , Doenças das Aves Domésticas/imunologia , Vacinação/veterinária , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Embrião não Mamífero/imunologia , Ensaio de Imunoadsorção Enzimática/veterinária , Interferon gama/biossíntese , Interleucina-10/biossíntese , Interleucina-18/biossíntese , Metapneumovirus/isolamento & purificação , Mitógenos/imunologia , Infecções por Paramyxoviridae/sangue , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/virologia , Doenças das Aves Domésticas/sangue , Doenças das Aves Domésticas/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Perus
20.
J Clin Immunol ; 31(2): 228-39, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21042929

RESUMO

Important roles played by invariant natural killer T (iNKT) cells in asthma pathogenesis have been demonstrated. We identified functional iNKT cells and CD1d molecules in pig lungs. Pig iNKT cells cultured in the presence of α-GalCer proliferated and secreted Th1 and Th2 cytokines. Like in other animal models, direct activation of pig lung iNKT cells using α-GalCer resulted in acute airway hyperreactivity (AHR). Clinically, acute AHR-induced pigs had increased respiratory rate, enhanced mucus secretion in the airways, fever, etc. In addition, we observed petechial hemorrhages, infiltration of CD4(+) cells, and increased Th2 cytokines in AHR-induced pig lungs. Ex vivo proliferated iNKT cells of asthma induced pigs in the presence of C-glycoside analogs of α-GalCer had predominant Th2 phenotype and secreted more of Th2 cytokine, IL-4. Thus, baby pigs may serve as a useful animal model to study iNKT cell-mediated AHR caused by various environmental and microbial CD1d-specific glycolipid antigens.


Assuntos
Hiper-Reatividade Brônquica/imunologia , Pulmão/imunologia , Células T Matadoras Naturais/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Antígenos CD1d/imunologia , Asma/imunologia , Hiper-Reatividade Brônquica/patologia , Modelos Animais de Doenças , Glicosídeos , Pulmão/patologia , Ativação Linfocitária/efeitos dos fármacos , Monossacarídeos/farmacologia , Células T Matadoras Naturais/efeitos dos fármacos , Pneumonia/imunologia , Pneumonia/patologia , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...