RESUMO
Despite the successful control of highly contagious tumorigenic Marek's disease (MD) by vaccination, a continuous increase in MD virus (MDV) virulence over recent decades has put emphasis on the development of more MD-resistant chickens. The cell types and genes involved in resistance therefore need to be recognized. The virus is primarily lymphotropic, but research should also focus on innate immunity, as innate immune cells are among the first to encounter MDV. Our previous study on MDV-macrophage interaction revealed significant differences between MHC-congenic lines 61 (MD-resistant) and 72 (MD-susceptible). To investigate the role of dendritic cells (DCs) in MD resistance, bone-marrow-derived DCs from these lines were infected with MDV in vitro. They were then characterized by cell sorting, and the respective transcriptomes analysed by RNA-seq. The differential expression (DE) of genes revealed a strong immune activation in DCs of the susceptible line, although an inherent immune supremacy was shown by the resistant line, including a significant expression of tumour-suppressor miRNA, gga-mir-124a, in line 61 control birds. Enrichment analysis of DE genes revealed high expression of an oncogenic transcription factor, AP-1, in the susceptible line following MDV challenge. This research highlights genes and pathways that may play a role in DCs in determining resistance or susceptibility to MDV infection.
RESUMO
Macrophage colony-stimulating factor (CSF1) is an essential growth factor to control the proliferation, differentiation and survival of cells of the macrophage lineage in vertebrates. We have previously produced a recombinant chicken CSF1-Fc fusion protein and administrated it to birds which produced a substantial expansion of tissue macrophage populations. To further study the biology of CSF1 in the chicken, here we generated anti-chicken CSF1 antibodies (ROS-AV181 and 183) using CSF1-Fc as an immunogen. The specific binding of each monoclonal antibody was confirmed by ELISA, Western blotting and immunohistochemistry on tissue sections. Using the anti-CSF1 antibodies, we show that chicken bone marrow derived macrophages (BMDM) express CSF1 on their surface, and that the level appears to be regulated further by exogenous CSF1. By capture ELISA circulating CSF1 levels increased transiently in both layer and broiler embryos around the day of hatch. The levels of CSF1 in broilers was higher than in layers during the first week after hatch. Antibody ROS-AV183 was able to block CSF1 biological activity in vitro and treatment of hatchlings using this neutralising antibody in vivo impacted on some tissue macrophage populations, but not blood monocytes. After anti-CSF1 treatment, CSF1R-transgene reporter expressing cells were reduced in the bursa of Fabricius and cecal tonsil and TIM4+ Kupffer cells in the liver were almost completely ablated. Anti-CSF1 treatment also produced a reduction in overall bone density, trabecular volume and TRAP+ osteoclasts. Our novel neutralising antibody provides a new tool to study the roles of CSF1 in birds.
Assuntos
Anticorpos Bloqueadores/isolamento & purificação , Anticorpos/isolamento & purificação , Proteínas Aviárias/genética , Bolsa de Fabricius/metabolismo , Galinhas/imunologia , Fator Estimulador de Colônias de Macrófagos/genética , Macrófagos/fisiologia , Animais , Proteínas Aviárias/metabolismo , Diferenciação Celular , Células Cultivadas , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento , Fragmentos Fc das Imunoglobulinas/genética , Fator Estimulador de Colônias de Macrófagos/imunologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Proteínas Recombinantes de Fusão/genéticaRESUMO
Despite successful control by vaccination, Marek's disease (MD) has continued evolving to greater virulence over recent years. To control MD, selection and breeding of MD-resistant chickens might be a suitable option. MHC-congenic inbred chicken lines, 61 and 72, are highly resistant and susceptible to MD, respectively, but the cellular and genetic basis for these phenotypes is unknown. Marek's disease virus (MDV) infects macrophages, B-cells, and activated T-cells in vivo. This study investigates the cellular basis of resistance to MD in vitro with the hypothesis that resistance is determined by cells active during the innate immune response. Chicken bone marrow-derived macrophages from lines 61 and 72 were infected with MDV in vitro. Flow cytometry showed that a higher percentage of macrophages were infected in line 72 than in line 61. A transcriptomic study followed by in silico functional analysis of differentially expressed genes was then carried out between the two lines pre- and post-infection. Analysis supports the hypothesis that macrophages from susceptible and resistant chicken lines display a marked difference in their transcriptome following MDV infection. Resistance to infection, differential activation of biological pathways, and suppression of oncogenic potential are among host defense strategies identified in macrophages from resistant chickens.
Assuntos
Resistência à Doença/genética , Macrófagos/metabolismo , Doença de Marek/imunologia , Transcriptoma , Animais , Células Cultivadas , Embrião de Galinha , Doença de Marek/genética , Aves Domésticas/genética , Aves Domésticas/imunologiaRESUMO
Avian pathogenic Escherichia coli (APEC) cause severe respiratory and systemic disease in chickens, commonly termed colibacillosis. Early immune responses after initial infection are highly important for the outcome of the infection. In this study, the early interactions between GFP-expressing APEC strains of serotypes O1:K1:H7 and O2:K1:H5 and phagocytic cells in the lung of CSF1R-reporter transgenic chickens were investigated. CSF1R-reporter transgenic chickens express fluorescent protein under the control of elements of the CSF1R promoter and enhancer, such that cells of the myeloid lineage can be visualized in situ and sorted. Chickens were separately inoculated with APEC strains expressing GFP and culled 6 h post-infection. Flow cytometric analysis was performed to phenotype and sort the cells that harbored bacteria in the lung, and the response of the sorted cells was defined by transcriptomic analysis. Both APEC strains were mainly detected in CSF1R-transgeneneg (CSF1R-tgneg) and CSF1R-tglow MHC IIneg MRC1L-Bneg cells and low numbers of APEC were detected in CSF1R-tghigh MHC IIpos MRC1L-Bpos cells. Transcriptomic and flow cytometric analysis identified the APECposCSF1R-tgneg and CSF1R-tglow cells as heterophils and the APECposCSF1R-tghigh cells as macrophages and dendritic cells. Both APEC strains induced strong inflammatory responses, however in both CSF1R-tgneg/low and CSF1R-tghigh cells, many immune related pathways were repressed to a greater extent or less activated in birds inoculated with APEC O2-GFP compared to APEC O1-GFP inoculated birds. Comparison of the immune pathways revealed the aryl hydrocarbon receptor (AhR) pathway, IL17 and STAT3 signaling, heterophil recruitment pathways and the acute phase response, are modulated particularly post-APEC O2-GFP inoculation. In contrast to in vivo data, APEC O2-GFP was more invasive in CSF1R-tghigh cells in vitro than APEC O1-GFP and had higher survival rates for up to 6 h post-infection. Our data indicate significant differences in the responses induced by APEC strains of prevalent serotypes, with important implications for the design and interpretation of future studies. Moreover, we show that bacterial invasion and survival in phagocyte populations in vitro is not predictive of events in the chicken lung.
Assuntos
Galinhas/imunologia , Escherichia coli/imunologia , Granulócitos/imunologia , Imunomodulação/imunologia , Pulmão/imunologia , Macrófagos/imunologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/imunologia , Animais , Animais Geneticamente Modificados/imunologia , Animais Geneticamente Modificados/microbiologia , Galinhas/microbiologia , Infecções por Escherichia coli/imunologia , Granulócitos/microbiologia , Pulmão/microbiologia , Macrófagos/microbiologia , Fagócitos/imunologia , Fagócitos/microbiologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/microbiologia , Transdução de Sinais/imunologia , Virulência/imunologia , Fatores de Virulência/imunologiaRESUMO
Avian pathogenic E. coli (APEC) cause severe respiratory and systemic disease. To address the genetic and immunological basis of resistance, inbred chicken lines were used to establish a model of differential resistance to APEC, using strain O1 of serotype O1:K1:H7. Inbred lines 72, 15I and C.B12 and the outbred line Novogen Brown were inoculated via the airsac with a high dose (107 colony-forming units, CFU) or low dose (105 CFU) of APEC O1. Clinical signs, colibacillosis lesion score and bacterial colonization of tissues after high dose challenge were significantly higher in line 15I and C.B12 birds. The majority of the 15I and C.B12 birds succumbed to the infection by 14â h post-infection, whilst none of the line 72 and the Novogen Brown birds developed clinical signs. No difference was observed after low dose challenge. In a repeat study, inbred lines 72 and 15I were inoculated with low, intermediate or high doses of APEC O1 ranging from 105 to 107 CFU. The colonization of lung was highest in line 15I after high dose challenge and birds developed clinical signs; however, colonization of blood and spleen, clinical signs and lesion score were not different between lines. No difference was observed after intermediate or low dose challenge. Ex vivo, the phagocytic and bactericidal activity of lung leukocytes from line 72 and 15I birds did not differ. Our data suggest that although differential resistance of inbred lines 72, 15I and C.B12 to APEC O1 challenge is apparent, it is dependent on the infectious dose. Research Highlights Lines 15I and C.B12 are more susceptible than line 72 to a high dose of APEC O1. Differential resistance is dose-dependent in lines 15I and 72. Phagocytic and bactericidal activity is similar and dose independent.
Assuntos
Galinhas , Resistência à Doença , Infecções por Escherichia coli/veterinária , Escherichia coli/imunologia , Imunidade Inata , Doenças das Aves Domésticas/imunologia , Sacos Aéreos/microbiologia , Animais , Animais Endogâmicos , Anticorpos Heterófilos/imunologia , Carga Bacteriana , Relação Dose-Resposta Imunológica , Escherichia coli/genética , Escherichia coli/patogenicidade , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Feminino , Macrófagos/imunologia , Masculino , Doenças das Aves Domésticas/microbiologia , Organismos Livres de Patógenos EspecíficosRESUMO
The respiratory tract is a key organ for many avian pathogens as well as a major route for vaccination in the poultry industry. To improve immune responses after vaccination of chickens through increased uptake of vaccines and targeting to antigen presenting cells, a better understanding of the avian respiratory immune system is required. Transgenic MacReporter birds were used expressing a reporter gene (eGFP or mApple) under the control of the CSF1R promoter and enhancer in cells of the mononuclear phagocyte (MNP) lineage to visualize the ontogeny of the lymphoid tissue, macrophages and dendritic cells, in the trachea, lung and air sac of birds from embryonic day 18-63 weeks of age. Small aggregates of CSF1R-transgene+ cells start to form at the openings of the secondary bronchi at 1 week of age, indicative of the early development of the organised bronchus-associated lymphoid tissue. Immunohistochemical staining revealed subpopulations of MNPs in the lung, based on expression of CSF1R-transgene, CD11, TIM4, LAMP1, and MHC II. Specialised epithelial cells or M cells covering the bronchus-associated lymphoid tissue expressed CSF1R-transgene and type II pneumocytes expressed LAMP1 suggesting that these epithelial cells are phagocytic and transcytose antigen. Highly organised lymphoid tissue was seen in trachea from 4 weeks onwards. Throughout the air sacs at all ages, CSF1R-transgene+ cells were scattered and at later stages, CSF1R-transgene+ cells lined capillaries. These results will serve as a base for further functional characterization of macrophages and dendritic cells and their role in respiratory diseases and vaccine responses.
Assuntos
Galinhas/genética , Galinhas/imunologia , Macrófagos/imunologia , Monócitos/metabolismo , Sacos Aéreos/imunologia , Sacos Aéreos/metabolismo , Animais , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/imunologia , Animais Geneticamente Modificados/metabolismo , Galinhas/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Traqueia/imunologia , Traqueia/metabolismoRESUMO
Toll-like receptor (TLR) ligands (TLR-Ls) are critical activators of immunity and are successfully being developed as vaccine adjuvants in both mammals and birds. In this study, we investigated the synergistic effect of co-stimulation of membrane and endosomal TLRs on the innate immune responses using chicken bone marrow-derived macrophages (BMMs), and studied the effect of age on the induction of innate responses. BMMs from 1 and 4-week-old birds were stimulated with Pam3Cys-SK4 (PCSK; TLR2), synthetic monophosphoryl lipid A (MPLA), Di[3-deoxy-d-manno-octulosonyl]-lipid A ammonium salt (KLA; TLR4), Gardiquimod, Resiquimod (R848; TLR7), CpG class B and C (TLR21). Nitric oxide (NO) production and mRNA levels of IL-1ß, IL-10 and IL-12p40 showed macrophages from 4-week-old birds showed more sensitive responses compared to 1-week-old birds. The most potent TLR-Ls, PCSK, MPLA and CpG B were used to study the effect of co-stimulation on macrophages. Co-stimulation with TLR21 and TLR4 synergistically up-regulated inflammatory-related genes, as well as NO production. However, incubation of splenocytes with PCSK, MPLA and CpG B did not induce cell proliferation. Moreover, treatment with CpG B led to significant cell death.
Assuntos
Imunidade Inata/imunologia , Receptor 4 Toll-Like/imunologia , Receptores Toll-Like/imunologia , Fatores Etários , Animais , Galinhas/imunologia , Endossomos/imunologia , Imunidade Inata/efeitos dos fármacos , Lipídeo A/análogos & derivados , Lipídeo A/farmacologia , Lipopeptídeos/farmacologia , Macrófagos/imunologia , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Receptor 4 Toll-Like/efeitos dos fármacos , Receptores Toll-Like/efeitos dos fármacosRESUMO
Campylobacter jejuni is the leading cause of bacterial food-borne gastroenteritis worldwide and human infections are frequently associated with handling and consumption of contaminated poultry. The polysaccharide capsule of C. jejuni plays important roles in colonisation of the chicken gut, invasion of epithelial cells and serum resistance and is subject to modification with O-methyl phosphoramidate (MeOPN) in most strains. In this study, the cytokine responses of mouse bone marrow-derived macrophages (mBMMs), chicken bone marrow-derived macrophages (chBMMs) and human monocyte-derived macrophages (hMDMs) were measured following infection with C. jejuni 11168H wild-type (WT) or isogenic mutants lacking either the capsule (Δcj1439) or its MeOPN modification (Δcj1417). Consistent with previous observations using murine bone marrow-derived dendritic cells, mutants lacking the capsule or MeOPN elicited enhanced transcription of IL-6 and IL-10 in mBMMs compared to wild-type C. jejuni. However, the lack of capsule and MeOPN did not alter IL-6 and IL-10 expression in chBMMs and hMDMs compared to C. jejuni WT. Phagocytosis assays showed the acapsular mutant was not impaired in uptake or net intracellular survival after phagocytosis in both chicken and human macrophages; however, the phagocytosis of the MeOPN mutant was significantly decreased in both chicken and human macrophages. In conclusion, differences in the response of macrophages of varying host origin to Campylobacter were detected. The absence of MeOPN modification on the capsule of C. jejuni did not alter the levels of innate cytokine expression in both chicken and human macrophages compared to the 11168H WT, but affected phagocytosis by host macrophages.
Assuntos
Infecções por Campylobacter/veterinária , Campylobacter jejuni/fisiologia , Galinhas , Macrófagos/metabolismo , Doenças das Aves Domésticas/microbiologia , Amidas/metabolismo , Animais , Cápsulas Bacterianas/metabolismo , Medula Óssea , Infecções por Campylobacter/microbiologia , Campylobacter jejuni/genética , Citocinas/metabolismo , Humanos , Monócitos/metabolismo , Mutação , Ácidos Fosfóricos/metabolismo , RatosRESUMO
Marek's disease virus (MDV) is an alphaherpesvirus that induces T-cell lymphomas in chickens. Natural infections in vivo are caused by the inhalation of infected poultry house dust and it is presumed that MDV infection is initiated in the macrophages from where the infection is passed to B cells and activated T cells. Virus can be detected in B and T cells and macrophages in vivo, and both B and T cells can be infected in vitro. However, attempts to infect macrophages in vitro have not been successful. The aim of this study was to develop a model for infecting phagocytes [macrophages and dendritic cells (DCs)] with MDV in vitro and to characterize the infected cells. Chicken bone marrow cells were cultured with chicken CSF-1 or chicken IL-4 and chicken CSF-2 for 4 days to produce macrophages and DCs, respectively, and then co-cultured with FACS-sorted chicken embryo fibroblasts (CEFs) infected with recombinant MDV expressing EGFP. Infected phagocytes were identified and sorted by FACS using EGFP expression and phagocyte-specific mAbs. Detection of MDV-specific transcripts of ICP4 (immediate early), pp38 (early), gB (late) and Meq by RT-PCR provided evidence for MDV replication in the infected phagocytes. Time-lapse confocal microscopy was also used to demonstrate MDV spread in these cells. Subsequent co-culture of infected macrophages with CEFs suggests that productive virus infection may occur in these cell types. This is the first report of in vitro infection of phagocytic cells by MDV.
Assuntos
Herpesvirus Galináceo 2/fisiologia , Fagócitos/virologia , Replicação Viral , Animais , Células Cultivadas , Galinhas , Técnicas de Cocultura , Doença de Marek/virologia , Modelos BiológicosRESUMO
Marek's disease virus (MDV) is a cell associated alphaherpesvirus that causes fatal lymphoma in chickens. One factor that plays a crucial role in MDV pathogenesis is the viral CXC chemokine vIL-8 that was originally named after chicken interleukin 8 (cIL-8). However, a recent study demonstrated that vIL-8 recruits B cells and a subset of T cells but not neutrophils, suggesting that vIL-8 is not a cIL-8 orthologue. In this study, we set to identify the cellular orthologues and receptor of vIL-8 using in silico analyses, binding and chemotaxis assays. Sequence and phylogenetic analyses of all chicken CXC chemokines present in the recently published chicken genome revealed that vIL-8 shares the highest amino acid similarity with the CXCL13L1 variant. To evaluate if vIL-8 and CXCL13L1 are also functional orthologues, we assessed their binding properties and chemotaxis activity. We demonstrated that both vIL-8 and CXCL13 variants bind B cells and subsets of T cells, confirming that they target the same cell types. In addition, the chemokines not only bound the target cells but also induced chemotaxis. Finally, we identified CXCR5 as the receptor of vIL-8 and CXCL13 variants and confirmed that the receptor is expressed on MDV target cells. Taken together, our data demonstrate the conservation of the receptor-ligand interaction between CXCR5 and CXCL13 and shed light on the origin and function of the MDV-encoded vIL-8 chemokine, which plays a crucial role in the pathogenesis of this highly oncogenic virus.
RESUMO
In mammals, the inducible cytokine interleukin 10 is a feedback negative regulator of inflammation. To determine the extent to which this function is conserved in birds, recombinant chicken IL-10 was expressed as a secreted human Ig Fc fusion protein (chIL-10-Fc) and used to immunise mice. Five monoclonal antibodies (mAb) which specifically recognise chicken IL-10 were generated and characterised. Two capture ELISA assays were developed which detected native chIL-10 secreted from chicken bone marrow-derived macrophages (chBMMs) stimulated with lipopolysaccharide (LPS). Three of the mAbs detected intracellular IL-10. This was detected in only a subset of the same LPS-stimulated chBMMs. The ELISA assay also detected massive increases in circulating IL-10 in chickens challenged with the coccidial parasite, Eimeria tenella. The same mAbs neutralised the bioactivity of recombinant chIL-10. The role of IL-10 in feedback control was tested in vitro. The neutralising antibodies prevented IL-10-induced inhibition of IFN-γ synthesis by mitogen-activated lymphocytes and increased nitric oxide production in LPS-stimulated chBMMs. The results confirm that IL-10 is an inducible feedback regulator of immune response in chickens, and could be the target for improved vaccine efficacy or breeding strategies.
Assuntos
Galinhas/imunologia , Coccidiose/imunologia , Eimeria tenella/imunologia , Interleucina-10/metabolismo , Macrófagos/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Neutralizantes/metabolismo , Evolução Biológica , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Humanos , Imunidade , Interleucina-10/imunologia , Mamíferos , CamundongosRESUMO
Immunoneuroendocrinology studies have identified conserved communicational paths in birds and mammals, e.g. the Hypothalamus-Pituitary-Adrenal axis with anti-inflammatory activity mediated by glucocorticoids. Immune neuroendocrine phenotypes (INPs) have been proposed for mammals implying the categorization of a population in subgroups underlying divergent immune-neuroendocrine interactions. These phenotypes were studied in the context of the LEWIS/FISCHER paradigm (rats expressing high or low pro-inflammatory profiles, respectively). Although avian species have some common immunological mechanisms with mammals, they have also evolved some distinct strategies and, until now, it has not been studied whether birds may also share with mammals similar INPs. Based on corticosterone levels we determined the existence of two divergent groups in Coturnix coturnix that also differed in other immune-neuroendocrine responses. Quail with lowest corticosterone showed higher lymphoproliferative and antibody responses, interferon-γ and interleukin-1ß mRNA expression levels and lower frequencies of leukocyte subpopulations distribution and interleukin-13 levels, than their higher corticosterone counterparts. Results suggest the existence of INPs in birds, comparable to mammalian LEWIS/FISCHER profiles, where basal corticosterone also underlies responses of comparable variables associated to the phenotypes. Concluding, INP may not be a mammalian distinct feature, leading to discuss whether these profiles represent a parallel phenomenon evolved in birds and mammals, or a common feature inherited from a reptilian ancestor millions of years ago.
Assuntos
Coturnix/fisiologia , Animais , Feminino , Sistema Hipotálamo-Hipofisário , Masculino , Modelos Biológicos , Fenótipo , Sistema Hipófise-SuprarrenalRESUMO
A new member of the chicken TNF superfamily has recently been identified, namely receptor activator of NF-κB ligand (RANKL), as have its signalling receptor, RANK, and its decoy receptor, osteoprotegerin (OPG). In mammals, RANKL and RANK are transmembrane proteins expressed on the surface of Th1 cells and dendritic cells (DC) respectively, whereas OPG is expressed as a soluble protein from osteoblasts and DC. Recombinant soluble chicken RANKL (chRANKL) forms homotrimers whereas chicken OPG (chOPG) forms homodimers, characteristic of these molecules in mammals. ChRANKL, chRANK and chOPG are expressed at the mRNA level in most tissues and organs. ChRANKL is transcriptionally regulated by Ca(2+) mobilisation and enhances the mRNA expression levels of pro-inflammatory cytokines in bone marrow-derived DC (BMDC); this is inhibited by both chOPG-Fc and soluble chRANK-Fc. However, chRANKL does not enhance the expression of cell surface markers in either BMDC or BM-derived macrophages (BMM). Furthermore, chRANKL enhances the survival of APC similar to its mammalian orthologue.
Assuntos
Proteínas Aviárias/metabolismo , Galinhas/imunologia , Células Dendríticas/imunologia , Osteoblastos/imunologia , Osteoprotegerina/metabolismo , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Células Th1/imunologia , Animais , Proteínas Aviárias/genética , Evolução Biológica , Sinalização do Cálcio , Sobrevivência Celular , Sequência Conservada/genética , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Osteoprotegerina/genética , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/genéticaRESUMO
We have generated the first transgenic chickens in which reporter genes are expressed in a specific immune cell lineage, based upon control elements of the colony stimulating factor 1 receptor (CSF1R) locus. The Fms intronic regulatory element (FIRE) within CSF1R is shown to be highly conserved in amniotes and absolutely required for myeloid-restricted expression of fluorescent reporter genes. As in mammals, CSF1R-reporter genes were specifically expressed at high levels in cells of the macrophage lineage and at a much lower level in granulocytes. The cell lineage specificity of reporter gene expression was confirmed by demonstration of coincident expression with the endogenous CSF1R protein. In transgenic birds, expression of the reporter gene provided a defined marker for macrophage-lineage cells, identifying the earliest stages in the yolk sac, throughout embryonic development and in all adult tissues. The reporter genes permit detailed and dynamic visualisation of embryonic chicken macrophages. Chicken embryonic macrophages are not recruited to incisional wounds, but are able to recognise and phagocytose microbial antigens.
Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Macrófagos/citologia , Animais , Animais Geneticamente Modificados , Sequência de Bases , Aves , Linhagem da Célula , Galinhas , Células Dendríticas/citologia , Genes Reporter , Técnicas Genéticas , Sistema Imunitário , Íntrons , Dados de Sequência Molecular , Fagocitose , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie , Transgenes , Saco Vitelino/fisiologiaRESUMO
Macrophages contribute to innate and acquired immunity as well as many aspects of homeostasis and development. Studies of macrophage biology and function in birds have been hampered by a lack of definitive cell surface markers. As in mammals, avian macrophages proliferate and differentiate in response to CSF1 and IL34, acting through the shared receptor, CSF1R. CSF1R mRNA expression in the chicken is restricted to macrophages and their progenitors. To expedite studies of avian macrophage biology, we produced an avian CSF1R-Fc chimeric protein and generated a monoclonal antibody (designated ROS-AV170) against the chicken CSF1R using the chimeric protein as immunogen. Specific binding of ROS-AV170 to CSF1R was confirmed by FACS, ELISA and immunohistochemistry on tissue sections. CSF1 down-regulated cell surface expression of the CSF1R detected with ROS-AV170, but the antibody did not block CSF1 signalling. Expression of CSF1R was detected on the surface of bone marrow progenitors only after culture in the absence of CSF1, and was induced during macrophage differentiation. Constitutive surface expression of CSF1R distinguished monocytes from other myeloid cells, including heterophils and thrombocytes. This antibody will therefore be of considerable utility for the study of chicken macrophage biology.
Assuntos
Anticorpos Monoclonais/imunologia , Galinhas/imunologia , Macrófagos/imunologia , Receptor de Fator Estimulador de Colônias de Macrófagos/imunologia , Animais , Células da Medula Óssea/imunologia , Células CHO , Diferenciação Celular/imunologia , Linhagem Celular , Linhagem da Célula/imunologia , Cricetulus , Feminino , Fator Estimulador de Colônias de Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/imunologia , RNA Mensageiro/biossíntese , Receptor de Fator Estimulador de Colônias de Macrófagos/biossíntese , Receptor de Fator Estimulador de Colônias de Macrófagos/genéticaRESUMO
Salmonella enterica subspecies enterica serovar Gallinarum biovar Gallinarum (SG) is a non-flagellated bacterium which causes fowl typhoid, a systemic disease associated with high mortality in birds. It has been suggested that the absence of flagella in SG is advantageous in the early stages of systemic infection through absence of TLR-5 activation. In order to investigate this hypothesis in more detail a flagellated and motile SG mutant (SG Fla(+)) was constructed. The presence of flagella increased invasiveness for chicken kidney cells (CKC) while its presence did not alter survival in HD11 macrophages. SG Fla(+) induced higher levels of CXCLi2, IL-6 and iNOS mRNA expression in CKC than the SG parent strain. The expression of genes responsible for immune response mediators in infected HD11 macrophages were not related to the presence of flagella. Mortality rates were lower in birds challenged with SG Fla(+) when compared with the SG parent. SG Fla(+) was recovered from caecal contents which showed pathological changes suggestive of inflammation and suggested increased colonization ability.
Assuntos
Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Flagelos/genética , Macrófagos , Doenças das Aves Domésticas , Salmonelose Animal , Virulência/genética , Animais , Ceco/microbiologia , Linhagem Celular , Galinhas , Regulação Bacteriana da Expressão Gênica , Macrófagos/imunologia , Macrófagos/microbiologia , Viabilidade Microbiana , Mutação , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/microbiologia , Salmonelose Animal/imunologia , Salmonelose Animal/microbiologia , Salmonella enterica/genética , Salmonella enterica/imunologia , Salmonella enterica/patogenicidadeRESUMO
The chicken, being a non-mammal, has perhaps surprisingly made many seminal contributions towards our understanding of immune responses in all species. Despite this, pre-genome our ability to study immune responses in detail in birds was limited both in terms of reagents and also in our understanding of the immune gene repertoire in birds. Post-genome things have radically changed, although there are still gaps, both in the genome sequence and in the repertoire, the latter not conclusive until the genome is complete. We now know that for many immune gene families (e.g. the major histocompatibility complex, the tumour necrosis factor superfamily and the tumour necrosis factor receptor superfamily, the chemokines, the interleukin-1 family) the chicken repertoire is smaller, although there are interesting/confusing examples (e.g. the chicken immunoglobulin-like receptors family) where the chicken repertoire is greater than that of mammals. The future for chicken immunology is exciting, and in particular we have the potential to develop novel solutions for disease (such as novel vaccines or breeding for disease resistance) based on our improving knowledge of the chicken's immune response to disease.
Assuntos
Galinhas/genética , Galinhas/imunologia , Genoma/imunologia , Doenças das Aves Domésticas/imunologia , Animais , Genoma/genética , Genômica , Imunidade/genética , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/prevenção & controleRESUMO
Poultry-derived food is a common source of infection of human with the non-host-adapted salmonellae while fowl typhoid and pullorum disease are serious diseases in poultry. Development of novel immune-based control strategies against Salmonella infection necessitates a better understanding of the host-pathogen interactions at the cellular level. Intestinal epithelial cells are the first line of defence against enteric infections and the role of macrophages is crucial in Salmonella infection and pathogenesis. While gene expression following Salmonella infection has been investigated, a comparison between different serovars has not been, as yet, extensively studied in poultry. In this study, chicken macrophage-like cells (HD11) and chick kidney epithelial cells (CKC) were used to study and compare the immune responses and mechanisms that develop after infection with different Salmonella serotypes. Salmonella serovars Typhimurium, Enteritidis, Hadar and Infantis showed a greater level of invasion and/or uptake characters when compared with S. Pullorum or S. Gallinarum. Nitrate and reactive oxygen species were greater in Salmonella-infected HD11 cells with the expression of iNOS and nuclear factor-κB by chicken macrophages infected with both systemic and broad host range serovars. HD11 cells revealed higher mRNA gene expression for CXCLi2, IL-6 and iNOS genes in response to S. Enteritidis infection when compared to S. Pullorum-infected cells. S. Typhimurium- and S. Hadar-infected HD11 showed higher gene expression for CXCLi2 versus S. Pullorum-infected cells. Higher mRNA gene expression levels of pro-inflammatory cytokine IL-6, chemokines CXCLi1 and CXCLi2 and iNOS genes were detected in S. Typhimurium- and S. Enteritidis-infected CKC followed by S. Hadar and S. Infantis while no significant changes were observed in S. Pullorum or S. Gallinarum-infected CKC.
Assuntos
Galinhas , Macrófagos/microbiologia , NF-kappa B/imunologia , Óxido Nítrico/imunologia , Doenças das Aves Domésticas/microbiologia , Espécies Reativas de Oxigênio/imunologia , Salmonelose Animal/imunologia , Salmonella/imunologia , Animais , Western Blotting/veterinária , Linhagem Celular , Sobrevivência Celular/imunologia , Citocinas/genética , Citocinas/imunologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Interações Hospedeiro-Patógeno/imunologia , Macrófagos/imunologia , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/imunologia , Doenças das Aves Domésticas/imunologia , RNA/química , RNA/genética , Espécies Reativas de Nitrogênio/análise , Espécies Reativas de Nitrogênio/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Salmonella/genética , Salmonelose Animal/microbiologiaRESUMO
In mammals, interleukin-21 (IL-21) is an immunomodulatory cytokine with pleiotropic effects on the proliferation, differentiation and effector functions of T, B, NK and dendritic cells. A cDNA encoding the chicken orthologue of IL-21 (chIL-21) was cloned by RT-PCR from RNA isolated from activated chicken splenocytes and consists of 438 nucleotides, encoding an open reading frame of 145 amino acids (aa). Chicken IL-21 has 20-30% aa identity to its orthologues in mammals, Xenopus and fish, but is more highly conserved within Aves (50-80%). The four alpha-helical bundle structure of mammalian IL-21 appears to be conserved in the predicted chicken protein, as are the four cysteine residues required for the formation of two disulphide bridges. A glutamine residue in aa position 129, which has been implicated in the binding of IL-21 to the IL-2 receptor γ-chain in mammals, is also conserved. ChIL-21 is expressed in most lymphoid tissues, predominantly by CD4+ TCRαß+ T cells. As in mammals, chIL-21 synergistically enhances T-cell proliferation and inhibits maturation of dendritic cells.
Assuntos
Galinhas/imunologia , Células Dendríticas/imunologia , Interleucinas/imunologia , Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Bioensaio/veterinária , Clonagem Molecular/métodos , Sequência Conservada , Células Dendríticas/citologia , Interleucinas/genética , Dados de Sequência Molecular , RNA/química , RNA/genética , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Alinhamento de Sequência , Análise de Sequência de DNA , Organismos Livres de Patógenos Específicos , Linfócitos T/citologiaRESUMO
Marek's disease virus (MDV) is a highly contagious oncogenic alphaherpesvirus that causes disease that is both a cancer model and a continuing threat to the world's poultry industry. This comprehensive gene expression study analyzes the host response to infection in both resistant and susceptible lines of chickens and inherent expression differences between the two lines following the infection of the host. A novel pathogenicity mechanism, involving the downregulation of genes containing HIC1 transcription factor binding sites as early as 4 days postinfection, was suggested from this analysis. HIC1 drives antitumor mechanisms, suggesting that MDV infection switches off genes involved in antitumor regulation several days before the expression of the MDV oncogene meq. The comparison of the gene expression data to previous QTL data identified several genes as candidates for involvement in resistance to MD. One of these genes, IRG1, was confirmed by single nucleotide polymorphism analysis to be involved in susceptibility. Its precise mechanism remains to be elucidated, although the analysis of gene expression data suggests it has a role in apoptosis. Understanding which genes are involved in susceptibility/resistance to MD and defining the pathological mechanisms of the disease gives us a much greater ability to try to reduce the incidence of this virus, which is costly to the poultry industry in terms of both animal welfare and economics.