Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 8795, 2024 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-38627516

RESUMO

In mammals, a subset of follicle-associated epithelial (FAE) cells, known as M cells, conduct the transcytosis of antigens across the epithelium into the underlying lymphoid tissues. We previously revealed that M cells in the FAE of the chicken lung, bursa of Fabricius (bursa), and caecum based on the expression of CSF1R. Here, we applied RNA-seq analysis on highly enriched CSF1R-expressing bursal M cells to investigate their transcriptome and identify novel chicken M cell-associated genes. Our data show that, like mammalian M cells, those in the FAE of the chicken bursa also express SOX8, MARCKSL1, TNFAIP2 and PRNP. Immunohistochemical analysis also confirmed the expression of SOX8 in CSF1R-expressing cells in the lung, bursa, and caecum. However, we found that many other mammalian M cell-associated genes such as SPIB and GP2 were not expressed by chicken M cells or represented in the chicken genome. Instead, we show bursal M cells express high levels of related genes such as SPI1. Whereas our data show that bursal M cells expressed CSF1R-highly, the M cells in the small intestine lacked CSF1R and both expressed SOX8. This study offers insights into the transcriptome of chicken M cells, revealing the expression of CSF1R in M cells is tissue-specific.


Assuntos
Galinhas , Células M , Animais , Bolsa de Fabricius/metabolismo , Galinhas/genética , Galinhas/metabolismo , Epitélio , Tecido Linfoide , Receptores de Fator Estimulador de Colônias/metabolismo
2.
Viruses ; 15(11)2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-38005955

RESUMO

The avian coronavirus, infectious bronchitis virus (IBV), is an economically important infectious disease affecting chickens, with a diverse range of serotypes found globally. The major surface protein, spike (S), has high diversity between serotypes, and amino acid differences in the S1 sub-unit are thought to be responsible for poor cross-protection afforded by vaccination. Here, we attempt to address this, by using epitope mapping technology to identify shared and serotype-specific immunogenic epitopes of the S glycoprotein of three major circulating strains of IBV, M41, QX, and 4/91, via CLIPS peptide arrays based on peptides from the S1 sub-units. The arrays were screened with sera from chickens immunised with recombinant IBV, based on Beau-R backbone expressing heterologous S, generated in two independent vaccination/challenge trials. The screening of sera from rIBV vaccination experiments led to the identification of 52 immunogenic epitopes on the S1 of M41, QX, and 4/91. The epitopes were assigned into six overlapping epitope binding regions. Based on accessibility and location in the hypervariable regions of S, three sequences, 25YVYYYQSAFRPPNGWHLQGGAYAVVNSTN54, 67TVGVIKDVYNQSVASI82, and 83AMTVPPAGMSWSVS96, were selected for further investigation, and synthetic peptide mimics were recognised by polyclonal sera. These epitopes may have the potential to contribute towards a broader cross-protective IBV vaccine.


Assuntos
Infecções por Coronavirus , Vírus da Bronquite Infecciosa , Doenças das Aves Domésticas , Vacinas Virais , Animais , Epitopos , Mapeamento de Epitopos , Galinhas , Peptídeos , Glicoproteínas , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/veterinária , Glicoproteína da Espícula de Coronavírus
3.
Vet Res ; 53(1): 15, 2022 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-35236416

RESUMO

Three-dimensional (3D) intestinal enteroids are powerful in vitro models for studying intestinal biology. However, due to their closed structure direct access to the apical surface is impeded, limiting high-throughput applications of exogenous compounds and pathogens. In this study, we describe a method for generating confluent 2D enteroids from single-cell suspensions of enzymatically-dissociated ileum-derived bovine 3D enteroids. Confluent monolayers were first achieved using IntestiCult media but to establish a defined, cost-effective culture media, we also developed a bovine enteroid monolayer (BEM) medium. The monolayers cultured in BEM media proliferated extensively and formed confluent cell layers on both Matrigel-coated plastic plates and transwell inserts by day 3 of culture. The 2D enteroids maintained the epithelial cell lineages found in 3D enteroids and ileum tissue. In addition, the monolayers formed a functional epithelial barrier based on the presence of the adherens and tight junction proteins, E-cadherin and ZO-1, and electrical resistance across the monolayer was measured from day 3 and maintained for up to 7 days in culture. The method described here will provide a useful model to study bovine epithelial cell biology with ease of access to the apical surface of epithelial cells and has potential to investigate host-pathogen interactions and screen bioactive compounds.


Assuntos
Células Epiteliais , Mucosa Intestinal , Animais , Bovinos , Interações Hospedeiro-Patógeno , Íleo , Intestinos
4.
Front Immunol ; 13: 1064084, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36618373

RESUMO

Chicken bone marrow-derived macrophages (BMMΦ) and dendritic cells (BMDC) are utilized as models to study the mononuclear phagocytic system (MPS). A widely used method to generate macrophages and DC in vitro is to culture bone marrow cells in the presence of colony-stimulating factor-1 (CSF1) to differentiate BMMΦ and granulocyte-macrophage-CSF (GM-CSF, CSF2) and interleukin-4 (IL-4) to differentiate BMDC, while CSF2 alone can lead to the development of granulocyte-macrophage-CSF-derived DC (GMDC). However, in chickens, the MPS cell lineages and their functions represented by these cultures are poorly understood. Here, we decipher the phenotypical, functional and transcriptional differences between chicken BMMΦ and BMDC along with examining differences in DC cultures grown in the absence of IL-4 on days 2, 4, 6 and 8 of culture. BMMΦ cultures develop into a morphologically homogenous cell population in contrast to the BMDC and GMDC cultures, which produce morphologically heterogeneous cell cultures. At a phenotypical level, all cultures contained similar cell percentages and expression levels of MHCII, CD11c and CSF1R-transgene, whilst MRC1L-B expression decreased over time in BMMΦ. All cultures were efficiently able to uptake 0.5 µm beads, but poorly phagocytosed 1 µm beads. Little difference was observed in the kinetics of phagosomal acidification across the cultures on each day of analysis. Temporal transcriptomic analysis indicated that all cultures expressed high levels of CSF3R, MERTK, SEPP1, SPI1 and TLR4, genes associated with macrophages in mammals. In contrast, low levels of FLT3, XCR1 and CAMD1, genes associated with DC, were expressed at day 2 in BMDC and GMDC after which expression levels decreased. Collectively, chicken CSF2 + IL-4- and CSF2-dependent BM cultures represent cells of the macrophage lineage rather than inducing conventional DC.


Assuntos
Galinhas , Interleucina-4 , Animais , Galinhas/metabolismo , Interleucina-4/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Medula Óssea/metabolismo , Células Dendríticas/metabolismo , Macrófagos/metabolismo , Mamíferos/metabolismo
5.
Commun Biol ; 4(1): 377, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33742093

RESUMO

Mammalian three-dimensional (3D) enteroids mirror in vivo intestinal organisation and are powerful tools to investigate intestinal cell biology and host-pathogen interactions. We have developed complex multilobulated 3D chicken enteroids from intestinal embryonic villi and adult crypts. These avian enteroids develop optimally in suspension without the structural support required to produce mammalian enteroids, resulting in an inside-out enteroid conformation with media-facing apical brush borders. Histological and transcriptional analyses show these enteroids comprise of differentiated intestinal epithelial cells bound by cell-cell junctions, and notably, include intraepithelial leukocytes and an inner core of lamina propria leukocytes. The advantageous polarisation of these enteroids has enabled infection of the epithelial apical surface with Salmonella Typhimurium, influenza A virus and Eimeria tenella without the need for micro-injection. We have created a comprehensive model of the chicken intestine which has the potential to explore epithelial and leukocyte interactions and responses in host-pathogen, food science and pharmaceutical research.


Assuntos
Eimeria tenella/patogenicidade , Células Epiteliais , Vírus da Influenza A/patogenicidade , Mucosa Intestinal , Leucócitos , Salmonella typhimurium/patogenicidade , Animais , Células Cultivadas , Microambiente Celular , Galinhas , Eimeria tenella/imunologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Células Epiteliais/parasitologia , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Vírus da Influenza A/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/parasitologia , Mucosa Intestinal/virologia , Leucócitos/imunologia , Leucócitos/microbiologia , Leucócitos/parasitologia , Leucócitos/virologia , Camundongos Endogâmicos C57BL , Organoides , Permeabilidade , Fagocitose , Fenótipo , Codorniz , Salmonella typhimurium/imunologia
6.
Front Immunol ; 10: 2495, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31695701

RESUMO

The follicle-associated epithelium (FAE) is a specialized structure that samples luminal antigens and transports them into mucosa-associated lymphoid tissues (MALT). In mammals, transcytosis of antigens across the gut epithelium is performed by a subset of FAE cells known as M cells. Here we show that colony-stimulating factor 1 receptor (CSF1R) is expressed by a subset of cells in the avian bursa of Fabricius FAE. Expression was initially detected using a CSF1R-reporter transgene that also label subsets of bursal macrophages. Immunohistochemical detection using a specific monoclonal antibody confirmed abundant expression of CSF1R on the basolateral membrane of FAE cells. CSF1R-transgene expressing bursal FAE cells were enriched for expression of markers previously reported as putative M cell markers, including annexin A10 and CD44. They were further distinguished from a population of CSF1R-transgene negative epithelial cells within FAE by high apical F-actin expression and differential staining with the lectins jacalin, PHA-L and SNA. Bursal FAE cells that express the CSF1R-reporter transgene were responsible for the bulk of FAE transcytosis of labeled microparticles in the size range 0.02-0.1 µm. Unlike mammalian M cells, they did not readily take up larger bacterial sized microparticles (0.5 µm). Their role in uptake of bacteria was tested using Salmonella, which can enter via M cells in mammals. Labeled Salmonella enterica serovar Typhimurium entered bursal tissue via the FAE. Entry was partially dependent upon Type III secretion system-1. However, the majority of invading bacteria were localized to CSF1R-negative FAE cells and in resident phagocytes that express the phosphatidylserine receptor TIM4. CSF1R-expressing FAE cells in infected follicles showed evidence of cell death and shedding into the bursal lumen. In mammals, CSF1R expression in the gut is restricted to macrophages which only indirectly control M cell differentiation. The novel expression of CSF1R in birds suggests that these functional equivalents to mammalian M cells may have different ontological origins and their development and function are likely to be regulated by different growth factors.


Assuntos
Apresentação de Antígeno/imunologia , Proteínas Aviárias/imunologia , Bolsa de Fabricius/imunologia , Células Epiteliais/imunologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Animais , Antígenos de Bactérias , Antígenos de Diferenciação/imunologia , Bolsa de Fabricius/patologia , Galinhas , Humanos , Infecções por Salmonella/patologia
7.
Genes (Basel) ; 10(2)2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30678299

RESUMO

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/imunologia
8.
Front Immunol ; 10: 3055, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31998322

RESUMO

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/imunologia
9.
Avian Pathol ; 48(2): 157-167, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30570345

RESUMO

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 14 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 Específicos
10.
Vet Res ; 49(1): 104, 2018 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-30305141

RESUMO

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/metabolismo
11.
Dev Comp Immunol ; 86: 156-170, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29729283

RESUMO

Mammalian type I interferons (IFNα/ß) are known to modulate inflammatory processes in addition to their antiviral properties. Indeed, virus-induced type I interferons regulate the mammalian phagocyte immune response to bacteria during superinfections. However, it remains unresolved whether type I IFNs similarly impact the chicken macrophage immune response. We first evidenced that IFNα and IFNß act differently in terms of gene expression stimulation and activation of intracellular signaling pathways in chicken macrophages. Next, we showed that priming of chicken macrophages with IFNα increased bacteria uptake, boosted bacterial-induced ROS/NO production and led to an increased transcriptional expression or production of NOS2/NO, IL1B/IL-1ß and notably IFNB/IFNß. Neutralization of IFNß during bacterial challenge limited IFNα-induced augmentation of the pro-inflammatory response. In conclusion, we demonstrated that type I IFNs differently regulate chicken macrophage functions and drive a pro-inflammatory response to bacterial challenge. These findings shed light on the diverse functions of type I IFNs in chicken macrophages.


Assuntos
Bactérias/imunologia , Galinhas/imunologia , Inflamação/imunologia , Interferon-alfa/imunologia , Interferon beta/imunologia , Macrófagos/imunologia , Animais , Galinhas/microbiologia , Expressão Gênica/imunologia , Inflamação/microbiologia , Macrófagos/microbiologia , Óxidos de Nitrogênio/imunologia , Espécies Reativas de Oxigênio/imunologia , Transdução de Sinais/imunologia
12.
Vet Immunol Immunopathol ; 199: 15-21, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29678225

RESUMO

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 fármacos
13.
Vet Res ; 49(1): 3, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29316981

RESUMO

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 , Ratos
14.
J Gen Virol ; 98(5): 1080-1088, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28548038

RESUMO

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 Biológicos
15.
Dev Comp Immunol ; 63: 206-12, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27108075

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 , Camundongos
16.
BMC Biol ; 13: 12, 2015 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-25857347

RESUMO

BACKGROUND: Macrophages have many functions in development and homeostasis as well as innate immunity. Recent studies in mammals suggest that cells arising in the yolk sac give rise to self-renewing macrophage populations that persist in adult tissues. Macrophage proliferation and differentiation is controlled by macrophage colony-stimulating factor (CSF1) and interleukin 34 (IL34), both agonists of the CSF1 receptor (CSF1R). In the current manuscript we describe the origin, function and regulation of macrophages, and the role of CSF1R signaling during embryonic development, using the chick as a model. RESULTS: Based upon RNA-sequencing comparison to bone marrow-derived macrophages grown in CSF1, we show that embryonic macrophages contribute around 2% of the total embryo RNA in day 7 chick embryos, and have similar gene expression profiles to bone marrow-derived macrophages. To explore the origins of embryonic and adult macrophages, we injected Hamburger-Hamilton stage 16 to 17 chick embryos with either yolk sac-derived blood cells, or bone marrow cells from EGFP+ donors. In both cases, the transferred cells gave rise to large numbers of EGFP+ tissue macrophages in the embryo. In the case of the yolk sac, these cells were not retained in hatched birds. Conversely, bone marrow EGFP+ cells gave rise to tissue macrophages in all organs of adult birds, and regenerated CSF1-responsive marrow macrophage progenitors. Surprisingly, they did not contribute to any other hematopoietic lineage. To explore the role of CSF1 further, we injected embryonic or hatchling CSF1R-reporter transgenic birds with a novel chicken CSF1-Fc conjugate. In both cases, the treatment produced a large increase in macrophage numbers in all tissues examined. There were no apparent adverse effects of chicken CSF1-Fc on embryonic or post-hatch development, but there was an unexpected increase in bone density in the treated hatchlings. CONCLUSIONS: The data indicate that the yolk sac is not the major source of macrophages in adult birds, and that there is a macrophage-restricted, self-renewing progenitor cell in bone marrow. CSF1R is demonstrated to be limiting for macrophage development during development in ovo and post-hatch. The chicken provides a novel and tractable model to study the development of the mononuclear phagocyte system and CSF1R signaling.


Assuntos
Galinhas/imunologia , Sistema Fagocitário Mononuclear/embriologia , Sistema Fagocitário Mononuclear/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Transdução de Sinais , Animais , Células Sanguíneas/efeitos dos fármacos , Células Sanguíneas/metabolismo , Densidade Óssea/efeitos dos fármacos , Células da Medula Óssea , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Embrião de Galinha , Galinhas/genética , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Sistema Fagocitário Mononuclear/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sequência de RNA , Transdução de Sinais/efeitos dos fármacos , Saco Vitelino/citologia
17.
Dev Comp Immunol ; 51(1): 170-84, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25796577

RESUMO

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/genética
18.
J Immunol ; 194(5): 2338-44, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25637020

RESUMO

We have identified differences in gene expression in macrophages grown from the bone marrow of male and female chickens in recombinant chicken M-CSF (CSF1). Cells were profiled with or without treatment with bacterial LPS for 24 h. Approximately 600 transcripts were induced by prolonged LPS stimulation to an equal extent in the male and female macrophages. Many transcripts encoded on the Z chromosome were expressed ∼1.6-fold higher in males, reflecting a lack of dosage compensation in the homogametic sex. A smaller set of W chromosome-specific genes was expressed only in females. LPS signaling in mammals is associated with induction of type 1 IFN-responsive genes. Unexpectedly, because IFNs are encoded on the Z chromosome of chickens, unstimulated macrophages from the female birds expressed a set of known IFN-inducible genes at much higher levels than male cells under the same conditions. To confirm that these differences were not the consequence of the actions of gonadal hormones, we induced gonadal sex reversal to alter the hormonal environment of the developing chick and analyzed macrophages cultured from male, female, and female sex-reversed embryos. Gonadal sex reversal did not alter the sexually dimorphic expression of either sex-linked or IFN-responsive genes. We suggest that female birds compensate for the reduced dose of inducible IFN with a higher basal set point of IFN-responsive genes.


Assuntos
Proteínas Aviárias/imunologia , Galinhas/imunologia , Gônadas/imunologia , Macrófagos/imunologia , RNA Mensageiro/imunologia , Cromossomos Sexuais/imunologia , Animais , Inibidores da Aromatase/farmacologia , Proteínas Aviárias/genética , Células Cultivadas , Embrião de Galinha , Galinhas/genética , Mecanismo Genético de Compensação de Dose , Fadrozol/farmacologia , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Gônadas/efeitos dos fármacos , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon beta/genética , Interferon beta/imunologia , Lipopolissacarídeos/farmacologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Masculino , RNA Mensageiro/genética , Caracteres Sexuais
19.
Development ; 141(16): 3255-65, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25063453

RESUMO

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/fisiologia
20.
Dev Comp Immunol ; 41(3): 341-51, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23542704

RESUMO

Macrophages (MPh) and dendritic cells (DC) are members of the mononuclear phagocyte system. In chickens, markers to distinguish MPh from DC are lacking, but whether MPh and DC can be distinguished in humans and mice is under debate, despite the availability of numerous markers. Mucosal MPh and DC are strategically located to ingest foreign antigens, suggesting they can rapidly respond to invading pathogens. This review addresses our current understanding of DC and MPh function, the receptors expressed by MPh and DC involved in pathogen recognition, and the responses of DC and MPh against respiratory and intestinal pathogens in the chicken. Furthermore, potential opportunities are described to modulate MPh and DC responses to enhance disease resistance, highlighting modulation through nutraceuticals and vaccination.


Assuntos
Galinhas/imunologia , Células Dendríticas/imunologia , Trato Gastrointestinal/imunologia , Macrófagos/imunologia , Sistema Respiratório/imunologia , Animais , Coccidiose/imunologia , Coccidiose/prevenção & controle , Células Dendríticas/microbiologia , Células Dendríticas/parasitologia , Células Dendríticas/virologia , Suplementos Nutricionais/estatística & dados numéricos , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/prevenção & controle , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/parasitologia , Trato Gastrointestinal/virologia , Imunidade Inata , Imunomodulação , Influenza Aviária/imunologia , Influenza Aviária/prevenção & controle , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/imunologia , Macrófagos/microbiologia , Macrófagos/parasitologia , Macrófagos/virologia , Sistema Respiratório/microbiologia , Sistema Respiratório/parasitologia , Sistema Respiratório/virologia , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/genética , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico/imunologia , Vacinação/estatística & dados numéricos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA