Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Leukoc Biol ; 107(2): 237-250, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31487076

RESUMO

Macrophage colony-stimulating factor (CSF1 or M-CSF) and interleukin 34 (IL34) are secreted cytokines that control macrophage survival and differentiation. Both act through the CSF1 receptor (CSF1R), a type III transmembrane receptor tyrosine kinase. The functions of CSF1R and both ligands are conserved in birds. We have analyzed protein-coding sequence divergence among avian species. The intracellular tyrosine kinase domain of CSF1R was highly conserved in bird species as in mammals but the extracellular domain of avian CSF1R was more divergent in birds with multiple positively selected amino acids. Based upon crystal structures of the mammalian CSF1/IL34 receptor-ligand interfaces and structure-based alignments, we identified amino acids involved in avian receptor-ligand interactions. The contact amino acids in both CSF1 and CSF1R diverged among avian species. Ligand-binding domain swaps between chicken and zebra finch CSF1 confirmed the function of variants that confer species specificity on the interaction of CSF1 with CSF1R. Based upon genomic sequence analysis, we identified prevalent amino acid changes in the extracellular domain of CSF1R even within the chicken species that distinguished commercial broilers and layers and tropically adapted breeds. The rapid evolution in the extracellular domain of avian CSF1R suggests that at least in birds this ligand-receptor interaction is subjected to pathogen selection. We discuss this finding in the context of expression of CSF1R in antigen-sampling and antigen-presenting cells.


Assuntos
Evolução Biológica , Interleucinas/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Polimorfismo Genético , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Animais , Galinhas , Interleucinas/genética , Ligantes , Fator Estimulador de Colônias de Macrófagos/genética , Filogenia , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Especificidade da Espécie , Peixe-Zebra
2.
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
3.
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
4.
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
5.
Mol Cell Endocrinol ; 382(1): 150-158, 2014 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-23994026

RESUMO

Obesity is characterised by hyperleptinaemia and hypoadiponectinaemia and these metabolic abnormalities may contribute to the progression of several obesity-associated cancers including oesophageal adenocarcinoma (OAC). We have examined the effects of leptin and adiponectin on OE33 OAC cells. Leptin stimulated proliferation, invasion and migration and inhibited apoptosis in a STAT3-dependant manner. Leptin-stimulated MMP-2 secretion in a partly STAT3-dependent manner and MMP-9 secretion via a STAT3-independent pathway. Adiponectin inhibited leptin-induced proliferation, migration, invasion, MMP secretion and reduced the anti-apoptotic effects: these effects of adiponectin were ameliorated by both a non-specific tyrosine phosphatase inhibitor and a specific PTP1B inhibitor. Adiponectin reduced leptin-stimulated JAK2 activation and STAT3 transcriptional activity in a PTP1B-sensitive manner and adiponectin increased both PTP1B protein and activity. We conclude that adiponectin restrains leptin-induced signalling and pro-carcinogenic behaviour by inhibiting the early events in leptin-induced signal transduction by activating PTP1B. Relative adiponectin deficiency in obesity may contribute to the promotion of OAC.


Assuntos
Adiponectina/farmacologia , Neoplasias Esofágicas/enzimologia , Neoplasias Esofágicas/patologia , Leptina/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Janus Quinase 2/metabolismo , Metaloproteinases da Matriz/metabolismo , Modelos Biológicos , Invasividade Neoplásica , Fator de Transcrição STAT3/metabolismo
6.
Dev Comp Immunol ; 42(2): 278-85, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24084378

RESUMO

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/genética
7.
Mech Dev ; 125(11-12): 1059-70, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18684398

RESUMO

Macrophages are essential in development, repair and pathology of a variety of tissues via their roles in tissue remodelling, wound healing and inflammation. These biological functions are also associated with a number of human diseases, for example tumour associated macrophages have well defined functions in cancer progression. Xenopus embryonic macrophages arise from a haematopoietic stem cell population by direct differentiation and act as the main mechanism of host defence, before lymphoid cells and a circulatory system have developed. This function is conserved in mouse and human development. Macrophages express a number of matrix metalloproteinases (MMPs), which are central to their function. MMPs are a large family of zinc-dependent endoproteases with multiple roles in extracellular matrix remodelling and the modulation of signalling pathways. We have previously shown MMP-7 to be expressed by Xenopus embryonic macrophages. Here we investigate the role of MMP-7 and two other MMPs (MMP-18 and MMP-9) that are also expressed in the migrating macrophages. Using morpholino (MO) mediated knockdown of each of the MMPs we demonstrate that they are necessary for normal macrophage migration in vivo. The loss-of-function effect can be rescued using the specific MMPs, altered to be resistant to morpholinos but not by overexpression of the other MMPs. Double and triple morpholino knockdowns further suggest that these MMPs act combinatorily to promote embryonic macrophage migration. Thus, our results imply that these three MMPs have distinct functions, which together are crucial to mediate macrophage migration in the developing embryo. This demonstrates conclusively that MMPs are required for normal macrophage cell migration in the whole organism.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Macrófagos/citologia , Metaloproteinases da Matriz/fisiologia , Animais , Movimento Celular , Matriz Extracelular/metabolismo , Técnicas Genéticas , Coração/embriologia , Macrófagos/enzimologia , Macrófagos/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinases da Matriz/metabolismo , Metaloproteinases da Matriz Secretadas/metabolismo , Modelos Genéticos , Fases de Leitura Aberta , Temperatura , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA