Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
BMC Biol ; 13: 12, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25857347

ABSTRACT

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.


Subject(s)
Chickens/immunology , Mononuclear Phagocyte System/embryology , Mononuclear Phagocyte System/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction , Animals , Blood Cells/drug effects , Blood Cells/metabolism , Bone Density/drug effects , Bone Marrow Cells , Cell Differentiation/drug effects , Cell Line , Chick Embryo , Chickens/genetics , Flow Cytometry , Gene Expression Regulation, Developmental/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Mononuclear Phagocyte System/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, RNA , Signal Transduction/drug effects , Yolk Sac/cytology
3.
Development ; 141(16): 3255-65, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25063453

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Developmental , Macrophages/cytology , Animals , Animals, Genetically Modified , Base Sequence , Birds , Cell Lineage , Chickens , Dendritic Cells/cytology , Genes, Reporter , Genetic Techniques , Immune System , Introns , Molecular Sequence Data , Phagocytosis , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Sequence Homology, Nucleic Acid , Species Specificity , Transgenes , Yolk Sac/physiology
4.
Dev Comp Immunol ; 42(2): 278-85, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24084378

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/immunology , Chickens/immunology , Macrophages/immunology , Receptor, Macrophage Colony-Stimulating Factor/immunology , Animals , Bone Marrow Cells/immunology , CHO Cells , Cell Differentiation/immunology , Cell Line , Cell Lineage/immunology , Cricetulus , Female , Macrophage Colony-Stimulating Factor/immunology , Mice , Mice, Inbred BALB C , Monocytes/immunology , RNA, Messenger/biosynthesis , Receptor, Macrophage Colony-Stimulating Factor/biosynthesis , Receptor, Macrophage Colony-Stimulating Factor/genetics
5.
J Biol Chem ; 288(35): 25362-25374, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23853092

ABSTRACT

Broad-spectrum inhibitors of histone deacetylases (HDACs) constrain Toll-like receptor (TLR)-inducible production of key proinflammatory mediators. Here we investigated HDAC-dependent inflammatory responses in mouse macrophages. Of the classical Hdacs, Hdac7 was expressed at elevated levels in inflammatory macrophages (thioglycollate-elicited peritoneal macrophages) as compared with bone marrow-derived macrophages and the RAW264 cell line. Overexpression of a specific, alternatively spliced isoform of Hdac7 lacking the N-terminal 22 amino acids (Hdac7-u), but not the Refseq Hdac7 (Hdac7-s), promoted LPS-inducible expression of Hdac-dependent genes (Edn1, Il-12p40, and Il-6) in RAW264 cells. A novel class IIa-selective HDAC inhibitor reduced recombinant human HDAC7 enzyme activity as well as TLR-induced production of inflammatory mediators in thioglycollate-elicited peritoneal macrophages. Both LPS and Hdac7-u up-regulated the activity of the Edn1 promoter in an HDAC-dependent fashion in RAW264 cells. A hypoxia-inducible factor (HIF) 1 binding site in this promoter was required for HDAC-dependent TLR-inducible promoter activity and for Hdac7- and HIF-1α-mediated trans-activation. Coimmunoprecipitation assays showed that both Hdac7-u and Hdac7-s interacted with HIF-1α, whereas only Hdac7-s interacted with the transcriptional repressor CtBP1. Thus, Hdac7-u positively regulates HIF-1α-dependent TLR signaling in macrophages, whereas an interaction with CtBP1 likely prevents Hdac7-s from exerting this effect. Hdac7 may represent a potential inflammatory disease target.


Subject(s)
Gene Expression Regulation , Histone Deacetylases/metabolism , Macrophages/metabolism , Toll-Like Receptor 4/metabolism , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , Histone Deacetylases/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/pharmacology , Macrophages/pathology , Mice , Signal Transduction/drug effects , Signal Transduction/genetics , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics
6.
Cytokine ; 61(2): 630-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23260168

ABSTRACT

Colony stimulating factor (CSF-1) and its receptor, CSF-1R, have been previously well studied in humans and rodents to dissect the role they play in development of cells of the mononuclear phagocyte system. A second ligand for the CSF-1R, IL-34 has been described in several species. In this study, we have cloned and expressed the feline CSF-1R and examined the responsiveness to CSF-1 and IL-34 from a range of species. The results indicate that pig and human CSF-1 and human IL-34 are equally effective in cats, where both mouse CSF-1 and IL-34 are significantly less active. Recombinant human CSF-1 can be used to generate populations of feline bone marrow and monocyte derived macrophages that can be used to further dissect macrophage-specific gene expression in this species, and to compare it to data derived from mouse, human and pig. These results set the scene for therapeutic use of CSF-1 and IL-34 in cats.


Subject(s)
Interleukins/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Receptors, Colony-Stimulating Factor/genetics , Amino Acid Sequence , Animals , Blotting, Western , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cats , Cloning, Molecular , DNA, Complementary/genetics , Humans , Male , Mice , Models, Molecular , Molecular Sequence Data , Phagocytosis/drug effects , Receptors, Colony-Stimulating Factor/chemistry , Receptors, Colony-Stimulating Factor/metabolism , Recombinant Proteins/pharmacology , Species Specificity , Sus scrofa
7.
Cytokine ; 60(3): 793-805, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22974529

ABSTRACT

Macrophage Colony Stimulating Factor (CSF-1) controls the survival, differentiation and proliferation of cells of the mononuclear phagocyte system. A second ligand for the CSF-1R, Interleukin 34 (IL-34), has been described, but its physiological role is not yet known. The domestic pig provides an alternative to traditional rodent models for evaluating potential therapeutic applications of CSF-1R agonists and antagonists. To enable such studies, we cloned and expressed active pig CSF-1. To provide a bioassay, pig CSF-1R was expressed in the factor-dependent Ba/F3 cell line. On this transfected cell line, recombinant porcine CSF-1 and human CSF-1 had identical activity. Mouse CSF-1 does not interact with the human CSF-1 receptor but was active on pig. By contrast, porcine CSF-1 was active on mouse, human, cat and dog cells. IL-34 was previously shown to be species-specific, with mouse and human proteins demonstrating limited cross-species activity. The pig CSF-1R was equally responsive to both mouse and human IL-34. Based upon the published crystal structures of CSF-1/CSF-1R and IL34/CSF-1R complexes, we discuss the molecular basis for the species specificity.


Subject(s)
Interleukins/immunology , Macrophage Colony-Stimulating Factor/immunology , Macrophage Colony-Stimulating Factor/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Amino Acid Sequence , Animals , Cats , Cell Line , Cloning, Molecular , Dogs , HEK293 Cells , Humans , Macrophage Colony-Stimulating Factor/chemistry , Macrophage Colony-Stimulating Factor/genetics , Macrophages/cytology , Macrophages/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Signal Transduction , Species Specificity , Swine
8.
PLoS One ; 6(1): e15723, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21249125

ABSTRACT

BACKGROUND: The ten mouse and six human members of the Schlafen (Slfn) gene family all contain an AAA domain. Little is known of their function, but previous studies suggest roles in immune cell development. In this report, we assessed Slfn regulation and function in macrophages, which are key cellular regulators of innate immunity. METHODOLOGY/PRINCIPAL FINDINGS: Multiple members of the Slfn family were up-regulated in mouse bone marrow-derived macrophages (BMM) by the Toll-like Receptor (TLR)4 agonist lipopolysaccharide (LPS), the TLR3 agonist Poly(I∶C), and in disease-affected joints in the collagen-induced model of rheumatoid arthritis. Of these, the most inducible was Slfn4. TLR agonists that signal exclusively through the MyD88 adaptor protein had more modest effects on Slfn4 mRNA levels, thus implicating MyD88-independent signalling and autocrine interferon (IFN)-ß in inducible expression. This was supported by the substantial reduction in basal and LPS-induced Slfn4 mRNA expression in IFNAR-1⁻/⁻ BMM. LPS causes growth arrest in macrophages, and other Slfn family genes have been implicated in growth control. Slfn4 mRNA levels were repressed during macrophage colony-stimulating factor (CSF-1)-mediated differentiation of bone marrow progenitors into BMM. To determine the role of Slfn4 in vivo, we over-expressed the gene specifically in macrophages in mice using a csf1r promoter-driven binary expression system. Transgenic over-expression of Slfn4 in myeloid cells did not alter macrophage colony formation or proliferation in vitro. Monocyte numbers, as well as inflammatory macrophages recruited to the peritoneal cavity, were reduced in transgenic mice that specifically over-expressed Slfn4, while macrophage numbers and hematopoietic activity were increased in the livers and spleens. CONCLUSIONS: Slfn4 mRNA levels were up-regulated during macrophage activation but down-regulated during differentiation. Constitutive Slfn4 expression in the myeloid lineage in vivo perturbs myelopoiesis. We hypothesise that the down-regulation of Slfn4 gene expression during macrophage differentiation is a necessary step in development of this lineage.


Subject(s)
Carrier Proteins/genetics , Macrophage Activation , Macrophages/cytology , Myelopoiesis , Signal Transduction , Animals , Carrier Proteins/physiology , Cell Cycle/immunology , Gene Expression Regulation/immunology , Immunity, Innate , Macrophages/immunology , Mice , Mice, Transgenic , RNA, Messenger/analysis , Signal Transduction/genetics , Signal Transduction/immunology , Signal Transduction/physiology
9.
J Leukoc Biol ; 89(6): 823-36, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21233413

ABSTRACT

The nucleotide exchange factor cytohesin-1 was previously reported to interact with the cytoplasmic domains of the integrin ß-chain common to all ß(2) integrins such as LFA-1 and Mac-1. We show here that cytohesin-1, which contributes to fMLF-induced functional responses in PMNs through activation of Arf6, restrains the activation of the ß(2) integrin Mac-1 (αMß(2)) in PMNs or dcAMP-differentiated PLB-985 cells. We found that the cytohesin-1 inhibitor SecinH3 or siRNA increased cell adhesion to immobilized fibrinogen and fMLF-mediated conformational changes of Mac-1, monitored using mAb CBRM1/5, specific for the activation epitope of the αM subunit. In contrast, PLB-985 cells overexpressing cytohesin-1 showed little adhesion to fibrinogen. The use of SecinH3 and siRNA also revealed that interference with cytohesin-1 signaling also enhanced phagocytosis of zymosan particles and chemotaxis toward fMLF in transwell migration assays. These increments of phagocytosis and chemotaxis in cells treated with SecinH3 and cytohesin-1 siRNA were reversed by a blocking mAb to the integrin-αM subunit. We provide evidence for increased polymerized cortical actin in cells treated with SecinH3 and that altered signaling through cytohesin-1 increased cell surface expression of FPRL-1 and impairs the late calcium mobilization response elicited by fMLF. The data provide evidence that stimulation with fMLF initiates a signaling cascade that restrains Mac-1 activation in PMNs. Such crosstalk between FPRL-1 and Mac-1 involves cytohesin-1. We suggest that cytohesin-1 may coordinate activation of the ß(2) integrins to regulate PMN adhesion, phagocytosis, and chemotaxis.


Subject(s)
CD18 Antigens/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Macrophage-1 Antigen/chemistry , Macrophage-1 Antigen/metabolism , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , Actins/metabolism , Adult , Blotting, Western , Calcium/metabolism , Cell Adhesion , Cell Differentiation , Cell Movement , Chemotaxis , Fibrinogen/metabolism , Flow Cytometry , Humans , Immunoprecipitation , Lymphocyte Function-Associated Antigen-1/metabolism , Neutrophil Activation/drug effects , Phagocytosis , Protein Conformation/drug effects
10.
J Leukoc Biol ; 87(5): 753-64, 2010 May.
Article in English | MEDLINE | ID: mdl-20051473

ABSTRACT

Macrophages are involved in many aspects of development, host defense, pathology, and homeostasis. Their normal differentiation, proliferation, and survival are controlled by CSF-1 via the activation of the CSF1R. A recently discovered cytokine, IL-34, was shown to bind the same receptor in humans. Chicken is a widely used model organism in developmental biology, but the factors that control avian myelopoiesis have not been identified previously. The CSF-1, IL-34, and CSF1R genes in chicken and zebra finch were identified from respective genomic/cDNA sequence resources. Comparative analysis of the avian CSF1R loci revealed likely orthologs of mammalian macrophage-specific promoters and enhancers, and the CSF1R gene is expressed in the developing chick embryo in a pattern consistent with macrophage-specific expression. Chicken CSF-1 and IL-34 were expressed in HEK293 cells and shown to elicit macrophage growth from chicken BM cells in culture. Comparative sequence and co-evolution analysis across all vertebrates suggests that the two ligands interact with distinct regions of the CSF1R. These studies demonstrate that there are two separate ligands for a functional CSF1R across all vertebrates.


Subject(s)
Interleukins/genetics , Macrophage Colony-Stimulating Factor/genetics , Macrophages/metabolism , Receptor, Macrophage Colony-Stimulating Factor/genetics , Amino Acid Sequence , Animals , Biological Evolution , Birds , Chick Embryo , Chickens , Conserved Sequence , Finches , Gene Expression , Humans , In Situ Hybridization , Interleukins/chemistry , Interleukins/metabolism , Macrophage Colony-Stimulating Factor/chemistry , Macrophage Colony-Stimulating Factor/metabolism , Molecular Sequence Data , Phylogeny , Protein Structure, Quaternary , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Sequence Homology, Amino Acid , Structural Homology, Protein
11.
J Immunol ; 184(2): 637-49, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20018626

ABSTRACT

Polymorphonuclear neutrophil (PMN) stimulation with fMLP stimulates small G proteins such as ADP-ribosylation factors (Arfs) Arf1 and Arf6, leading to phospholipase D (PLD) activation and functions such as degranulation and the oxidative burst. However, the molecular links between fMLF receptors and PLD remain unclear. PMNs express cytohesin-1, an Arf-guanine exchange factor that activates Arfs, and its expression is strongly induced during the acquisition of the neutrophilic phenotype by neutrophil-like cells. The role of cytohesin-1 in the activation of the fMLF-Arf-PLD signaling axis, and the accomplishment of superoxide anion production, and degranulation was investigated in PMNs using the selective inhibitor of cytohesin, Sec 7 inhibitor H3 (secinH3). Cytohesin-1 inhibition with secinH3 leads to Arf6 but not Arf1 inhibition, demonstrating the specificity for Arf6, and fMLF-mediated activation of PLD and of the oxidative burst as well. We observed a decrease in fMLF-mediated protein secretion and expression of cell surface markers corresponding to primary (CD63/myeloperoxidase), secondary (CD66/lactoferrin), and tertiary (matrix metalloproteinase-9) granules in PMNs incubated with secinH3. Similarly, silencing cytohesin-1 or Arf6 in PLB-985 cells negatively affected fMLF-induced activation of PLD, superoxide production, and expression of granule markers on the cell surface. In contrast, stable overexpression of cytohesin-1 in PLB-985 cells enhanced fMLF-induced activation of Arf6, PLD, and NADPH oxidase. The results of this study provide evidence for an involvement of cytohesin-1 in the regulation of the functional responses of human PMNs and link these events, in part at least, to the activation of Arf6.


Subject(s)
ADP-Ribosylation Factors/metabolism , Guanine Nucleotide Exchange Factors/physiology , Neutrophils/metabolism , Phospholipase D/metabolism , Superoxides/metabolism , ADP-Ribosylation Factor 6 , Cell Degranulation/drug effects , Cells, Cultured , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Humans , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , NADPH Oxidases/metabolism , Triazoles/pharmacology
12.
Science ; 323(5917): 1057-60, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19131592

ABSTRACT

The mammalian innate immune system is activated by foreign nucleic acids. Detection of double-stranded DNA (dsDNA) in the cytoplasm triggers characteristic antiviral responses and macrophage cell death. Cytoplasmic dsDNA rapidly activated caspase 3 and caspase 1 in bone marrow-derived macrophages. We identified the HIN-200 family member and candidate lupus susceptibility factor, p202, as a dsDNA binding protein that bound stably and rapidly to transfected DNA. Knockdown studies showed p202 to be an inhibitor of DNA-induced caspase activation. Conversely, the related pyrin domain-containing HIN-200 factor, AIM2 (p210), was required for caspase activation by cytoplasmic dsDNA. This work indicates that HIN-200 proteins can act as pattern recognition receptors mediating responses to cytoplasmic dsDNA.


Subject(s)
Caspase 1/metabolism , Caspase 3/metabolism , Cytoplasm/metabolism , DNA/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Membrane Proteins/metabolism , Receptors, Pattern Recognition/metabolism , Animals , Cell Line , DNA/immunology , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , Enzyme Activation , Immunity, Innate , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/isolation & purification , Macrophages/immunology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Inbred Strains , RNA, Small Interfering , Symporters , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...