Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Am J Obstet Gynecol ; 2024 Jun 11.
Article En | MEDLINE | ID: mdl-38871238

BACKGROUND: In recent years, pragmatic metformin use in pregnancy has stretched to include prediabetes, type 2 diabetes, gestational diabetes and (most recently) pre-eclampsia. With its expanded use, however, concerns of unintended harm have been raised. OBJECTIVE: We developed an experimental primate model and applied triple-quadruple pole LC mass spectrometry (UHPLC-QQQ) for direct quantitation of maternal and fetal tissue metformin levels with detailed fetal biometry and histopathology. STUDY DESIGN: Within 30 days of confirmed conception (defined as early pregnancy), n=13 time-bred (TMB) Rhesus dams with gestations designated for fetal necropsy were initiated on twice daily human dose-equivalent 10 mg/kg metformin or vehicle control. Pregnant dams were maintained as pairs and fed either a control chow or 36% fat Western-style diet (WSD). Metformin or placebo vehicle control were delivered in a variety of treats while animals were separated via a slide. A Cesarean was performed at G145, and amniotic fluid and blood were collected and the fetus and placenta were delivered. The fetus was immediately necropsied by trained primate center personnel. All fetal organs were dissected, measured, sectioned, and processed per clinical standards. Fluid and tissue metformin levels were assayed using validated UHPLC-QQQ in SRM against standard curves. RESULTS: Among the n=13 G145 pregnancies with fetal necropsy, n=1 dam and its fetal tissues had detectable metformin levels despite being allocated to the vehicle control group (>1 µM metformin/kg maternal weight or fetal/placental tissue), while a second fetus allocated to the vehicle control group had severe fetal growth restriction (birthweight 248.32 g, <1%) and was suspected of having a fetal congenital condition. After excluding these two fetal gestations from further analyses, 11 fetuses from dams initiated on either vehicle control (n=4, 3 female, 1 male fetuses) or 10 mg/kg metformin (n=7, 5 female, 2 male fetuses) were available for analyses. Among dams initiated on metformin by G30 (regardless of maternal diet), we observed significant bioaccumulation within the fetal kidney (0.78-6.06 µmol/kg, mean 2.48 µmol/kg) , liver (0.16-0.73 µmol/kg, mean 0.38 µmol/kg), fetal gut (0.28-1.22 µmol/kg, mean 0.70 µmol/kg), amniotic fluid (0.43-3.33 µmol/L, mean 1.88 µmol/L), placenta (0.16-1.0 µmol/kg , mean 0.50 µmol/kg) and fetal serum (0 -0.66 µmol/L , mean 0.23 µmol/L ), and fetal urine (4.1-174.1 µmol/L mean 38.5 µmol/L ), with fetal levels near biomolar equivalent to maternal levels (maternal serum 0.18-0.86 µmol/L , mean 0.46 µmol/L; maternal urine 42.6-254.0 µmol/L , mean 149.3 µmol/L). WSD feeding neither accelerated nor reduced metformin bioaccumulations in maternal or fetal serum, urine, amniotic fluid, placenta nor fetal tissues. In these 11 animals, fetal bioaccumulation of metformin was associated with less fetal skeletal muscle (57% lower cross-sectional area of gastrocnemius) and decreased liver, heart, and retroperitoneal fat masses (p<0.05), collectively driving lower delivery weight (p<0.0001) without changing the crown-rump length. Sagittal sections of fetal kidneys demonstrated delayed maturation, with disorganized glomerular generations and increased cortical thickness; this renal dysmorphology was not accompanied by structural nor functional changes indicative of renal insufficiency. CONCLUSIONS: We demonstrate fetal bioaccumulation of metformin with associated fetal growth restriction and renal dysmorphology following maternal initiation of the drug within 30 days of conception in primates. Given these results and the prevalence of metformin use during pregnancy, additional investigation of any potential immediate and enduring effects of prenatal metformin use is warranted.

2.
Vet Immunol Immunopathol ; 266: 110681, 2023 Dec.
Article En | MEDLINE | ID: mdl-37992576

Macrophage colony-stimulating factor (CSF1) controls the proliferation and differentiation of cells of the mononuclear phagocyte system through binding to the receptor CSF1R. The expression and function of CSF1 has been well-studied in rodents and humans, but knowledge is lacking in other veterinary species. The development of a novel mouse anti-porcine CSF1 monoclonal antibody (mAb) facilitates the characterisation of this growth factor in pigs. Cell surface expression of CSF1 was confirmed on differentiated macrophage populations derived from blood and bone marrow monocytes, and on lung resident macrophages, the first species for this to be confirmed. However, monocytes isolated from blood and bone marrow lacked CSF1 expression. This species-specific mAb delivers the opportunity to further understanding of porcine myeloid cell biology. This is not only vital for the role of pigs as a model for human health, but also as a veterinary species of significant economic and agricultural importance.


Antibodies, Monoclonal , Macrophage Colony-Stimulating Factor , Swine , Mice , Animals , Humans , Macrophages , Monocytes , Mononuclear Phagocyte System/metabolism
3.
Vet Immunol Immunopathol ; 226: 110073, 2020 Aug.
Article En | MEDLINE | ID: mdl-32559524

Intestinal macrophages are the largest group of mononuclear phagocytes in the body and play a role in intestinal innate immunity, neuroimmune interactions and maintaining intestinal homeostasis. Conversely, they also are implicated in numerous pathologies of the gastrointestinal tract, such as postoperative ileus and inflammatory bowel disease. As a result, macrophages could be potential therapeutic targets. To date, there are limited studies on the morphology and distribution of macrophages in the equine gastrointestinal tract (GIT). The aim of this study was to identify the location and abundance of resident macrophages in the equine GIT using CD163 as an immunohistochemical marker. Tissue samples were obtained post-mortem from 14 sites along the gastrointestinal tracts of 10 horses free from gastrointestinal disease; sample sites extended from the stomach to the small colon. CD163+ve cells were present in all regions of the equine GIT from stomach to small colon. CD163+ve cells were also identified in all tissue layers of the intestinal wall, namely, mucosa, submucosa, muscularis externa (ME), myenteric plexus and serosa. Consistent with a proposed function in regulation of intestinal motility, CD163+ve cells were regularly distributed within the ME, with accumulations closely associated with the myenteric plexus and effector cells such as neurons and the interstitial cells of Cajal (ICC).


Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Gastrointestinal Tract/cytology , Gastrointestinal Tract/immunology , Macrophages/immunology , Receptors, Cell Surface/immunology , Animals , Colon/cytology , Colon/immunology , Female , Horses , Immunohistochemistry , Macrophages/pathology , Male , Stomach/cytology , Stomach/immunology
5.
Immunohorizons ; 2(1): 27-37, 2018 Jan 01.
Article En | MEDLINE | ID: mdl-30467554

Activated mouse macrophages metabolize arginine via NO synthase (NOS2) to produce NO as an antimicrobial effector. Published gene expression datasets provide little support for the activation of this pathway in human macrophages. Generation of NO requires the coordinated regulation of multiple genes. We have generated RNA-sequencing data from bone marrow-derived macrophages from representative rodent (rat), monogastric (pig and horse), and ruminant (sheep, goat, cattle, and water buffalo) species, and analyzed the expression of genes involved in arginine metabolism in response to stimulation with LPS. In rats, as in mice, LPS strongly induced Nos2, the arginine transporter Slc7a2, arginase 1 (Arg1), GTP cyclohydrolase (Gch1), and argininosuccinate synthase (Ass1). None of these responses was conserved across species. Only cattle and water buffalo showed substantial NOS2 induction. The species studied also differed in expression and regulation of arginase (ARG2, rather than ARG1), and amino acid transporters. Variation between species was associated with rapid promoter evolution. Differential induction of NOS2 and ARG2 between the ruminant species was associated with insertions of the Bov-A2 retrotransposon in the promoter region. Bov-A2 was shown to possess LPS-inducible enhancer activity in transfected RAW264.7 macrophages. Consistent with a function in innate immunity, NO production and arginine metabolism vary greatly between species and differences may contribute to pathogen host restriction.

6.
Front Immunol ; 9: 2246, 2018.
Article En | MEDLINE | ID: mdl-30327653

The F4/80 antigen, encoded by the Adgre1 locus, has been widely-used as a monocyte-macrophage marker in mice, but its value as a macrophage marker in other species is unclear, and has even been questioned. ADGRE1 is a seven transmembrane G protein-coupled receptor with an extracellular domain containing repeated Epidermal Growth Factor (EGF)-like calcium binding domains. Using a new monoclonal antibody, we demonstrated that ADGRE1 is a myeloid differentiation marker in pigs, absent from progenitors in bone marrow, highly-expressed in mature granulocytes, monocytes, and tissue macrophages and induced by macrophage colony-stimulating factor (CSF1) treatment in vivo. Based upon these observations, we utilized RNA-Seq to assess the expression of ADGRE1 mRNA in bone marrow or monocyte-derived macrophages (MDM) and alveolar macrophages from 8 mammalian species including pig, human, rat, sheep, goat, cow, water buffalo, and horse. ADGRE1 mRNA was expressed by macrophages in each species, with inter-species variation both in expression level and response to lipopolysaccharide (LPS) stimulation. Analysis of the RNA-Seq data also revealed additional exons in several species compared to current Ensembl annotations. The ruminant species and horses appear to encode a complete duplication of the 7 EGF-like domains. In every species, Sashimi plots revealed evidence of exon skipping of the EGF-like domains, which are highly-variable between species and polymorphic in humans. Consistent with these expression patterns, key elements of the promoter and a putative enhancer are also conserved across all species. The rapid evolution of this molecule and related ADGRE family members suggests immune selection and a role in pathogen recognition.


Antigens, Differentiation/genetics , Macrophages/physiology , Membrane Glycoproteins/genetics , Mucins/genetics , Receptors, G-Protein-Coupled/genetics , Sus scrofa/genetics , Alternative Splicing , Animals , Antibodies, Monoclonal, Murine-Derived , Antigens, Differentiation/immunology , Base Sequence , Biomarkers , Bone Marrow Cells/cytology , Calcium-Binding Proteins , Cell Differentiation/physiology , Cells, Cultured , Epidermal Growth Factor/genetics , Exons , Female , Gene Expression , HEK293 Cells , Humans , Membrane Glycoproteins/immunology , Mice , Mucins/immunology , Receptors, G-Protein-Coupled/immunology , Transcription, Genetic
7.
J Immunol ; 201(9): 2683-2699, 2018 11 01.
Article En | MEDLINE | ID: mdl-30249809

We have produced Csf1r-deficient rats by homologous recombination in embryonic stem cells. Consistent with the role of Csf1r in macrophage differentiation, there was a loss of peripheral blood monocytes, microglia in the brain, epidermal Langerhans cells, splenic marginal zone macrophages, bone-associated macrophages and osteoclasts, and peritoneal macrophages. Macrophages of splenic red pulp, liver, lung, and gut were less affected. The pleiotropic impacts of the loss of macrophages on development of multiple organ systems in rats were distinct from those reported in mice. Csf1r-/- rats survived well into adulthood with postnatal growth retardation, distinct skeletal and bone marrow abnormalities, infertility, and loss of visceral adipose tissue. Gene expression analysis in spleen revealed selective loss of transcripts associated with the marginal zone and, in brain regions, the loss of known and candidate novel microglia-associated transcripts. Despite the complete absence of microglia, there was little overt phenotype in brain, aside from reduced myelination and increased expression of dopamine receptor-associated transcripts in striatum. The results highlight the redundant and nonredundant functions of CSF1R signaling and of macrophages in development, organogenesis, and homeostasis.


Macrophages , Microglia , Organogenesis/genetics , Rats/growth & development , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Animals , Models, Animal , Mutation , Rats/genetics
8.
Nat Commun ; 9(1): 1272, 2018 03 28.
Article En | MEDLINE | ID: mdl-29593242

Colony-stimulating factor 1 (CSF1) controls the growth and differentiation of macrophages.CSF1R signaling has been implicated in the maintenance of the intestinal stem cell niche and differentiation of Paneth cells, but evidence of expression of CSF1R within the crypt is equivocal. Here we show that CSF1R-dependent macrophages influence intestinal epithelial differentiation and homeostasis. In the intestinal lamina propria CSF1R mRNA expression is restricted to macrophages which are intimately associated with the crypt epithelium, and is undetectable in Paneth cells. Macrophage ablation following CSF1R blockade affects Paneth cell differentiation and leads to a reduction of Lgr5+ intestinal stem cells. The disturbances to the crypt caused by macrophage depletion adversely affect the subsequent differentiation of intestinal epithelial cell lineages. Goblet cell density is enhanced, whereas the development of M cells in Peyer's patches is impeded. We suggest that modification of the phenotype or abundance of macrophages in the gut wall alters the development of the intestinal epithelium and the ability to sample gut antigens.


Intestinal Mucosa/metabolism , Macrophages/cytology , Mucous Membrane/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Animals , Cell Differentiation , Cell Lineage , Female , Goblet Cells/metabolism , Green Fluorescent Proteins/metabolism , Homeostasis , Intestines , Macrophage Colony-Stimulating Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Paneth Cells/cytology , Peyer's Patches/cytology , Phenotype , RNA, Messenger/metabolism , Signal Transduction , Stem Cell Niche , Stem Cells/cytology
9.
J Immunol ; 200(6): 2209-2223, 2018 03 15.
Article En | MEDLINE | ID: mdl-29440354

CSF1 is the primary growth factor controlling macrophage numbers, but whether expression of the CSF1 receptor differs between discrete populations of mononuclear phagocytes remains unclear. We have generated a Csf1r-mApple transgenic fluorescent reporter mouse that, in combination with lineage tracing, Alexa Fluor 647-labeled CSF1-Fc and CSF1, and a modified ΔCsf1-enhanced cyan fluorescent protein (ECFP) transgene that lacks a 150 bp segment of the distal promoter, we have used to dissect the differentiation and CSF1 responsiveness of mononuclear phagocyte populations in situ. Consistent with previous Csf1r-driven reporter lines, Csf1r-mApple was expressed in blood monocytes and at higher levels in tissue macrophages, and was readily detectable in whole mounts or with multiphoton microscopy. In the liver and peritoneal cavity, uptake of labeled CSF1 largely reflected transgene expression, with greater receptor activity in mature macrophages than monocytes and tissue-specific expression in conventional dendritic cells. However, CSF1 uptake also differed between subsets of monocytes and discrete populations of tissue macrophages, which in macrophages correlated with their level of dependence on CSF1 receptor signaling for survival rather than degree of transgene expression. A double ΔCsf1r-ECFP-Csf1r-mApple transgenic mouse distinguished subpopulations of microglia in the brain, and permitted imaging of interstitial macrophages distinct from alveolar macrophages, and pulmonary monocytes and conventional dendritic cells. The Csf1r-mApple mice and fluorescently labeled CSF1 will be valuable resources for the study of macrophage and CSF1 biology, which are compatible with existing EGFP-based reporter lines.


Mononuclear Phagocyte System/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Transgenes/genetics , Animals , Cell Differentiation/genetics , Dendritic Cells/metabolism , Green Fluorescent Proteins/genetics , Macrophage Colony-Stimulating Factor/genetics , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Monocytes/metabolism , Signal Transduction/genetics
10.
Am J Physiol Gastrointest Liver Physiol ; 314(3): G388-G398, 2018 03 01.
Article En | MEDLINE | ID: mdl-29351395

Signaling via the colony-stimulating factor 1 receptor (CSF1R) controls the survival, differentiation, and proliferation of macrophages. Mutations in CSF1 or CSF1R in mice and rats have pleiotropic effects on postnatal somatic growth. We tested the possible application of pig CSF1-Fc fusion protein as a therapy for low birth weight (LBW) at term, using a model based on maternal dexamethasone treatment in rats. Neonatal CSF1-Fc treatment did not alter somatic growth and did not increase the blood monocyte count. Instead, there was a substantial increase in the size of liver in both control and LBW rats, and the treatment greatly exacerbated lipid droplet accumulation seen in the dexamethasone LBW model. These effects were reversed upon cessation of treatment. Transcriptional profiling of the livers supported histochemical evidence of a large increase in macrophages with a resident Kupffer cell phenotype and revealed increased expression of many genes implicated in lipid droplet formation. There was no further increase in hepatocyte proliferation over the already high rates in neonatal liver. In conclusion, treatment of neonatal rats with CSF1-Fc caused an increase in liver size and hepatic lipid accumulation, due to Kupffer cell expansion and/or activation rather than hepatocyte proliferation. Increased liver macrophage numbers and expression of endocytic receptors could mitigate defective clearance functions in neonates. NEW & NOTEWORTHY This study is based on extensive studies in mice and pigs of the role of CSF1/CSF1R in macrophage development and postnatal growth. We extended the study to neonatal rats as a possible therapy for low birth weight. Unlike our previous studies in mice and pigs, there was no increase in hepatocyte proliferation and no increase in monocyte numbers. Instead, neonatal rats treated with CSF1 displayed reversible hepatic steatosis and Kupffer cell expansion.


Adiposity/drug effects , Cell Proliferation/drug effects , Fatty Liver/chemically induced , Fetal Growth Retardation/drug therapy , Kupffer Cells/drug effects , Lipid Metabolism/drug effects , Liver/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Animals , Animals, Newborn , Birth Weight , Cells, Cultured , Dexamethasone , Disease Models, Animal , Fatty Liver/metabolism , Fatty Liver/pathology , Female , Fetal Growth Retardation/chemically induced , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Kupffer Cells/metabolism , Kupffer Cells/pathology , Liver/growth & development , Liver/metabolism , Liver/pathology , Macrophage Colony-Stimulating Factor/toxicity , Male , Pregnancy , Rats, Sprague-Dawley , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/agonists , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction/drug effects , Spleen/drug effects , Spleen/metabolism , Spleen/pathology , Sus scrofa
11.
Gut ; 66(12): 2098-2109, 2017 12.
Article En | MEDLINE | ID: mdl-28615302

OBJECTIVE: Postoperative ileus (POI) is assumed to result from myeloid cells infiltrating the intestinal muscularis externa (ME) in patients undergoing abdominal surgery. In the current study, we investigated the role of infiltrating monocytes in a murine model of intestinal manipulation (IM)-induced POI in order to clarify whether monocytes mediate tissue damage and intestinal dysfunction or they are rather involved in the recovery of gastrointestinal (GI) motility. DESIGN: IM was performed in mice with defective monocyte migration to tissues (C-C motif chemokine receptor 2, Ccr2-/ - mice) and wild-type (WT) mice to study the role of monocytes and monocyte-derived macrophages (MΦs) during onset and resolution of ME inflammation. RESULTS: At early time points, IM-induced GI transit delay and inflammation were equal in WT and Ccr2 -/- mice. However, GI transit recovery after IM was significantly delayed in Ccr2 -/- mice compared with WT mice, associated with increased neutrophil-mediated immunopathology and persistent impaired neuromuscular function. During recovery, monocyte-derived MΦs acquire pro-resolving features that aided in the resolution of inflammation. In line, bone marrow reconstitution and treatment with MΦ colony-stimulating factor 1 enhanced monocyte recruitment and MΦ differentiation and ameliorated GI transit in Ccr2 -/- mice. CONCLUSION: Our study reveals a critical role for monocyte-derived MΦs in restoring intestinal homeostasis after surgical trauma. From a therapeutic point of view, our data indicate that inappropriate targeting of monocytes may increase neutrophil-mediated immunopathology and prolong the clinical outcome of POI, while future therapies should be aimed at enhancing MΦ physiological repair functions.


Ileus/immunology , Ileus/pathology , Macrophages/immunology , Monocytes/immunology , Postoperative Complications/immunology , Postoperative Complications/pathology , Receptors, CCR2/immunology , Animals , Cell Differentiation , Cell Movement , Disease Models, Animal , Gastrointestinal Motility , Gastrointestinal Transit , Homeostasis/immunology , Inflammation/immunology , Inflammation/pathology , Mice , Muscle, Smooth/pathology
12.
J Immunol ; 197(6): 2297-305, 2016 09 15.
Article En | MEDLINE | ID: mdl-27521343

Expression of Csf1r in adults is restricted to cells of the macrophage lineage. Transgenic reporters based upon the Csf1r locus require inclusion of the highly conserved Fms-intronic regulatory element for expression. We have created Csf1r-EGFP transgenic sheep via lentiviral transgenesis of a construct containing elements of the mouse Fms-intronic regulatory element and Csf1r promoter. Committed bone marrow macrophage precursors and blood monocytes express EGFP in these animals. Sheep monocytes were divided into three populations, similar to classical, intermediate, and nonclassical monocytes in humans, based upon CD14 and CD16 expression. All expressed EGFP, with increased levels in the nonclassical subset. Because Csf1r expression coincides with the earliest commitment to the macrophage lineage, Csf1r-EGFP bone marrow provides a tool for studying the earliest events in myelopoiesis using the sheep as a model.


Animals, Genetically Modified/immunology , Biomarkers/blood , Green Fluorescent Proteins/genetics , Macrophages/physiology , Monocytes/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Animals , Cell Differentiation , Humans , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/immunology , Macrophages/immunology , Mice , Myelopoiesis , Promoter Regions, Genetic , Receptors, IgG/genetics , Receptors, IgG/immunology , Sheep/genetics , Transgenes
13.
Am J Physiol Gastrointest Liver Physiol ; 311(3): G533-47, 2016 09 01.
Article En | MEDLINE | ID: mdl-27445344

Macrophage colony-stimulating factor (CSF1) is an essential growth and differentiation factor for cells of the macrophage lineage. To explore the role of CSF1 in steady-state control of monocyte production and differentiation and tissue repair, we previously developed a bioactive protein with a longer half-life in circulation by fusing pig CSF1 with the Fc region of pig IgG1a. CSF1-Fc administration to pigs expanded progenitor pools in the marrow and selectively increased monocyte numbers and their expression of the maturation marker CD163. There was a rapid increase in the size of the liver, and extensive proliferation of hepatocytes associated with increased macrophage infiltration. Despite the large influx of macrophages, there was no evidence of liver injury and no increase in circulating liver enzymes. Microarray expression profiling of livers identified increased expression of macrophage markers, i.e., cytokines such as TNF, IL1, and IL6 known to influence hepatocyte proliferation, alongside cell cycle genes. The analysis also revealed selective enrichment of genes associated with portal, as opposed to centrilobular regions, as seen in hepatic regeneration. Combined with earlier data from the mouse, this study supports the existence of a CSF1-dependent feedback loop, linking macrophages of the liver with bone marrow and blood monocytes, to mediate homeostatic control of the size of the liver. The results also provide evidence of safety and efficacy for possible clinical applications of CSF1-Fc.


Liver/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Monocytes/physiology , Swine , Animals , Antibodies , Antigens, CD , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Female , Gene Expression Regulation, Enzymologic , Leukocytes, Mononuclear/physiology , Male
14.
BMC Biol ; 13: 12, 2015 Feb 19.
Article En | MEDLINE | ID: mdl-25857347

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.


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
15.
J Immunol ; 194(5): 2338-44, 2015 Mar 01.
Article En | MEDLINE | ID: mdl-25637020

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.


Avian Proteins/immunology , Chickens/immunology , Gonads/immunology , Macrophages/immunology , RNA, Messenger/immunology , Sex Chromosomes/immunology , Animals , Aromatase Inhibitors/pharmacology , Avian Proteins/genetics , Cells, Cultured , Chick Embryo , Chickens/genetics , Dosage Compensation, Genetic , Fadrozole/pharmacology , Female , Gene Expression , Gene Expression Profiling , Gonads/drug effects , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Lipopolysaccharides/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/cytology , Macrophages/drug effects , Male , RNA, Messenger/genetics , Sex Characteristics
16.
PLoS One ; 9(8): e105429, 2014.
Article En | MEDLINE | ID: mdl-25137049

The MacBlue transgenic mouse uses the Csf1r promoter and first intron to drive expression of gal4-VP16, which in turn drives a cointegrated gal4-responsive UAS-ECFP cassette. The Csf1r promoter region used contains a deletion of a 150 bp conserved region covering trophoblast and osteoclast-specific transcription start sites. In this study, we examined expression of the transgene in embryos and adult mice. In embryos, ECFP was expressed in the large majority of macrophages derived from the yolk sac, and as the liver became a major site of monocytopoiesis. In adults, ECFP was detected at high levels in both Ly6C+ and Ly6C- monocytes and distinguished them from Ly6C+, F4/80+, CSF1R+ immature myeloid cells in peripheral blood. ECFP was also detected in the large majority of microglia and Langerhans cells. However, expression was lost from the majority of tissue macrophages, including Kupffer cells in the liver and F4/80+ macrophages of the lung, kidney, spleen and intestine. The small numbers of positive cells isolated from the liver resembled blood monocytes. In the gut, ECFP+ cells were identified primarily as classical dendritic cells or blood monocytes in disaggregated cell preparations. Immunohistochemistry showed large numbers of ECFP+ cells in the Peyer's patch and isolated lymphoid follicles. The MacBlue transgene was used to investigate the effect of treatment with CSF1-Fc, a form of the growth factor with longer half-life and efficacy. CSF1-Fc massively expanded both the immature myeloid cell (ECFP-) and Ly6C+ monocyte populations, but had a smaller effect on Ly6C- monocytes. There were proportional increases in ECFP+ cells detected in lung and liver, consistent with monocyte infiltration, but no generation of ECFP+ Kupffer cells. In the gut, there was selective infiltration of large numbers of cells into the lamina propria and Peyer's patches. We discuss the use of the MacBlue transgene as a marker of monocyte/macrophage/dendritic cell differentiation.


Biomarkers/metabolism , Kupffer Cells/metabolism , Langerhans Cells/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Monocytes/metabolism , Transgenes/genetics , Animals , Cell Differentiation/genetics , Female , Green Fluorescent Proteins/genetics , Intestinal Mucosa/metabolism , Kidney/metabolism , Langerhans Cells/drug effects , Liver/metabolism , Male , Mice , Mice, Transgenic/genetics , Microglia/metabolism , Peyer's Patches/metabolism , Receptor, Macrophage Colony-Stimulating Factor/genetics , Spleen/metabolism , Trans-Activators/genetics , Yolk Sac/metabolism
17.
Mol Ther ; 22(9): 1580-92, 2014 Sep.
Article En | MEDLINE | ID: mdl-24962162

We have produced an Fc conjugate of colony-stimulating factor (CSF) 1 with an improved circulating half-life. CSF1-Fc retained its macrophage growth-promoting activity, and did not induce proinflammatory cytokines in vitro. Treatment with CSF1-Fc did not produce adverse effects in mice or pigs. The impact of CSF1-Fc was examined using the Csf1r-enhanced green fluorescent protein (EGFP) reporter gene in MacGreen mice. Administration of CSF1-Fc to mice drove extensive infiltration of all tissues by Csf1r-EGFP positive macrophages. The main consequence was hepatosplenomegaly, associated with proliferation of hepatocytes. Expression profiles of the liver indicated that infiltrating macrophages produced candidate mediators of hepatocyte proliferation including urokinase, tumor necrosis factor, and interleukin 6. CSF1-Fc also promoted osteoclastogenesis and produced pleiotropic effects on other organ systems, notably the testis, where CSF1-dependent macrophages have been implicated in homeostasis. However, it did not affect other putative CSF1 targets, notably intestine, where Paneth cell numbers and villus architecture were unchanged. CSF1 has therapeutic potential in regenerative medicine in multiple organs. We suggest that the CSF1-Fc conjugate retains this potential, and may permit daily delivery by injection rather than continuous infusion required for the core molecule.


Hepatocytes/metabolism , Hepatomegaly/chemically induced , Immunoglobulin Fc Fragments/metabolism , Macrophage Colony-Stimulating Factor/administration & dosage , Macrophage Colony-Stimulating Factor/adverse effects , Splenomegaly/chemically induced , Swine/immunology , Animals , CHO Cells , Cell Proliferation , Cricetulus , Female , Gene Expression Regulation/drug effects , HEK293 Cells , Half-Life , Humans , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Regenerative Medicine
18.
J Leukoc Biol ; 96(2): 265-74, 2014 Aug.
Article En | MEDLINE | ID: mdl-24652541

We investigated the role of CSF1R signaling in adult mice using prolonged treatment with anti-CSF1R antibody. Mutation of the CSF1 gene in the op/op mouse produces numerous developmental abnormalities. Mutation of the CSF1R has an even more penetrant phenotype, including perinatal lethality, because of the existence of a second ligand, IL-34. These effects on development provide limited insight into functions of CSF1R signaling in adult homeostasis. The carcass weight and weight of several organs (spleen, kidney, and liver) were reduced in the treated mice, but overall body weight gain was increased. Despite the complete loss of Kupffer cells, there was no effect on liver gene expression. The treatment ablated OCL, increased bone density and trabecular volume, and prevented the decline in bone mass seen in female mice with age. The op/op mouse has a deficiency in pancreatic ß cells and in Paneth cells in the gut wall. Only the latter was reproduced by the antibody treatment and was associated with increased goblet cell number but no change in villus architecture. Male op/op mice are infertile as a result of testosterone insufficiency. Anti-CSF1R treatment ablated interstitial macrophages in the testis, but there was no sustained effect on testosterone or LH. The results indicate an ongoing requirement for CSF1R signaling in macrophage and OCL homeostasis but indicate that most effects of CSF1 and CSF1R mutations are due to effects on development.


Aging/immunology , Homeostasis/immunology , Receptor, Macrophage Colony-Stimulating Factor/immunology , Signal Transduction/immunology , Aging/genetics , Aging/pathology , Animals , Female , Goblet Cells/immunology , Goblet Cells/pathology , Homeostasis/genetics , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Interleukins/genetics , Interleukins/immunology , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Mutant Strains , Mutation , Paneth Cells/immunology , Paneth Cells/pathology , Receptor, Macrophage Colony-Stimulating Factor/genetics , Sex Characteristics , Signal Transduction/genetics , Testis/immunology , Testis/pathology
19.
Sci Rep ; 3: 3013, 2013 Oct 22.
Article En | MEDLINE | ID: mdl-24145216

Hereditary diffuse leukoencephalopathy with spheroids (HDLS) in humans is a rare autosomal dominant disease characterized by giant neuroaxonal swellings (spheroids) within the CNS white matter. Symptoms are variable and can include personality and behavioural changes. Patients with this disease have mutations in the protein kinase domain of the colony-stimulating factor 1 receptor (CSF1R) which is a tyrosine kinase receptor essential for microglia development. We investigated the effects of these mutations on Csf1r signalling using a factor dependent cell line. Corresponding mutant forms of murine Csf1r were expressed on the cell surface at normal levels, and bound CSF1, but were not able to sustain cell proliferation. Since Csf1r signaling requires receptor dimerization initiated by CSF1 binding, the data suggest a mechanism for phenotypic dominance of the mutant allele in HDLS.


Gliosis/congenital , Leukoencephalopathies/genetics , Leukoencephalopathies/metabolism , Mutation , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Animals , Cell Line , Cell Survival/genetics , Gene Expression , Genetic Association Studies , Gliosis/genetics , Gliosis/metabolism , Humans , Mice , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Transport , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Signal Transduction
20.
Berl Munch Tierarztl Wochenschr ; 126(5-6): 220-9, 2013.
Article En | MEDLINE | ID: mdl-23758037

This study was to identify risk factors for bovine spongiform encephalopathy (BSE) by means of individual case-control data. 43 BSE cases in a defined region in Lower Saxony and Schleswig-Holstein were compared with 84 control animals. Purchase of new breeding stock and cross contamination between feed on the farm did not seem to have influence on the BSE incidence in these regions. The results indicate independent risk patterns. Pattern 1: Cows with high milk yield seemed to be at risk on big farms with adjacent pig production and when they were not fed milk replacer. Pattern 2: Milk replacer (esp. from certain producers) is a risk factor for Non-Red Holstein cattle, low yielding cows and farms without pig production. Pattern 3: Red Holstein cattle not being fed milk replacer have a higher BSE risk than other breeds when they have a low milk yield and live on small farms with pig production. This study, like findings in Bavaria, Lower Saxony and Schleswig-Holstein, strengthens the hypothesis that BSE in Germany was caused by a feed mediated ubiquitous exposure to PrP(sc) during a confined time period. Producers, in need of buying animal derived feed components during that time slot, were more likely to spread the PrP(sc) than others. Their increased risk is not necessarily due to an inadequate purchasing policy, but can also be coincidental. The breed Red Holstein is not the risk factor itself but represents the risk from concentrated feed for animals during a susceptible age period (calves). Therefore, the authors suggest a continuous exclusion of animal-derived fat components from milk replacers.


Encephalopathy, Bovine Spongiform/epidemiology , Age Factors , Analysis of Variance , Animal Feed/adverse effects , Animals , Breeding , Case-Control Studies , Cattle , Encephalopathy, Bovine Spongiform/etiology , Encephalopathy, Bovine Spongiform/physiopathology , Factor Analysis, Statistical , Female , Germany/epidemiology , Lactation , Milk/metabolism , Risk Factors
...