Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
Add more filters

Publication year range
1.
Am J Hum Genet ; 104(5): 925-935, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30982609

ABSTRACT

Colony stimulating factor 1 receptor (CSF1R) plays key roles in regulating development and function of the monocyte/macrophage lineage, including microglia and osteoclasts. Mono-allelic mutations of CSF1R are known to cause hereditary diffuse leukoencephalopathy with spheroids (HDLS), an adult-onset progressive neurodegenerative disorder. Here, we report seven affected individuals from three unrelated families who had bi-allelic CSF1R mutations. In addition to early-onset HDLS-like neurological disorders, they had brain malformations and skeletal dysplasia compatible to dysosteosclerosis (DOS) or Pyle disease. We identified five CSF1R mutations that were homozygous or compound heterozygous in these affected individuals. Two of them were deep intronic mutations resulting in abnormal inclusion of intron sequences in the mRNA. Compared with Csf1r-null mice, the skeletal and neural phenotypes of the affected individuals appeared milder and variable, suggesting that at least one of the mutations in each affected individual is hypomorphic. Our results characterized a unique human skeletal phenotype caused by CSF1R deficiency and implied that bi-allelic CSF1R mutations cause a spectrum of neurological and skeletal disorders, probably depending on the residual CSF1R function.


Subject(s)
Brain/abnormalities , Leukoencephalopathies/etiology , Mutation , Osteochondrodysplasias/etiology , Osteosclerosis/etiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Adolescent , Adult , Alleles , Animals , Brain/metabolism , Brain/pathology , Child, Preschool , Female , Humans , Leukoencephalopathies/pathology , Male , Mice , Mice, Knockout , Osteochondrodysplasias/pathology , Osteosclerosis/pathology , Phenotype , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Young Adult
2.
Int J Mol Sci ; 22(14)2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34298983

ABSTRACT

The use of autologous tolerogenic dendritic cells (tolDC) has become a promising strategy to re-establish immune tolerance in autoimmune diseases. Among the different strategies available, the use of vitamin D3 for the generation of tolDC (VitD3-tolDC) has been widely tested because of their immune regulatory properties. To identify molecules and pathways involved in the generation of VitD3-tolDC, we established an easy and fast gene silencing method based on the use of Viromer blue to introduce siRNA into monocytes on day 1 of culture differentiation. The analysis of the effect of CD209 (DC-SIGN) and CD115 (CSF1R) down-modulation on the phenotype and functionality of transfected VitD3-tolDC revealed a partial role of CD115 in their tolerogenicity. Further investigations showed that CSF1R-CSF1 signaling is involved in the induction of cell metabolic reprogramming, triggering glycolysis to produce high amounts of lactate, a novel suppressive mechanism of T cell proliferation, recently found in autologous tolerogenic dendritic cells (ATDCs).


Subject(s)
Cholecalciferol/pharmacology , Dendritic Cells/immunology , Glycolysis/physiology , Immune Tolerance/genetics , Leukocytes, Mononuclear/immunology , Macrophage Colony-Stimulating Factor/physiology , Monocytes/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned , Dendritic Cells/drug effects , Glucose/metabolism , Humans , Hydrogen-Ion Concentration , Interleukins/pharmacology , Lactates/metabolism , Signal Transduction , Transfection
3.
Blood ; 130(16): 1809-1818, 2017 10 19.
Article in English | MEDLINE | ID: mdl-28807982

ABSTRACT

The transcription factor CCAAT/enhancer-binding protein ß (C/EBPß) is highly expressed in monocytes/macrophages. However, its roles in monopoiesis are largely unknown. Here, we investigated the roles of C/EBPß in monopoiesis. Further subdivision of monocytes revealed that Cebpb messenger RNA was highly upregulated in Ly6C- monocytes in bone marrow. Accordingly, the number of Ly6C- monocytes was significantly reduced in Cebpb-/- mice. Bone marrow chimera experiments and Mx1-Cre-mediated deletion of Cebpb revealed a cell-intrinsic and monocyte-specific requirement for C/EBPß in monopoiesis. In Cebpb-/- mice, turnover of Ly6C- monocytes was highly accelerated and apoptosis of Ly6C- monocytes was increased. Expression of Csf1r, which encodes a receptor for macrophage colony-stimulating factor, was significantly reduced in Ly6C- monocytes of Cebpb-/- mice. C/EBPß bound to positive regulatory elements of Csf1r and promoted its transcription. Collectively, these results indicate that C/EBPß is a critical factor for Ly6C- monocyte survival, at least in part through upregulation of Csf1r.


Subject(s)
Apoptosis/genetics , CCAAT-Enhancer-Binding Protein-beta/physiology , Monocytes/physiology , Animals , Antigens, Ly/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , COS Cells , Cell Differentiation/genetics , Cell Survival/genetics , Cells, Cultured , Chlorocebus aethiops , Gene Expression Regulation , Mice , Mice, Knockout , Monocytes/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology
4.
Brain Behav Immun ; 73: 682-697, 2018 10.
Article in English | MEDLINE | ID: mdl-30056204

ABSTRACT

Microglia are the resident immune cells in the central nervous system (CNS). Originally thought to be primarily responsible for disposing of cellular debris and responding to neural insults, emerging research now shows that microglia are highly dynamic cells involved in a variety of neurodevelopmental processes. The hypothalamus is a brain region critical for maintaining homeostatic processes such as energy balance, thirst, food intake, reproduction, and circadian rhythms. Given that microglia colonize the embryonic brain alongside key steps of hypothalamic development, here we tested whether microglia are required for the proper establishment of this brain region. The Colony-stimulating factor-1 receptor (Csf1r) is expressed by microglia, macrophages and osteoclasts, and is required for their proliferation, differentiation, and survival. Therefore, to eliminate microglia from the fetal brain, we treated pregnant dams with the CSF1R inhibitor PLX5622. We showed that approximately 99% of microglia were eliminated by embryonic day 15.5 (E15.5) after pregnant dams were placed on a PLX5622 diet starting at E3.5. Following microglia depletion, we observed elevated numbers of apoptotic cells accumulating throughout the developing hypothalamus. Once the PLX5622 diet was removed, microglia repopulated the postnatal brain within 7 days and did not appear to repopulate from Nestin+ precursors. Embryonic microglia depletion also resulted in a decreased litter size, as well as an increase in the number of pups that died within the first two postnatal days of life. In pups that survived, the elimination of microglia in the fetal brain resulted in a decrease in the number of Pro-opiomelanocortin (POMC) neurons and a concomitant accelerated weight gain starting at postnatal day 5 (P5), suggesting that microglia could be important for the development of cell types key to hypothalamic satiety centers. Moreover, surviving PLX5622 exposed animals displayed craniofacial and dental abnormalities, perhaps due to non-CNS effects of PLX5622 on macrophages and/or osteoclasts. Finally, depletion of microglia during embryogenesis had long-term sex-specific effects on behaviour, including the development of hyperactivity and anxiolytic-like behaviour in juvenile and adult female mice, respectively. Together, these data demonstrate an important role for microglia during the development of the embryonic hypothalamus, and perhaps the CNS more broadly.


Subject(s)
Microglia/drug effects , Microglia/metabolism , Organic Chemicals/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Brain/metabolism , Embryo, Mammalian , Female , Hyperkinesis/physiopathology , Macrophages/metabolism , Male , Mice , Microglia/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Sex Factors , Weight Gain/physiology
5.
Brain Behav Immun ; 55: 179-190, 2016 07.
Article in English | MEDLINE | ID: mdl-26541819

ABSTRACT

Neurogenesis is altered in neurodegenerative disorders, partly regulated by inflammatory factors. We have investigated whether microglia, the innate immune brain cells, regulate hippocampal neurogenesis in neurodegeneration. Using the ME7 model of prion disease we applied gain- or loss-of CSF1R function, as means to stimulate or inhibit microglial proliferation, respectively, to dissect the contribution of these cells to neurogenesis. We found that increased hippocampal neurogenesis correlates with the expansion of the microglia population. The selective inhibition of microglial proliferation caused a reduction in neurogenesis and a restoration of normal neuronal differentiation, supporting a pro-neurogenic role for microglia. Using a gene screening strategy, we identified TGFß as a molecule controlling the microglial pro-neurogenic response in chronic neurodegeneration, supported by loss-of-function mechanistic experiments. By the selective targeting of microglial proliferation we have been able to uncover a pro-neurogenic role for microglia in chronic neurodegeneration, suggesting promising therapeutic targets to normalise the neurogenic niche during neurodegeneration.


Subject(s)
Hippocampus/physiology , Microglia/physiology , Neurogenesis/physiology , Prion Diseases/physiopathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Transforming Growth Factor beta/physiology , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL
6.
Am J Physiol Gastrointest Liver Physiol ; 306(6): G455-65, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24503766

ABSTRACT

Current literature consolidates the view of Crohn's disease (CD) as a form of immunodeficiency highlighting dysregulation of intestinal innate immunity in the pathogenesis of CD. Intestinal macrophages derived from blood monocytes play a key role in sustaining the innate immune homeostasis in the intestine, suggesting that the monocyte/macrophage compartment might be an attractive therapeutic target for the management of CD. Granulocyte macrophage colony-stimulating factor (GM-CSF) is a hematopoietic growth factor that also promotes myeloid cell activation, proliferation, and differentiation. GM-CSF has a protective effect in human CD and mouse models of colitis. However, the role of GM-CSF in immune and inflammatory reactions in the intestine is not well defined. Beneficial effects exerted by GM-CSF during intestinal inflammation could relate to modulation of the mucosal barrier function in the intestine, including epithelial cell proliferation, survival, restitution, and immunomodulatory actions. The aim of this review is to summarize potential mechanistic roles of GM-CSF in intestinal innate immune cell homeostasis and to highlight its central role in maintenance of the intestinal immune barrier in the context of immunodeficiency in CD.


Subject(s)
Crohn Disease/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Immunity, Innate/immunology , Inflammation/immunology , Intestines/immunology , Animals , Autoantibodies/immunology , Colitis/pathology , Colitis/physiopathology , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Homeostasis , Humans , Immunity, Innate/genetics , Inflammation/pathology , Intestinal Mucosa/pathology , Intestines/pathology , Macrophages/immunology , Mice , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology
7.
Blood ; 119(13): 3155-63, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-22223820

ABSTRACT

The t(8;21)(q22;q22) is common in adult acute myeloid leukemia (AML). The RUNX1-ETO fusion protein that is expressed by this translocation is poorly leukemogenic and requires additional mutations for transformation. Loss of sex chromosome (LOS) is frequently observed in t(8;21) AML. In the present study, to evaluate whether LOS cooperates with t(8;21) in leukemogenesis, we first used a retroviral transduction/transplantation model to express RUNX1-ETO in hematopoietic cells from XO mice. The low frequency of leukemia in these mice suggests that the potentially critical gene for suppression of t(8;21) leukemia in humans is not conserved on mouse sex chromosomes. The gene encoding the GM-CSF receptor α subunit (CSF2RA) is located on X and Y chromosomes in humans but on chromosome 19 in mice. GM-CSF promotes myeloid cell survival, proliferation, and differentiation. To determine whether GM-CSF signaling affects RUNX1-ETO leukemogenesis, hematopoietic stem/progenitor cells that lack GM-CSF signaling were used to express RUNX1-ETO and transplanted into lethally irradiated mice, and a high penetrance of AML was observed in recipients. Furthermore, GM-CSF reduced the replating ability of RUNX1-ETO-expressing cells. These results suggest a possible tumor-suppressor role of GM-CSF in RUNX1-ETO leukemia. Loss of the CSF2RA gene may be a critical mutation explaining the high incidence of LOS associated with the t(8;21)(q22;q22) translocation.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Leukemia, Myeloid, Acute/genetics , Signal Transduction/physiology , Translocation, Genetic , Adult , Animals , Cells, Cultured , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 8/genetics , Chromosomes, Mammalian/genetics , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Sex Chromosomes/genetics , Sex Chromosomes/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/genetics
8.
Acta Haematol ; 132(2): 220-5, 2014.
Article in English | MEDLINE | ID: mdl-24714374

ABSTRACT

BACKGROUND: Sweet's syndrome (SS) is an acute febrile neutrophilic dermatosis. It can occur as an idiopathic, drug-induced or malignancy-associated entity. SS is also seen in patients with myelodysplastic syndrome (MDS) where it may present atypically, both clinically and histologically. In a few rare cases of MDS, lymphocytic infiltrates are the presenting feature of SS. METHODS: MEDLINE and Scopus were the data sources for our review. RESULTS: A clinicopathological subsetemerged of 12 male SS patients with MDS and a mean age of 67.3 years in which the initial SS lesions were lymphocytic infiltrates. However, from 0.5 to 8 years later, sequential biopsies revealed neutrophilic dermal infiltration typical of SS. CONCLUSION: Initially lymphocytic infiltrates in this subset could be attributed either to an early timing of the biopsy concerning the age of the lesion or to the dysgranulopoiesis syndrome. A possible relationship between the dysfunction of the receptor of the granulocyte-macrophage colony stimulating factor, the gene of which is located on the pseudoautosomal X-Y region, may exist in MDS patients with initially lymphocytic SS. This could explain the male gender of this subset and might establish initially lymphocytic SS as a distinguished clinicopathological entity for predicting the occurrence and even the prognosis of MDS.


Subject(s)
Myelodysplastic Syndromes/complications , Sweet Syndrome/etiology , Aged , Anemia, Refractory, with Excess of Blasts/complications , Anemia, Refractory, with Excess of Blasts/pathology , Biopsy , Chromosomes, Human, X/genetics , Chromosomes, Human, Y/genetics , Disease Progression , Humans , Lymphocytes/pathology , Male , Middle Aged , Models, Immunological , Myelodysplastic Syndromes/pathology , Neutrophil Infiltration , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Sex Factors , Skin/pathology , Sweet Syndrome/genetics , Sweet Syndrome/immunology , Sweet Syndrome/pathology
9.
J Immunol ; 185(3): 1606-15, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20622119

ABSTRACT

GM-CSF, IL-3, and IL-5 are proinflammatory cytokines that control the production and function of myeloid and lymphoid cells. Their receptors are composed of a ligand-specific alpha subunit and a shared common signal-transducing beta subunit (beta common receptor or GM-CSFR beta [beta(c)]). The pleiotropic nature of biologic outcomes mediated by beta(c) and the presence of large, uncharacterized regions of its cytoplasmic domain suggest that much remains to be learned about its downstream signaling pathways. Although some previous work has attempted to link beta(c) with NF-kappaB activation, a definitive mechanism that mediates this pathway has not been described and, to date, it has not been clear whether the receptor can directly activate NF-kappaB. We demonstrate that NF-kappaB activation by beta(c) is dependent on TNFR-associated factor 6 (TRAF6) and that association of TRAF6 with beta(c) requires a consensus-binding motif found in other molecules known to interact with TRAF6. Furthermore, point mutation of this motif abrogated the ability of beta(c) to mediate NF-kappaB activation and reduced the viability of an IL-3-dependent hematopoietic cell line. Because this receptor plays a key role in hematopoiesis and the beta(c) cytoplasmic domain identified in this work mediates hematopoietic cell viability, this new pathway is likely to contribute to immune cell biology. This work is significant because it is the first description of a TRAF6-dependent signaling pathway associated with a type I cytokine receptor. It also suggests that TRAF6, a mediator of TNFR and TLR signaling, may be a common signaling intermediate in diverse cytokine receptor systems.


Subject(s)
Cytokine Receptor Common beta Subunit/physiology , NF-kappa B/physiology , TNF Receptor-Associated Factor 6/metabolism , Active Transport, Cell Nucleus/immunology , Animals , Cells, Cultured , Clone Cells , Consensus Sequence , Cytokine Receptor Common beta Subunit/antagonists & inhibitors , Cytokine Receptor Common beta Subunit/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/immunology , Fibroblasts/metabolism , Humans , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Protein Binding/genetics , Protein Binding/immunology , Protein Biosynthesis/immunology , Protein Structure, Tertiary/genetics , Protein Transport/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , TNF Receptor-Associated Factor 6/deficiency , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/physiology
10.
J Exp Med ; 201(6): 853-8, 2005 Mar 21.
Article in English | MEDLINE | ID: mdl-15781578

ABSTRACT

Granulocyte/macrophage colony-stimulating factor (GM-CSF) promotes the survival, proliferation, and differentiation of myeloid lineage cells and regulates chemotaxis and adhesion. However, mice in which the genes encoding GM-CSF (Gmcsf) or the beta common subunit of the GM-CSF receptor (betac) are inactivated display normal steady-state hematopoiesis. Here, we show that host GM-CSF signaling strongly modulates the ability of donor hematopoietic cells to radioprotect lethally irradiated mice. Although bone marrow mononuclear cells efficiently rescue Gmcsf mutant recipients, fetal liver cells and Sca1(+) lin(-/dim) marrow cells are markedly impaired. This defect is partially attributable to accessory cells that are more prevalent in bone marrow. In contrast, Gmcsf-deficient hematopoietic stem cells demonstrate normal proliferative potentials. Short-term survival is also impaired in irradiated betac mutant recipients transplanted with fetal liver or bone marrow. These data demonstrate a nonredundant function of GM-CSF in radioprotection by donor hematopoietic cells that may prove relevant in clinical transplantation.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Myeloid Progenitor Cells/physiology , Myelopoiesis/radiation effects , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Whole-Body Irradiation , Animals , Ataxin-1 , Ataxins , Bone Marrow Transplantation , Cell Proliferation/radiation effects , Cell Separation , Female , Fetus/cytology , Fetus/physiology , Gamma Rays , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Liver/cytology , Liver/physiology , Mice , Myelopoiesis/physiology , Nerve Tissue Proteins , Nuclear Proteins , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Signal Transduction/physiology , Signal Transduction/radiation effects
11.
Am J Respir Crit Care Med ; 182(10): 1292-304, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20622029

ABSTRACT

RATIONALE: We identified a 6-year-old girl with pulmonary alveolar proteinosis (PAP), impaired granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor function, and increased GM-CSF. OBJECTIVES: Increased serum GM-CSF may be useful to identify individuals with PAP caused by GM-CSF receptor dysfunction. METHODS: We screened 187 patients referred to us for measurement of GM-CSF autoantibodies to diagnose autoimmune PAP. Five were children with PAP and increased serum GM-CSF but without GM-CSF autoantibodies or any disease causing secondary PAP; all were studied with family members, subsequently identified patients, and controls. MEASUREMENT AND MAIN RESULTS: Eight children (seven female, one male) were identified with PAP caused by recessive CSF2RA mutations. Six presented with progressive dyspnea of insidious onset at 4.8 ± 1.6 years and two were asymptomatic at ages 5 and 8 years. Radiologic and histopathologic manifestations were similar to those of autoimmune PAP. Molecular analysis demonstrated that GM-CSF signaling was absent in six and severely reduced in two patients. The GM-CSF receptor ß chain was detected in all patients, whereas the α chain was absent in six and abnormal in two, paralleling the GM-CSF signaling defects. Genetic analysis revealed multiple distinct CSF2RA abnormalities, including missense, duplication, frameshift, and nonsense mutations; exon and gene deletion; and cryptic alternative splicing. All symptomatic patients responded well to whole-lung lavage therapy. CONCLUSIONS: CSF2RA mutations cause a genetic form of PAP presenting as insidious, progressive dyspnea in children that can be diagnosed by a combination of characteristic radiologic findings and blood tests and treated successfully by whole-lung lavage.


Subject(s)
Genetic Diseases, Inborn/etiology , Pulmonary Alveolar Proteinosis/genetics , Age of Onset , Autoantibodies/physiology , Child , Child, Preschool , Disease Progression , Dyspnea/etiology , Female , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Genetic Markers/genetics , Genotype , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Humans , Infant , Lung/pathology , Male , Mutation , Pedigree , Pulmonary Alveolar Proteinosis/diagnosis , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Alveolar Proteinosis/therapy , Receptors, Granulocyte Colony-Stimulating Factor/blood , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology
12.
Elife ; 92020 05 05.
Article in English | MEDLINE | ID: mdl-32367800

ABSTRACT

Macrophages derive from multiple sources of hematopoietic progenitors. Most macrophages require colony-stimulating factor 1 receptor (CSF1R), but some macrophages persist in the absence of CSF1R. Here, we analyzed mpeg1:GFP-expressing macrophages in csf1r-deficient zebrafish and report that embryonic macrophages emerge followed by their developmental arrest. In larvae, mpeg1+ cell numbers then increased showing two distinct types in the skin: branched, putative Langerhans cells, and amoeboid cells. In contrast, although numbers also increased in csf1r-mutants, exclusively amoeboid mpeg1+ cells were present, which we showed by genetic lineage tracing to have a non-hematopoietic origin. They expressed macrophage-associated genes, but also showed decreased phagocytic gene expression and increased epithelial-associated gene expression, characteristic of metaphocytes, recently discovered ectoderm-derived cells. We further demonstrated that juvenile csf1r-deficient zebrafish exhibit systemic macrophage depletion. Thus, csf1r deficiency disrupts embryonic to adult macrophage development. Zebrafish deficient for csf1r are viable and permit analyzing the consequences of macrophage loss throughout life.


Immune cells called macrophages are found in all organs in the body. These cells are highly effective at eating and digesting large particles including dead cells and debris, and microorganisms such as bacteria. Macrophages are also instrumental in shaping developing organs and repairing tissues during life. Macrophages were, until recently, thought to be constantly replenished from cells circulating in the bloodstream. However, it turns out that separate populations of macrophages become established in most tissues during embryonic development and are maintained throughout life without further input. Previous studies of zebrafish, rodents and humans have shown that, when a gene called CSF1R is non-functional, macrophages are absent from many organs including the brain. However, some tissue-specific macrophages still persist, and it was not clear why these cells do not rely on the CSF1R gene while others do. Kuil et al. set out to decipher the precise requirement for the CSF1R gene in macrophage development in living zebrafish. The experiments used zebrafish that make a green fluorescent protein in their macrophages. As these fish are transparent, this meant that Kuil et al. could observe the cells within the living fish and isolate them to determine which genes are switched on and off. This approach revealed that zebrafish with a mutated version of the CSF1R gene make macrophages as embryos but that these cells then fail to multiply and migrate into the developing organs. This results in fewer macrophages in the zebrafish's tissues, and an absence of these cells in the brain. Kuil et al. went on to show that new macrophages did emerge in zebrafish that were about two to three weeks old. However, unexpectedly, these new cells were not regular macrophages. Instead, they were a new recently identified cell-type called metaphocytes, which share similarities with macrophages but have a completely different origin, move faster and do not eat particles. Zebrafish lacking the CSF1R gene thus lose nearly all their macrophages but retain metaphocytes. These macrophage-free mutant zebrafish constitute an unprecedented tool for further studies looking to discriminate the different roles of macrophages and metaphocytes.


Subject(s)
Macrophages/physiology , Microglia/physiology , Protein-Tyrosine Kinases/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Zebrafish Proteins/physiology , Animals , Cell Proliferation , Gene Expression Profiling , Macrophages/metabolism , Microglia/metabolism , Receptor Protein-Tyrosine Kinases , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Zebrafish/embryology , Zebrafish Proteins/metabolism
13.
J Cell Biol ; 141(4): 1041-51, 1998 May 18.
Article in English | MEDLINE | ID: mdl-9585421

ABSTRACT

The cytokine Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) regulates proliferation, differentiation, and apoptosis during myelopoiesis and erythropoiesis. Structure-function relationships of GM-CSF interactions with its receptor (GM-R), the biochemistry of GM-R signal transduction, and GM-CSF action in vivo are relatively well understood. Much less is known, however, about GM-R function in primary hematopoietic cells. In this paper we show that expression of the human GM-R in a heterologous cell system (primary avian erythroid and myeloid cells) confirms respective results in murine or human cell lines, but also provides new insights how the GM-R regulates progenitor proliferation and differentiation. As expected, the hGM-CSF stimulated myeloid progenitor proliferation and differentiation and enhanced erythroid progenitor proliferation during terminal differentiation. In the latter cells, however, the hGM-R only partially substituted for the activities of the erythropoietin receptor (EpoR). It failed to replace the EpoR in its cooperation with c-Kit to induce long-term proliferation of erythroid progenitors. Furthermore, the hGM-R alpha chain specifically interfered with EpoR signaling, an activity neither seen for the betac subunit of the receptor complex alone, nor for the alpha chain of the closely related Interleukin-3 receptor. These results point to a novel role of the GM-R alpha chain in defining cell type-specific functions of the GM-R.


Subject(s)
Erythroblasts/cytology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/cytology , Macrophages/cytology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line, Transformed , Cells, Cultured , Chick Embryo , Erythroblasts/drug effects , Erythroblasts/physiology , Erythropoietin/pharmacology , Fibroblasts , Genetic Vectors , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/physiology , Hemoglobins/biosynthesis , Humans , Kinetics , Macrophages/drug effects , Mammals , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Retroviridae , Signal Transduction , Transfection
14.
Front Immunol ; 10: 2656, 2019.
Article in English | MEDLINE | ID: mdl-31798586

ABSTRACT

Viral encephalitis is a rare but clinically serious consequence of viral invasion of the brain and insight into its pathogenesis is urgently needed. Important research questions concern the involvement of the host innate immune response in pathogenesis, key to which is the role played by microglia, resident macrophages of the brain parenchyma. Do microglia have a protective function, by coordinating the innate immune response to viral infection, or do they drive pathogenic neuroinflammation? Here we synthesize recent data from mouse models of acute viral encephalitis, which reveal an unambiguously protective role for microglia. Depletion of microglia, via blockade of colony-stimulating factor 1 receptor (CSF1R) signaling, led to increased viral replication accompanied by more severe neurological disease and heightened mortality. Whilst the underlying mechanism(s) remain to be defined, microglial interactions with T cells and phagocytosis of infected neurones appear to play a role. Paradoxically, the production of inflammatory cytokines was increased in several instances following viral infection in microglia-depleted brains, suggesting that: (i) cells other than microglia mediate inflammatory responses and/or (ii) microglia may exert a regulatory function. Under certain circumstances the microglial antiviral response might contribute negatively to longer-term neurological sequelae, although fewer studies have focused on this aspect in encephalitis models. Understanding regulation of the microglial response, and how it contributes to disease is therefore a priority for future studies. Collectively, these findings demonstrate the central role of microglia in pathogenesis, suggesting the exciting possibility that defects of microglial function might contribute to encephalitis susceptibility and/or outcome in humans.


Subject(s)
Encephalitis, Viral/immunology , Microglia/physiology , Animals , Brain/immunology , Brain/virology , Disease Models, Animal , Humans , Immunity, Innate , Mice , Phagocytosis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , T-Lymphocytes/immunology
15.
Eur J Med Chem ; 162: 161-175, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30445265

ABSTRACT

Kinase irregularity has been correlated with several complex neurodegenerative tauopathies. Development of selective inhibitors of these kinases might afford promising anti-tauopathy therapies. While DAPK1 inhibitors halt the formation of tau aggregates and counteract neuronal death, CSF1R inhibitors could alleviate the tauopathies-associated neuroinflammation. Herein, we report the design, synthesis, biological evaluation, mechanistic study, and molecular docking study of novel CSF1R/DAPK1 dual inhibitors as multifunctional molecules inhibiting the formation of tau aggregates and neuroinflammation. Compound 3l, the most potent DAPK1 inhibitor in the in vitro kinase assay (IC50 = 1.25 µM) was the most effective tau aggregates formation inhibitor in the cellular assay (IC50 = 5.0 µM). Also, compound 3l elicited potent inhibition of CSF1R in the in vitro kinase assay (IC50 = 0.15 µM) and promising inhibition of nitric oxide production in LPS-induced BV-2 cells (55% inhibition at 10 µM concentration). Kinase profiling and hERG binding assay anticipated the absence of off-target toxicities while the PAMPA-BBB assay predicted potentially high BBB permeability. The mechanistic study and selectivity profile suggest compound 3l as a non-ATP-competitive DAPK1 inhibitor and an ATP-competitive CSF1R inhibitor while the in silico calculations illustrated binding of compound 3l to the substrate-binding site of DAPK1. Hence, compound 3l might act as a protein-protein interaction inhibitor by hindering DAPK1 kinase reaction through preventing the binding of DAPK1 substrates.


Subject(s)
Benzamides/pharmacology , Death-Associated Protein Kinases/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Tauopathies/drug therapy , Animals , Benzamides/chemistry , Binding Sites , Cell Line , Drug Design , Humans , Mice , Molecular Docking Simulation , Protein Binding/drug effects
16.
Cytokine ; 42(2): 234-242, 2008 May.
Article in English | MEDLINE | ID: mdl-18374598

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin (IL)-3 and IL-5 are related cytokines that play key roles in regulating the differentiation, proliferation, survival and activation of myeloid blood cells. The cell surface receptors for these cytokines are composed of cytokine-specific alpha-subunits and a common beta-receptor (betac), a shared subunit that is essential for receptor signaling in response to GM-CSF, IL-3 and IL-5. Previous studies have reached conflicting conclusions as to whether N-glycosylation of the betac-subunit is necessary for functional GM-CSF, IL-3 and IL-5 receptors. We sought to clarify whether betac N-glycosylation plays a role in receptor function, since all structural studies of human betac to date have utilized recombinant protein lacking N-glycosylation at Asn(328). Here, by eliminating individual N-glycans in human betac and the related murine homolog, beta(IL-3), we demonstrate unequivocally that ligand-binding and receptor activation are not critically dependent on individual N-glycosylation sites within the beta-subunit although the data do not preclude the possibility that N-glycans may exert some sort of fine control. These studies support the biological relevance of the X-ray crystal structures of the human betac domain 4 and the complete ectodomain, both of which lack N-glycosylation at Asn(328).


Subject(s)
Cytokine Receptor Common beta Subunit/physiology , Polysaccharides/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Receptors, Interleukin-3/physiology , Receptors, Interleukin-5/physiology , Animals , COS Cells , Chlorocebus aethiops , Cytokine Receptor Common beta Subunit/chemistry , Cytokine Receptor Common beta Subunit/genetics , Humans , Interleukin-3/metabolism , Interleukin-5/metabolism , Mice , Mutagenesis, Site-Directed , Polysaccharides/chemistry , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Receptors, Interleukin-3/chemistry , Receptors, Interleukin-5/chemistry
17.
Alcohol ; 42(2): 137-42, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18358993

ABSTRACT

The 11th annual meeting of the Alcohol and Immunology Research Interest Group was held at Loyola University Medical Center, Maywood, Illinois on November 17, 2006. The Alcohol and Immunology Research Interest Group meeting is held annually to exchange new findings and ideas that arise from ongoing research examining the effects of alcohol intake on the immune system. The event consisted of five sessions, two of which featured plenary talks from invited speakers, two with oral presentations from selected abstracts, and a final poster session. Participants presented new data on a variety of topics including the effects of ethanol on key cells of the immune system (neutrophils, dendritic cells, NK cells), B cell responses, the capacity to clear infectious agents, and the barrier functions of skin, lung, and intestine.


Subject(s)
Ethanol/toxicity , Immunity/drug effects , Infections/immunology , Inflammation/immunology , Animals , Cytokines/biosynthesis , Dendritic Cells/drug effects , Dendritic Cells/immunology , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lipopolysaccharides/toxicity , Peroxidase/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology
18.
Cell Signal ; 51: 191-198, 2018 11.
Article in English | MEDLINE | ID: mdl-30075184

ABSTRACT

Although epidermal growth factor receptor (EGFR) has been identified as a potent "oncogenic driver" in various tumors of epithelial origin, EGFR-targeted therapies are often of limited success. One of the challenges of improving targeted therapies is to overcome bypassing signaling pathways. Analysis of RNA-seq data of 1006 cell lines from the Cancer Cell Line Encyclopedia (CCLE) revealed that more than 12% of carcinoma cell lines expressed markedly elevated mRNA levels of colony-stimulating factor (CSF)-1 receptor (CSF-1R). Since epithelial cells also express CSF-1, elevated levels of CSF-1R may participate in providing alternative growth and survival signals under targeted therapies. To address this question, we ectopically expressed CSF-1R in A431 cells that express EGFR at high levels, but no biologically relevant level of CSF-1R. In the presence of EGFR inhibitor gefitinib, CSF-1R provided a significant growth advantage in A431 cells. As expected, activation of both receptors, EGFR or CSF-1R, induced phosphorylation of extracellular signal-regulated kinase (Erk)1/2, Akt, protein kinase C (PKC) and signal transducer and activator of transcription (STAT)3. However, EGFR, but not CSF-1R, also induced STAT5 phosphorylation. Inhibitor of phosphatidylinositol 3-kinase (PI3K) (AZD8186), MAPK/ERK kinase (MEK)1/2 (U0126), PKCs (Bisindolylmaleimide I or Gö6976) or STAT3 (Stattic) partially reduced proliferation of CSF-1R expressing A431 cells in the presence of gefitinib. Moreover, multi-kinase inhibitor, cabozantinib, suppressed CSF-1R activation and drastically reduced cell growth when combined with gefitinib. These data suggest that CSF-1R has the potential to reduce sensitivity to gefitinib and may be involved in resistance development.


Subject(s)
Carcinoma , Drug Resistance, Neoplasm/physiology , Gefitinib/therapeutic use , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Signal Transduction/drug effects , Skin Neoplasms , Anilides/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Carcinoma/metabolism , Cell Proliferation/drug effects , Databases, Genetic , ErbB Receptors/antagonists & inhibitors , HeLa Cells , Hep G2 Cells , Humans , Macrophage Colony-Stimulating Factor/metabolism , Macrophages , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Pyridines/therapeutic use , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism
19.
Mol Cell Biol ; 18(2): 742-52, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9447970

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) induces various functions, including the proliferation and differentiation of a broad range of hematopoietic cells. We previously reported that at least two distinct pathways are involved in human GM-CSF receptor signaling; both require the box 1 region of the common beta subunit (beta c). This region is essential for the activation of JAK2, which is necessary for all the biological functions of GM-CSF. The activation of JAK2 by GM-CSF leads to rapid tyrosine phosphorylation of cellular proteins, including the beta c. However, the significance of beta c phosphorylation with regard to the regulation of signaling molecules and the expression of GM-CSF functions is less well understood. Here we investigated the role of the cytoplasmic tyrosine residues of the beta c by using a series of beta c mutants expressed in murine BA/F3 cells. A mutant beta c with all eight cytoplasmic tyrosines converted to phenylalanine (Fall) activated JAK2 but not SHP-2, MAPK cascades, STAT5, or the c-fos promoter in BA/F3 cells, and it did not effectively induce proliferation. Adding back each tyrosine to Fall revealed that Tyr577, Tyr612, and Tyr695 are involved in the activation of SHP-2, MAPK cascades, and c-fos transcription, while every tyrosine, particularly Tyr612, Tyr695, Tyr750, and Tyr806, facilitated STAT5 activation. Impaired growth was also restored, at least partly, by any of the tyrosines. These results provide evidence that beta c tyrosines possess distinct yet overlapping functions in activating multiple signaling pathways induced by GM-CSF.


Subject(s)
Milk Proteins , Mitogen-Activated Protein Kinases , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Signal Transduction , Tyrosine/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division , DNA-Binding Proteins/metabolism , Enzyme Activation , Genes, fos , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Humans , JNK Mitogen-Activated Protein Kinases , Mice , Mitogen-Activated Protein Kinase 1 , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Proto-Oncogene Proteins c-raf/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , STAT5 Transcription Factor , Trans-Activators/metabolism , Tyrosine/chemistry
20.
Cancer Res ; 52(19): 5208-12, 1992 Oct 01.
Article in English | MEDLINE | ID: mdl-1382839

ABSTRACT

Mutations in the Steel locus, encoding a growth factor (Steel factor or SF) or c-kit, the gene encoding its receptor, result in severe anemia in the mouse. In the present study, we have addressed the mechanism of synergistic growth activation, at the cellular level, by SF and GM-CSF using the blast cells of acute myeloblastic leukemia (AML blasts). Our data indicate that SF drastically alleviates the requirement in cell interaction for blast colony formation in most of the samples tested. Analysis of cultures performed in the presence of SF and GM-CSF at different cell concentrations, ranging from 1,000 to 20,000 cells, suggested a single limiting element, i.e., the blast clonogenic cell, while 2 or more limiting elements were found in cultures stimulated with GM-CSF alone, suggesting interacting cell populations. The presence of membrane-bound SF was detected by immunofluorescence, suggesting the possibility that secreted or membrane-bound SF may, at least in part, contribute to the density-dependent growth of AML blasts. In all samples tested, SF appears to increase the responsiveness of AML blasts to GM-CSF, as demonstrated by a 3-fold decrease of GM-CSF half efficient concentration on addition of SF to the cultures. Exposure of AML blasts to SF did not affect GM-CSF receptor expression, suggesting that this increase in GM-CSF responsiveness is likely to occur at the postreceptor level. Interestingly, 2 of 15 AML samples surveyed did not respond to SF, and were both of the myelomonocytic or monocytic subtype, classified as M4 and M5, respectively.


Subject(s)
Cell Communication/drug effects , Hematopoietic Cell Growth Factors/genetics , Leukemia, Myeloid, Acute/pathology , Cell Communication/physiology , Cell Membrane/metabolism , Drug Synergism , Fluorescent Antibody Technique , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Cell Growth Factors/pharmacology , Humans , Intercellular Junctions/drug effects , Intercellular Junctions/physiology , Kinetics , Leukemia, Myeloid, Acute/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/drug effects , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Stem Cell Factor , Tumor Cells, Cultured , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL