Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Elife ; 102021 04 08.
Article in English | MEDLINE | ID: mdl-33830019

ABSTRACT

Innate immune cellular effectors are actively consumed during systemic inflammation, but the systemic traffic and the mechanisms that support their replenishment remain unknown. Here, we demonstrate that acute systemic inflammation induces the emergent activation of a previously unrecognized system of rapid migration of granulocyte-macrophage progenitors and committed macrophage-dendritic progenitors, but not other progenitors or stem cells, from bone marrow (BM) to regional lymphatic capillaries. The progenitor traffic to the systemic lymphatic circulation is mediated by Ccl19/Ccr7 and is NF-κB independent, Traf6/IκB-kinase/SNAP23 activation dependent, and is responsible for the secretion of pre-stored Ccl19 by a subpopulation of CD205+/CD172a+ conventional dendritic cells type 2 and upregulation of BM myeloid progenitor Ccr7 signaling. Mature myeloid Traf6 signaling is anti-inflammatory and necessary for lymph node myeloid cell development. This report unveils the existence and the mechanistic basis of a very early direct traffic of myeloid progenitors from BM to lymphatics during inflammation.


When the body becomes infected with disease-causing pathogens, such as bacteria, the immune system activates various mechanisms which help to fight off the infection. One of the immune system's first lines of defense is to launch an inflammatory response that helps remove the pathogen and recruit other immune cells. However, this response can become overactivated, leading to severe inflammatory conditions that damage healthy cells and tissues. A second group of cells counteract this over inflammation and are different to the ones involved in the early inflammatory response. Both types of cells ­ inflammatory and anti-inflammatory ­ develop from committed progenitors, which, unlike stem cells, are already destined to become a certain type of cell. These committed progenitors reside in the bone marrow and then rapidly travel to secondary lymphoid organs, such as the lymph nodes, where they mature into functioning immune cells. During this journey, committed progenitors pass from the bone marrow to the lymphatic vessels that connect up the different secondary lymphoid organs, and then spread to all tissues in the body. Yet, it is not fully understood what exact route these cells take and what guides them towards these lymphatic tissues during inflammation. To investigate this, Serrano-Lopez, Hegde et al. used a combination of techniques to examine the migration of progenitor cells in mice that had been treated with lethal doses of a bacterial product that triggers inflammation. This revealed that as early as one to three hours after the onset of infection, progenitor cells were already starting to travel from the bone marrow towards lymphatic vessels. Serrano-Lopez, Hegde et al. found that a chemical released by an "alarm" immune cell already residing in secondary lymphoid organs attracted these progenitor cells towards the lymphatic tissue. Further experiments showed that the progenitor cells travelling to secondary lymphoid organs were already activated by bacterial products. They then follow the chemical released by alarm immune cells ready to respond to the immune challenge and suppress inflammation. These committed progenitors were also found in the inflamed lymph nodes of patients. These findings suggest this rapid circulation of progenitors is a mechanism of defense that contributes to the fight against severe inflammation. Altering how these cells migrate from the bone marrow to secondary lymphoid organs could provide a more effective treatment for inflammatory conditions and severe infections. However, these approaches would need to be tested further in the laboratory and in clinical trials.


Subject(s)
Bone Marrow/metabolism , Cell Movement , Granulocyte-Macrophage Progenitor Cells/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Lymphadenopathy/metabolism , Lymphatic System/metabolism , Myeloid Progenitor Cells/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Bone Marrow/immunology , Bone Marrow/pathology , Cell Lineage , Cells, Cultured , Child , Child, Preschool , Disease Models, Animal , Female , Granulocyte-Macrophage Progenitor Cells/immunology , Granulocyte-Macrophage Progenitor Cells/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Lymphadenopathy/immunology , Lymphadenopathy/pathology , Lymphatic System/immunology , Lymphatic System/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myeloid Progenitor Cells/immunology , Myeloid Progenitor Cells/pathology , Phenotype , Signal Transduction , Time Factors , Young Adult
2.
Int J Radiat Biol ; 96(5): 584-595, 2020 05.
Article in English | MEDLINE | ID: mdl-31906761

ABSTRACT

Purpose: To study whether radiation-induced bleeding in the bone marrow induced iron accumulation, and subsequently caused ferroptosis in granulocyte-macrophage hematopoietic progenitor cells.Materials and methods: Male mice were subjected to different doses (0, 4, 8, or 10 Gy) of gamma radiation from a 137Cs source. The changes in iron metabolism or ferroptosis-related parameters of irradiated bone marrow were accessed with biochemical, histopathological, and antibody methods. Hematocytes were detected with a hematology analyzer. The counts of granulocyte-macrophage hematopoietic progenitor cells were measured with the granulocyte-macrophage colony-forming unit.Results: Iron accumulation occurred in the bone marrow, which caused by radiation-induced hemorrhage. The iron accumulation triggered an iron regulatory protein-ferroportin 1 axis to increase serum iron levels. Using LDN193189, radiation-induced iron accumulation was demonstrated to decrease white blood cell counts at least partly through a decrease in the counts of granulocyte-macrophage hematopoietic progenitor cells. The reduction in the counts of granulocyte-macrophage hematopoietic progenitor cells was subsequently demonstrated to attribute to ferroptosis with the use of ferroptosis inhibitors and through the detection of ferroptosis related-parameters. The survival rate of irradiated mice was improved using Ferrostatin-1 or LDN193189.Conclusions: These findings suggest that radiation-induced hemorrhage in the bone marrow causes ferroptosis in granulocyte-macrophage hematopoietic progenitor cells, and anti-ferroptosis has the potential to be a radioprotective strategy to ameliorate radiation-induced hematopoietic injury.


Subject(s)
Ferroptosis/radiation effects , Granulocyte-Macrophage Progenitor Cells/radiation effects , Animals , Cyclohexylamines/pharmacology , Gamma Rays , Granulocyte-Macrophage Progenitor Cells/metabolism , Granulocyte-Macrophage Progenitor Cells/pathology , Iron/metabolism , Leukocyte Count , Male , Mice , Mice, Inbred ICR , Phenylenediamines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology
3.
Inflamm Bowel Dis ; 26(5): 709-719, 2020 04 11.
Article in English | MEDLINE | ID: mdl-31821463

ABSTRACT

BACKGROUND: The aim of this study was to address whether the therapeutic effect of leukocytapheresis (LCAP) depends on calcitonin gene- related peptide (CGRP) induction. METHODS: An HLA-B27 transgenic rat model was treated with an LCAP column. The effects of LCAP on clinical, endoscopic, and histologic disease activity, the colony-forming ability of colony-forming unit (CFU)-granulocyte macrophages (GMs), colonic blood flow, and tissue expression of tumor necrosis factor (TNF)-α and CGRP were examined. Changes in the effects of LCAP after pretreatment with the CGRP antagonist CGRP8-37 were also observed. A dextran sulfate sodium-induced colitis rat model included treatment with CGRP, and the effect was assessed based on clinical, endoscopic, and histologic disease activity, colonic blood flow, the colony-forming ability of CFU-GMs, and tissue expression of inflammatory cytokines and CGRP receptor families. RESULTS: LCAP improved disease activity, enhanced colonic blood flow, and induced the bone marrow colony-forming ability of CFU-GMs with an increase in CGRP mRNA levels. These effects were abolished by pretreatment with CGRP8-37. The administration of CGRP suppressed colitis, promoting colonic blood flow, inducing bone marrow-derived cells, downregulating inflammatory cytokines, and upregulating receptor activity-modifying protein-1. The mRNA and protein levels of inflammatory cytokines in lipopolysaccharide-stimulated mononuclear cells were also decreased after CGRP treatment. CONCLUSIONS: The therapeutic effects of LCAP depend on CGRP induction. CGRP can effectively suppress colitis through the downregulation of inflammatory events and upregulation of protective events.


Subject(s)
Calcitonin Gene-Related Peptide/pharmacology , Colitis/therapy , Leukapheresis/methods , Animals , Colitis/chemically induced , Colon/blood supply , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Down-Regulation/drug effects , Granulocyte-Macrophage Progenitor Cells/metabolism , HLA-B27 Antigen , Male , Rats , Rats, Inbred Lew , Rats, Transgenic , Receptor Activity-Modifying Protein 1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
4.
Transfusion ; 60(1): 144-154, 2020 01.
Article in English | MEDLINE | ID: mdl-31756003

ABSTRACT

BACKGROUND: The CFU assay is considered the only in vitro assay that assesses the biologic function of hematopoietic stem and progenitor cells (HSPC). STUDY DESIGN AND METHODS: To investigate the impact of post-thaw CFU-GM counts on the quality of umbilical cord blood (UCB), we studied transplant outcomes in 269 patients receiving single UCB transplant. We also correlated the post-thaw CFU-GM counts of 1912 units with the pre-freeze and post-thaw graft characteristics, hoping to optimize selection criteria of UCB. Data analysis included: total nucleated cells, viability, CD34+, nucleated red blood cells (NRBC), hematocrit, frozen storage time, and cord blood bank (CBB). RESULTS: We demonstrated an association between post-thaw CFU-GM dose and the speed of neutrophil and platelet engraftment (p < 0.01). Higher post-thaw CFU-GM dose showed an increased benefit for neutrophil and platelet engraftment (p < 0.01). Post-thaw CD34+ cell dose and CFU-GM dose were strongly correlated (r = 0.78). However, CFU-GM dose showed additional benefit for patients receiving the lowest quartile of CD34+ dose. HLA disparity did not adversely impact either neutrophil or platelet engraftment. Post-thaw CFU-GM/million nucleated cells plated showed moderate correlation with pre-freeze and post-thaw CD34+ and weak correlation with other parameters. Post-thaw CFU-GM was not influenced by storage time, but was impacted by the CBB from which the unit is obtained (p < 0.01). CONCLUSION: Post-thaw CFU-GM is an effective measure of the quality and efficacy of the UCB graft, particularly adding valuable clinical information when the CD34+ cell dose is low. Consideration of pre-freeze CD34+ cell content and CBB as additional selection criteria is warranted.


Subject(s)
Blood Platelets/metabolism , Cord Blood Stem Cell Transplantation , Cryopreservation , Granulocyte-Macrophage Progenitor Cells/metabolism , Hematologic Neoplasms , Neutrophils/metabolism , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Graft Survival , Hematologic Neoplasms/blood , Hematologic Neoplasms/therapy , Humans , Infant , Male , Middle Aged
5.
Int J Mol Sci ; 22(1)2020 Dec 29.
Article in English | MEDLINE | ID: mdl-33383835

ABSTRACT

Calcium pyrophosphate dihydrate (CPPD) crystals are formed locally within the joints, leading to pseudogout. Although the mobilization of local granulocytes can be observed in joints where pseudogout has manifested, the mechanism of this activity remains poorly understood. In this study, CPPD crystals were administered to mice, and the dynamics of splenic and peripheral blood myeloid cells were analyzed. As a result, levels of both granulocytes and monocytes were found to increase following CPPD crystal administration in a concentration-dependent manner, with a concomitant decrease in lymphocytes in the peripheral blood. In contrast, the levels of other cells, such as dendritic cell subsets, T-cells, and B-cells, remained unchanged in the spleen, following CPPD crystal administration. Furthermore, an increase in granulocytes/monocyte progenitors (GMPs) and a decrease in megakaryocyte/erythrocyte progenitors (MEPs) were also observed in the bone marrow. In addition, CPPD administration induced production of IL-1ß, which acts on hematopoietic stem cells and hematopoietic progenitors and promotes myeloid cell differentiation and expansion. These results suggest that CPPD crystals act as a "danger signal" to induce IL-1ß production, resulting in changes in course of hematopoietic progenitor cell differentiation and in increased granulocyte/monocyte levels, and contributing to the development of gout.


Subject(s)
Calcium Pyrophosphate/chemistry , Calcium Pyrophosphate/metabolism , Cell Differentiation , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/metabolism , Granulocytes/metabolism , Monocytes/metabolism , Animals , Biomarkers , Bone Marrow , Cytokines/metabolism , Granulocytes/cytology , Immunophenotyping , Inflammation Mediators/metabolism , Leukocyte Count , Liquid Crystals , Mice , Monocytes/cytology
6.
Int J Biol Sci ; 15(13): 2763-2773, 2019.
Article in English | MEDLINE | ID: mdl-31853216

ABSTRACT

Protein arginine methyltransferase 1 (PRMT1) is the predominant asymmetric (type I) methyltransferase in mammalian cells. Mounting evidence suggested that PRMT1 is essential to embryonic development and tumor pathogenesis, but its role in normal adult hematopoiesis is less studied. We used a Prmt1 conditional knockout (KO) mouse model to identify the role of PRMT1 in normal adult hematopoiesis. The results indicated that deletion of PRMT1 results in anemia and leukopenia, reducing terminal erythroid and lymphocyte differentiation. Additionally, we found a significant decrease of megakaryocyte progenitors (MkPs) compared with similarly treated littermate control mice. The frequency of short-term hematopoietic stem cells (ST-HSCs) and granulocyte-macrophage progenitors (GMPs) populations were significantly lower in PRMT1f/f/Mx1-CRE bone marrow (BM) compared with littermate control mice. Importantly, in-vitro replating assays and BM transplantation results revealed that PRMT1 KO results in reduced hematopoietic stem and progenitor cells (HSPCs) self-renewal capacity. Thus, we conclude that PRMT1 is required for hematopoietic differentiation and the competitive fitness of HSPCs, and we believed that PRMT1 serves as a key epigenetic regulator of normal hematopoiesis that occurs throughout life.


Subject(s)
Granulocyte-Macrophage Progenitor Cells/metabolism , Hematopoiesis/physiology , Protein-Arginine N-Methyltransferases/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Flow Cytometry , Granulocyte-Macrophage Progenitor Cells/cytology , Hematopoiesis/genetics , Mice , Mice, Knockout , Protein-Arginine N-Methyltransferases/genetics
7.
Cell ; 178(6): 1509-1525.e19, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31491389

ABSTRACT

Most tissue-resident macrophage (RTM) populations are seeded by waves of embryonic hematopoiesis and are self-maintained independently of a bone marrow contribution during adulthood. A proportion of RTMs, however, is constantly replaced by blood monocytes, and their functions compared to embryonic RTMs remain unclear. The kinetics and extent of the contribution of circulating monocytes to RTM replacement during homeostasis, inflammation, and disease are highly debated. Here, we identified Ms4a3 as a specific gene expressed by granulocyte-monocyte progenitors (GMPs) and subsequently generated Ms4a3TdT reporter, Ms4a3Cre, and Ms4a3CreERT2 fate-mapping models. These models traced efficiently monocytes and granulocytes, but no lymphocytes or tissue dendritic cells. Using these models, we precisely quantified the contribution of monocytes to the RTM pool during homeostasis and inflammation. The unambiguous identification of monocyte-derived cells will permit future studies of their function under any condition.


Subject(s)
Cell Cycle Proteins/genetics , Gene Expression , Granulocyte-Macrophage Progenitor Cells/metabolism , Granulocytes/metabolism , Macrophages/metabolism , Membrane Proteins/genetics , Monocytes/metabolism , Animals , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocytes/cytology , Hematopoiesis/physiology , Homeostasis/physiology , Inflammation/metabolism , Macrophages/cytology , Mice , Monocytes/cytology
8.
Cell Death Dis ; 10(8): 598, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31395859

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) comprise a critical component of the tumor environment and CXCR2 reportedly plays a key role in the pathophysiology of various inflammatory diseases. Here, CXCR2 expression on granulocyte and macrophage progenitor cells (GMPs) was found to participate in myeloid cell differentiation within the tumor environment. In CXCR2-deficient tumor-bearing mice, GMPs exhibited fewer macrophage and dendritic cell progenitor cells than wild-type tumor-bearing mice, thereby decreasing monocytic MDSCs (mo-MDSCs) expansion. CXCR2 deficiency increased SAP18 expression in tumor-bearing mice, which reduced STAT3 phosphorylation through restraining ERK1/2 activation. Our findings reveal a critical role for CXCR2 in regulating hematopoietic progenitor cell differentiation under tumor conditions, and SAP18 is a key negative regulator in this process. Thus, inhibiting CXCR2 expression may alter the tumor microenvironment and attenuate tumor progression.


Subject(s)
Co-Repressor Proteins/genetics , Granulocyte-Macrophage Progenitor Cells/metabolism , Melanoma, Experimental/genetics , RNA-Binding Proteins/genetics , Receptors, Interleukin-8B/genetics , STAT3 Transcription Factor/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Granulocyte-Macrophage Progenitor Cells/pathology , Humans , MAP Kinase Signaling System/genetics , Melanoma, Experimental/pathology , Mice , Monocytes/metabolism , Monocytes/pathology , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , RNA, Small Interfering/pharmacology , Receptors, Interleukin-8B/antagonists & inhibitors , Tumor Microenvironment/genetics
9.
Blood ; 133(22): 2413-2426, 2019 05 30.
Article in English | MEDLINE | ID: mdl-30917956

ABSTRACT

Eosinophils and neutrophils are critical for host defense, yet gaps in understanding how granulocytes differentiate from hematopoietic stem cells (HSCs) into mature effectors remain. The pseudokinase tribbles homolog 1 (Trib1) is an important regulator of granulocytes; knockout mice lack eosinophils and have increased neutrophils. However, how Trib1 regulates cellular identity and function during eosinophilopoiesis is not understood. Trib1 expression markedly increases with eosinophil-lineage commitment in eosinophil progenitors (EoPs), downstream of the granulocyte/macrophage progenitor (GMP). Using hematopoietic- and eosinophil-lineage-specific Trib1 deletion, we found that Trib1 regulates both granulocyte precursor lineage commitment and mature eosinophil identity. Conditional Trib1 deletion in HSCs reduced the size of the EoP pool and increased neutrophils, whereas deletion following eosinophil lineage commitment blunted the decrease in EoPs without increasing neutrophils. In both modes of deletion, Trib1-deficient mice expanded a stable population of Ly6G+ eosinophils with neutrophilic characteristics and functions, and had increased CCAAT/enhancer binding protein α (C/EBPα) p42. Using an ex vivo differentiation assay, we found that interleukin 5 (IL-5) supports the generation of Ly6G+ eosinophils from Trib1-deficient cells, but is not sufficient to restore normal eosinophil differentiation and development. Furthermore, we demonstrated that Trib1 loss blunted eosinophil migration and altered chemokine receptor expression, both in vivo and ex vivo. Finally, we showed that Trib1 controls eosinophil identity by modulating C/EBPα. Together, our findings provide new insights into early events in myelopoiesis, whereby Trib1 functions at 2 distinct stages to guide eosinophil lineage commitment from the GMP and suppress the neutrophil program, promoting eosinophil terminal identity and maintaining lineage fidelity.


Subject(s)
Eosinophils/metabolism , Gene Expression Regulation , Granulocyte-Macrophage Progenitor Cells/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Myelopoiesis , Neutrophils/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Eosinophils/cytology , Granulocyte-Macrophage Progenitor Cells/cytology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Neutrophils/cytology , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics
10.
Cells ; 9(1)2019 12 28.
Article in English | MEDLINE | ID: mdl-31905606

ABSTRACT

BACKGROUND: Trypanosoma cruzi (Tc) causes Chagas disease (CD) that is the most frequent cause of heart failure in Latin America. TNF-α+ monocytes/macrophages (Mo/Mφ) are associated with inflammatory pathology in chronic CD. In this study, we determined the progenitor lineage of Mo/Mφ contributing to inflammation and examined the regulatory role of SIRT1 in modulating the Mo/Mφ response in Chagas disease. METHODS AND RESULTS: C57BL/6 mice were infected with Tc, treated with SIRT1 agonist (SRT1720) after control of acute parasitemia, and monitored during chronic phase (150 days post-infection). Flow cytometry studies showed an increase in maturation of bone marrow hematopoietic stem cell (HSC)-derived Mo of proinflammatory and anti-inflammatory phenotype in acutely- and chronically-infected mice; however, these cells were not increased in splenic compartment of infected mice. Instead, yolk-sac-derived CD11b+ F4/80+ Mo/Mφ were increased in sinusoidal compartment of Chagas mice. The splenic CD11b+ F4/80+ Mo/Mφ of Chagas (vs. control) mice exhibited increased mRNA, protein, and surface expression of markers of proinflammatory phenotype (CD80+/CD64+ > CD200+/CD206+) associated with proinflammatory cytokines response (IL-6+TNF-α >> Arg-1+IL-10), and these were also detected in the myocardium of chronically infected mice. Infected mice treated with SRT1720 (vs. infected/untreated) exhibited decreased splenic expansion and myocardial infiltration of proinflammatory Mo/Mφ. SRT1720 did not alter the inherent capability of splenic Mo/Mφ of Chagas mice to respond to pathogen stimulus. Instead, SRT1720 dampened the Tc-induced increase in the expression and/or phosphorylation of focal adhesion kinase (FAK) and downstream transcription factors (Pu.1, c-Myb, and Runx1) involved in Mφ proliferation and migration and Notch1 involved in functional activation. Studies in cultured Mφ confirmed the agonistic effects of SIRT1 in controlling the Tc-induced, FAK-dependent increase in the expression of transcription factors and showed that SIRT1 agonist and FAK inhibitor abrogated the NF-κB transcriptional activity and inflammatory cytokine gene expression in Tc-infected Mφ. CONCLUSIONS: The proinflammatory Mo/Mφ of yolk sac origin drive the splenic and tissue inflammatory response in chronic CD. SRT1720 reprogrammed the Tc-induced FAK-dependent transcription factors involved in Mφ proliferation and proinflammatory activation in Chagas disease.


Subject(s)
Chagas Disease/complications , Chagas Disease/metabolism , Inflammation/complications , Inflammation/metabolism , Macrophages/metabolism , Monocytes/metabolism , Animals , Biomarkers , Chagas Disease/mortality , Chagas Disease/parasitology , Chronic Disease , Cytokines/biosynthesis , Disease Models, Animal , Disease Susceptibility , Female , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Granulocyte-Macrophage Progenitor Cells/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Immunophenotyping , Inflammation/parasitology , Inflammation Mediators/metabolism , Macrophage Activation/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Monocytes/drug effects , Monocytes/immunology , Monocytes/pathology , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism , NF-kappaB-Inducing Kinase
11.
Stem Cells ; 37(3): 430-440, 2019 03.
Article in English | MEDLINE | ID: mdl-30537419

ABSTRACT

Previously, we reported that although the HSPC frequency in bone marrow cells (BMC) was comparable between ß2-/- and ß2+/+ mice, transplantation of ß2-/- BMC into lethally irradiated CD45.1 recipient resulted in more myeloid cell production than ß2+/+ BMC. The objective of this study is to address if integrin ß2 deficiency skews granulocyte/macrophage progenitor (GMP) proliferation. FACS analysis demonstrated that GMP frequency and cell number were higher and megakaryocyte/erythrocyte progenitor frequency and cell number were lower in ß2-/- mice than ß2+/+ mice. However, the common myeloid progenitors (CMP) frequency and cell number were similar between the two groups. The increased GMP number was due to GMP proliferation as evidenced by the percentage of BrdU-incorporating GMP. Whole genome transcriptome analysis identified increased FcεRIα expression in ß2-/- CMP compared to ß2+/+ CMP. FcεRIα expression on ß2-/- GMP was detected increased in ß2-/- mice by qRT-PCR and FACS. Although transplantation of FcεRIαhi GMP or FcεRIαlo GMP into lethally irradiated CD45.1 recipient resulted in comparable myeloid cell production, transplantation of ß2 deficient FcεRIαhi GMP generated more myeloid cells than ß2+/+ FcεRIαhi GMP. GATA2 expression was increased in ß2-/- GMP. Using a luciferase reporter assay, we demonstrated that mutation of the GATA2 binding site in the FcεRIα promoter region diminished FcεRIα transcription. In vitro, the addition of IgE, the ligand of FcεRIα, promoted GMP expansion, which was abrogated by inhibition of JNK phosphorylation. Integrin ß2 deficiency promoted GMP proliferation and myeloid cell production, which was mediated via FcεRIα/IgE-induced JNK phosphorylation in GMP. Stem Cells 2019;37:430-440.


Subject(s)
CD18 Antigens/metabolism , Cell Proliferation , Granulocyte-Macrophage Progenitor Cells/metabolism , Animals , CD18 Antigens/genetics , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gene Expression Regulation , MAP Kinase Kinase 4 , Mice , Mice, Knockout , Receptors, IgE/biosynthesis , Receptors, IgE/genetics , Transcription, Genetic
12.
Oncogene ; 38(5): 687-698, 2019 01.
Article in English | MEDLINE | ID: mdl-30171262

ABSTRACT

Myelodysplastic syndromes (MDSs) are stem cell disorders with risk of transformation to acute myeloid leukemia (AML). Gene expression profiling reveals transcriptional expression of GLI1, of Hedgehog (Hh) signaling, in poor-risk MDS/AML. Using a murine model of MDS we demonstrated that constitutive Hh/Gli1 activation accelerated leukemic transformation and decreased overall survival. Hh/Gli1 activation resulted in clonal expansion of phenotypically defined granulocyte macrophage progenitors (GMPs) and acquisition of self-renewal potential in a non-self-renewing progenitor compartment. Transcriptome analysis of GMPs revealed enrichment in gene signatures of self-renewal pathways, operating via direct Gli1 activation. Using human cell lines we demonstrated that in addition to canonical Hh signaling, GLI1 is activated in a Smoothened-independent manner. GLI1 knockdown or inhibition with GANT61 resulted in decreased proliferation and clonogenic potential. Our data suggest that GLI1 activation is frequent in MDS during disease progression and inhibition of GLI1 is an attractive therapeutic target for a subset of patients.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Granulocyte-Macrophage Progenitor Cells/metabolism , Hedgehog Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Myelodysplastic Syndromes/metabolism , Zinc Finger Protein GLI1/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Granulocyte-Macrophage Progenitor Cells/pathology , Hedgehog Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Pyridines/pharmacology , Pyrimidines/pharmacology , Zinc Finger Protein GLI1/genetics
13.
Immunity ; 49(1): 93-106.e7, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29958804

ABSTRACT

There is a growing body of research on the neural control of immunity and inflammation. However, it is not known whether the nervous system can regulate the production of inflammatory myeloid cells from hematopoietic progenitor cells in disease conditions. Myeloid cell numbers in diabetic patients were strongly correlated with plasma concentrations of norepinephrine, suggesting the role of sympathetic neuronal activation in myeloid cell production. The spleens of diabetic patients and mice contained higher numbers of tyrosine hydroxylase (TH)-expressing leukocytes that produced catecholamines. Granulocyte macrophage progenitors (GMPs) expressed the ß2 adrenergic receptor, a target of catecholamines. Ablation of splenic sympathetic neuronal signaling using surgical, chemical, and genetic approaches diminished GMP proliferation and myeloid cell development. Finally, mice lacking TH-producing leukocytes had reduced GMP proliferation, resulting in diminished myelopoiesis. Taken together, our study demonstrates that catecholamines produced by leukocytes and sympathetic nerve termini promote GMP proliferation and myeloid cell development.


Subject(s)
Diabetes Mellitus/physiopathology , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/metabolism , Myelopoiesis , Neuroimmunomodulation , Sympathetic Nervous System/metabolism , Adrenergic beta-2 Receptor Antagonists/pharmacology , Animals , Cell Proliferation/drug effects , Diabetes Mellitus/blood , Disease Models, Animal , Female , Humans , Leukocytes/enzymology , Leukocytes/metabolism , Male , Mice , Myeloid Cells/cytology , Myelopoiesis/drug effects , Neuroimmunomodulation/drug effects , Norepinephrine/blood , Signal Transduction/drug effects , Spleen/cytology , Spleen/innervation , Spleen/metabolism , Sympathetic Nervous System/drug effects
14.
J Leukoc Biol ; 104(1): 123-133, 2018 07.
Article in English | MEDLINE | ID: mdl-29645346

ABSTRACT

In recent years multi-parameter flow cytometry has enabled identification of cells at major stages in myeloid development; from pluripotent hematopoietic stem cells, through populations with increasingly limited developmental potential (common myeloid progenitors and granulocyte-macrophage progenitors), to terminally differentiated mature cells. Myeloid progenitors are heterogeneous, and the surface markers that define transition states from progenitors to mature cells are poorly characterized. Siglec-F is a surface glycoprotein frequently used in combination with IL-5 receptor alpha (IL5Rα) for the identification of murine eosinophils. Here, we describe a CD11b+ Siglec-F+ IL5Rα- myeloid population in the bone marrow of C57BL/6 mice. The CD11b+ Siglec-F+ IL5Rα- cells are retained in eosinophil deficient PHIL mice, and are not expanded upon overexpression of IL-5, indicating that they are upstream or independent of the eosinophil lineage. We show these cells to have GMP-like developmental potential in vitro and in vivo, and to be transcriptionally distinct from the classically described GMP population. The CD11b+ Siglec-F+ IL5Rα- population expands in the bone marrow of Myb mutant mice, which is potentially due to negative transcriptional regulation of Siglec-F by Myb. Lastly, we show that the role of Siglec-F may be, at least in part, to regulate GMP viability.


Subject(s)
Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Animals , Cell Differentiation/physiology , Mice , Mice, Inbred C57BL
15.
Eur J Immunol ; 48(5): 861-873, 2018 05.
Article in English | MEDLINE | ID: mdl-29315532

ABSTRACT

Basophils have been recently recognized to play important roles in type 2 immune responses during allergies and parasitic infection, largely due to the development of novel tools for the in vivo study of these cells. As such, the genetically-engineered MCPT8DTR mouse line has been used to specifically deplete basophils following treatment with diphtheria toxin (DT). In this study, we showed that DT-injected MCPT8DTR mice exhibited a striking decrease of eosinophils and neutrophils in skin when subjected to a hapten fluorescein isothiocyanate (FITC)-induced allergic contact dermatitis (ACD) experimental protocol. Unexpectedly, we found that loss of skin eosinophils and neutrophils was not due to a lack of basophil-mediated recruitment, as DT injection caused a systemic reduction of eosinophils and neutrophils in MCPT8DTR mice in a time-dependent manner. Furthermore, we found that hematopoietic stem-cell-derived granulocyte-macrophage progenitors (GMPs) expressed MCPT8 gene, and that these cells were depleted upon DT injection. Finally, we optimized a protocol in which a low-dose DT achieved a better specificity for depleting basophils, but not GMPs, in MCPT8DTR mice, and demonstrate that basophils do not play a major role in recruiting eosinophils and neutrophils to ACD skin. These data provide new and valuable information about functional studies of basophils.


Subject(s)
Basophils/immunology , Dermatitis, Allergic Contact/immunology , Diphtheria Toxin/toxicity , Eosinophils/immunology , Granulocyte-Macrophage Progenitor Cells/cytology , Neutrophils/immunology , Tryptases/metabolism , Animals , Basophils/cytology , Eosinophils/cytology , Female , Granulocyte-Macrophage Progenitor Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neutrophils/cytology , Tryptases/genetics
16.
Toxicol Sci ; 160(2): 268-283, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28973681

ABSTRACT

Lead (Pb) is a toxic heavy metal affecting human health; it is known to be harmful to various organs or systems, yet the mechanisms by which Pb influences immune cell development remain to be defined. In this study, we show that Pb exposure (1250 ppm via drinking water) selectively impacted the development of myeloid cells (myelopoiesis). After Pb treatment of adult C57BL/6 mice, the numbers of granulocyte-macrophage progenitors (GMP) were consistently reduced, whereas the numbers of myeloid cells were increased at week (wk) 1 and decreased at wk8 after initiating the Pb exposure. Functional assays indicate that Pb accelerated GMP differentiation in a reactive oxygen species-dependent manner after treatment for 1 week and inhibited common myeloid progenitor differentiation by upregulating interferon regulatory factor 8 (IRF8) expression after treatment for 8 weeks. Consistent with the distinct Pb influences on myeloid cells observed at wk1 and wk8, Pb caused an inflammatory environment in vivo at wk8, but not at wk1. Furthermore, like the observations in mice during the Pb exposure, bloods from humans occupationally exposed to Pb had their numbers of monocytes, neutrophils and GMP negatively associated with the Pb concentration, whereas IRF8 expression in common myeloid progenitor, but not GMP, was positively correlated with the Pb concentration. These data suggest an occupationally relevant level of Pb exposure preferentially influences myelopoiesis involving reactive oxygen species and IRF8, which may contribute to the current understanding of the hematopoietic toxicology of Pb.


Subject(s)
Cell Lineage/drug effects , Environmental Pollutants/adverse effects , Granulocyte-Macrophage Progenitor Cells/drug effects , Myeloid Progenitor Cells/drug effects , Myelopoiesis/drug effects , Occupational Exposure/adverse effects , Organometallic Compounds/adverse effects , Animals , Cells, Cultured , Coculture Techniques , Environmental Pollutants/blood , Female , Granulocyte-Macrophage Progenitor Cells/metabolism , Granulocyte-Macrophage Progenitor Cells/pathology , Humans , Interferon Regulatory Factors/metabolism , Leukocyte Count , Male , Mice, Inbred C57BL , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Organometallic Compounds/blood , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
17.
Blood ; 129(15): 2124-2131, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28130213

ABSTRACT

Hematopoietic stem cells (HSCs) reside in and are maintained by special microenvironments, termed niches. It is assumed that the HSC niche space remains occupied by endogenous cells and that myelosuppressive conditioning is required to achieve high levels of HSC engraftment. We herein demonstrate that upon the transplantation of very large numbers of purified HSCs into normal mice not exposed to myeloablation, donor HSCs engrafted in niches distant from filled HSC niches without replacing host HSCs and subsequently proliferated and generated hematopoietic progenitors, leading to marked increases in the overall HSC numbers in bone marrow. Additionally, stem cell factor that is produced by CXC chemokine ligand 12-abundant reticular cells is involved in HSC engraftment. In contrast, host granulocyte/macrophage progenitors (GMPs) were replaced by the progeny of transplanted donor HSCs, and overall GMP numbers remained unchanged. Thus, inconsistent with the classical concept, numerous empty HSC niches are available for engraftment and proliferation in bone marrow.


Subject(s)
Graft Survival , Granulocyte-Macrophage Progenitor Cells/metabolism , Hematopoietic Stem Cell Transplantation , Homeostasis , Stem Cell Niche , Transplantation Conditioning , Allografts , Animals , Bone Marrow/metabolism , Chemokine CXCL12/metabolism , Mice , Mice, Transgenic
18.
Stem Cell Reports ; 8(2): 346-359, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28111277

ABSTRACT

The lymphoid-myeloid transdifferentiation potentials of members of the C/EBP family (C/EBPα, ß, δ, and ε) were compared in v-Abl-immortalized primary B cells. Conversion of B cells to macrophages was readily induced by the ectopic expression of any C/EBP, and enhanced by endogenous C/EBPα and ß activation. High transgene expression of C/EBPß or C/EBPε, but not of C/EBPα or C/EBPδ, also induced the formation of granulocytes. Granulocytes and macrophages emerged in a mutually exclusive manner. C/EBPß-expressing B cells produced granulocyte-macrophage progenitor (GMP)-like progenitors when subjected to selective pressure to eliminate lymphoid cells. The GMP-like progenitors remained self-renewing and cytokine-independent, and continuously produced macrophages and granulocytes. In addition to their suitability to study myelomonocytic lineage bifurcation, lineage-switched GMP-like progenitors could reflect the features of the lympho-myeloid lineage switch observed in leukemic progression.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Cell Transdifferentiation/genetics , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , Cell Lineage/genetics , Cell Proliferation , Gene Dosage , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Mice , Multigene Family , Phenotype
19.
Nature ; 541(7635): 96-101, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28002407

ABSTRACT

Monocytes and macrophages comprise a variety of subsets with diverse functions. It is thought that these cells play a crucial role in homeostasis of peripheral organs, key immunological processes and development of various diseases. Among these diseases, fibrosis is a life-threatening disease of unknown aetiology. Its pathogenesis is poorly understood, and there are few effective therapies. The development of fibrosis is associated with activation of monocytes and macrophages. However, the specific subtypes of monocytes and macrophages that are involved in fibrosis have not yet been identified. Here we show that Ceacam1+Msr1+Ly6C-F4/80-Mac1+ monocytes, which we term segregated-nucleus-containing atypical monocytes (SatM), share granulocyte characteristics, are regulated by CCAAT/enhancer binding protein ß (C/EBPß), and are critical for fibrosis. Cebpb deficiency results in a complete lack of SatM. Furthermore, the development of bleomycin-induced fibrosis, but not inflammation, was prevented in chimaeric mice with Cebpb-/- haematopoietic cells. Adoptive transfer of SatM into Cebpb-/- mice resulted in fibrosis. Notably, SatM are derived from Ly6C-FcεRI+ granulocyte/macrophage progenitors, and a newly identified SatM progenitor downstream of Ly6C-FcεRI+ granulocyte/macrophage progenitors, but not from macrophage/dendritic-cell progenitors. Our results show that SatM are critical for fibrosis and that C/EBPß licenses differentiation of SatM from their committed progenitor.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Granulocyte-Macrophage Progenitor Cells/cytology , Monocytes/classification , Monocytes/metabolism , Pulmonary Fibrosis/pathology , Adoptive Transfer , Animals , Antigens, CD/metabolism , Antigens, Ly/metabolism , Biomarkers/metabolism , Bleomycin/toxicity , CCAAT-Enhancer-Binding Protein-beta/deficiency , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Adhesion Molecules/metabolism , Cell Differentiation , Dendritic Cells/cytology , Disease Models, Animal , Granulocyte-Macrophage Progenitor Cells/metabolism , Granulocytes/cytology , Granulocytes/metabolism , Inflammation , Male , Mice , Molecular Targeted Therapy/trends , Monocytes/pathology , Monocytes/transplantation , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/prevention & control , Receptors, IgE/metabolism , Scavenger Receptors, Class A/metabolism
20.
J Exp Med ; 213(8): 1513-35, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27377587

ABSTRACT

Our understanding of the perturbation of normal cellular differentiation hierarchies to create tumor-propagating stem cell populations is incomplete. In human acute myeloid leukemia (AML), current models suggest transformation creates leukemic stem cell (LSC) populations arrested at a progenitor-like stage expressing cell surface CD34. We show that in ∼25% of AML, with a distinct genetic mutation pattern where >98% of cells are CD34(-), there are multiple, nonhierarchically arranged CD34(+) and CD34(-) LSC populations. Within CD34(-) and CD34(+) LSC-containing populations, LSC frequencies are similar; there are shared clonal structures and near-identical transcriptional signatures. CD34(-) LSCs have disordered global transcription profiles, but these profiles are enriched for transcriptional signatures of normal CD34(-) mature granulocyte-macrophage precursors, downstream of progenitors. But unlike mature precursors, LSCs express multiple normal stem cell transcriptional regulators previously implicated in LSC function. This suggests a new refined model of the relationship between LSCs and normal hemopoiesis in which the nature of genetic/epigenetic changes determines the disordered transcriptional program, resulting in LSC differentiation arrest at stages that are most like either progenitor or precursor stages of hemopoiesis.


Subject(s)
Antigens, CD34/genetics , Granulocyte-Macrophage Progenitor Cells/metabolism , Leukemia, Myeloid, Acute , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Animals , Antigens, CD34/metabolism , Granulocyte-Macrophage Progenitor Cells/pathology , Heterografts , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Neoplastic Stem Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL