Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 102
1.
Shock ; 54(3): 386-393, 2020 09.
Article En | MEDLINE | ID: mdl-31764619

Myocardial injury in sepsis may be caused by a burst of several inflammatory mediators, leading to vascular endothelial injuries. However, the contribution of neutrophil elastase (NE) to myocardial injury in sepsis is still unknown. We aimed to evaluate whether endotoxemia-induced myocardial injury is associated with NE. Lipopolysaccharide (LPS) was injected intraperitoneally at a dose of 20 mg/kg into granulocyte-colony-stimulating-factor knockout mice (G-CSF-KO), which have few neutrophils, and littermate control mice. The survival rate of G-CSF-KO mice 48 hours after LPS injection was significantly greater than that of control mice. The serum level of troponin I in G-CSF-KO mice was significantly lower than that in control mice. In addition, the concentration of inflammatory cytokine interleukin-6 (IL-6) was significantly decreased 6 and 12 hours after LPS administration compared with that in control mice. Ultrastructural analysis revealed that vascular endothelial structures and the endothelial glycocalyx in G-CSF-KO mice were clearly preserved. Next, mice were injected with 0.2 mg/kg sivelestat (an NE inhibitor) after LPS administration. The survival rate was significantly higher and the serum level of troponin I was lower in sivelestat-injected mice than in control mice, respectively. Furthermore, IL-6 levels were significantly decreased 6 and 12 hours after LPS administration compared with those in control mice. Vascular endothelial structures and the endothelial glycocalyx in sivelestat-treated mice were clearly preserved at the ultrastructural level. In conclusion, NE is significantly associated with myocardial injury in endotoxemia. Inhibition of NE may be a useful tool for the management of endotoxemia.


Endotoxemia/drug therapy , Glycocalyx/metabolism , Leukocyte Elastase/antagonists & inhibitors , Leukocyte Elastase/metabolism , Animals , Endotoxemia/blood , Endotoxins/toxicity , Glycine/analogs & derivatives , Glycine/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Interleukin-6/blood , Male , Mice , Mice, Knockout , Microscopy, Electron , Sulfonamides/therapeutic use , Troponin I/blood
2.
Alcohol ; 80: 73-79, 2019 11.
Article En | MEDLINE | ID: mdl-31229291

Chronic alcohol consumption renders the lung more susceptible to infections by disrupting essential alveolar macrophage functions. Emerging evidence suggests that these functional deficits are due, in part, to a suppression of GM-CSF signaling, which is believed to compromise monocyte growth and maturation in the lung. However, in addition to controlling monocyte behaviors, GM-CSF also regulates surfactant homeostasis. For example, mice with targeted deletion of the gene for GM-CSF accumulate large amounts of surfactant phospholipids in their lungs. Moreover, decreased GM-CSF signaling in humans has been linked to the development of pulmonary alveolar proteinosis (PAP), a rare disorder in which surfactant lipids and proteins accumulate in alveolar macrophages and the lung exhibits enhanced susceptibility to infection. Consistent with parallel mechanisms in the PAP and alcoholic lung, we have recently reported that levels of intrapulmonary lipids, specifically triglycerides and free fatty acids, are increased in BAL fluid, whole lung digests and alveolar macrophages of chronically alcohol exposed rats. Additionally, we showed that uptake of saturated fatty acids alone could induce phenotypic and functional changes in alveolar macrophages that mimicked those in the alcohol-exposed rat and human lung. Herein, we discuss the role of GM-CSF in surfactant homeostasis and highlight the evidence that links decreased GM-CSF signaling to alveolar macrophage dysfunction in both the PAP and alcohol-exposed lung. Moreover, we discuss how lipid accumulation itself might contribute to altering alveolar macrophage function and propose how targeting these mechanisms could be employed for reducing the susceptibility to pulmonary infections in alcoholics.


Alcoholism/complications , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Lung/pathology , Pulmonary Alveolar Proteinosis/etiology , Alcoholism/pathology , Animals , Homeostasis , Lung/drug effects , Macrophages, Alveolar/pathology , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Surfactants/metabolism
3.
Am J Physiol Renal Physiol ; 317(7): F23-F29, 2019 07 01.
Article En | MEDLINE | ID: mdl-30943070

The etiology of chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) is still unknown. Granulocyte macrophage colony-stimulating factor (GM-CSF) has been shown to play an important role in the development of autoimmune and inflammatory diseases. Here, we investigated the expression and function of GM-CSF in patients with CP/CPPS and in a mouse model of experimental autoimmune prostatitis (EAP). GM-CSF mRNA levels were detected in expressed prostatic secretions samples from patients with CP/CPPS and in prostate tissue from a mouse model of EAP. The expression of GM-CSF receptor in mouse prostate and dorsal root ganglia were determined using PCR and immunohistochemistry. Behavioral testing and inflammation scoring were performed to evaluate the role of GM-CSF in disease development and symptom severity of EAP using GM-CSF knockout mice. mRNA levels of putative nociceptive and inflammatory markers were measured in the prostate after the induction of EAP. Elevated GM-CSF mRNA levels were observed in expressed prostatic secretions samples from patients with CP/CPPS compared with healthy volunteers. GM-CSF mRNA was also significantly increased in prostate tissue of the EAP mice model. The expression of GM-CSF receptors was confirmed in mouse prostate and dorsal root ganglia. GM-CSF knockout mice showed fewer Infiltrating leukocytes and pain symptoms after the induction of EAP. Deletion of GM-CSF significantly diminished EAP-induced increases of chemokine (C-C motif) ligand 2, chemokine (C-C motif) ligand 3, and nerve growth factor mRNA expression. The results indicated that GM-CSF plays a functional role in the pathogenesis of EAP. GM-CSF may function as a signaling mediator for both inflammation and pain transduction in CP/CPPS.


Autoimmune Diseases/physiopathology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Prostatitis/immunology , Animals , Autoimmune Diseases/etiology , Chronic Pain , Disease Models, Animal , Ganglia, Spinal/chemistry , Ganglia, Spinal/metabolism , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Male , Mice , Mice, Knockout , Pelvic Pain , Prostate/chemistry , Prostate/metabolism , Prostatitis/physiopathology , RNA, Messenger/analysis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Semen/chemistry
4.
Immunity ; 50(4): 796-811, 2019 04 16.
Article En | MEDLINE | ID: mdl-30995500

The ß common chain cytokines GM-CSF, IL-3, and IL-5 regulate varied inflammatory responses that promote the rapid clearance of pathogens but also contribute to pathology in chronic inflammation. Therapeutic interventions manipulating these cytokines are approved for use in some cancers as well as allergic and autoimmune disease, and others show promising early clinical activity. These approaches are based on our understanding of the inflammatory roles of these cytokines; however, GM-CSF also participates in the resolution of inflammation, and IL-3 and IL-5 may also have such properties. Here, we review the functions of the ß common cytokines in health and disease. We discuss preclinical and clinical data, highlighting the potential inherent in targeting these cytokine pathways, the limitations, and the important gaps in understanding of the basic biology of this cytokine family.


Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Inflammation/immunology , Interleukin-3/immunology , Interleukin-5/immunology , Animals , Autoimmune Diseases/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Hematopoiesis/immunology , Humans , Inflammation/therapy , Interleukin-3/antagonists & inhibitors , Interleukin-3/deficiency , Interleukin-3/genetics , Interleukin-5/antagonists & inhibitors , Interleukin-5/deficiency , Interleukin-5/genetics , Mice , Mice, Knockout , Multigene Family , Neoplasms/immunology , Neoplasms/therapy , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/immunology , Receptors, Interleukin-5/genetics , Receptors, Interleukin-5/immunology , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Signal Transduction , Structure-Activity Relationship , Vaccination , Wound Healing/immunology
5.
J Exp Med ; 213(10): 1983-98, 2016 09 19.
Article En | MEDLINE | ID: mdl-27595596

Kawasaki disease (KD) is the leading cause of pediatric heart disease in developed countries. KD patients develop cardiac inflammation, characterized by an early infiltrate of neutrophils and monocytes that precipitates coronary arteritis. Although the early inflammatory processes are linked to cardiac pathology, the factors that regulate cardiac inflammation and immune cell recruitment to the heart remain obscure. In this study, using a mouse model of KD (induced by a cell wall Candida albicans water-soluble fraction [CAWS]), we identify an essential role for granulocyte/macrophage colony-stimulating factor (GM-CSF) in orchestrating these events. GM-CSF is rapidly produced by cardiac fibroblasts after CAWS challenge, precipitating cardiac inflammation. Mechanistically, GM-CSF acts upon the local macrophage compartment, driving the expression of inflammatory cytokines and chemokines, whereas therapeutically, GM-CSF blockade markedly reduces cardiac disease. Our findings describe a novel role for GM-CSF as an essential initiating cytokine in cardiac inflammation and implicate GM-CSF as a potential target for therapeutic intervention in KD.


Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Inflammation/pathology , Mucocutaneous Lymph Node Syndrome/pathology , Myocardium/pathology , Animals , Candida albicans/metabolism , Cell Compartmentation , Chemokines/metabolism , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Hematopoiesis , Inflammation/complications , Inflammation/genetics , Kinetics , Mice, Inbred C57BL , Monocytes/pathology , Mucocutaneous Lymph Node Syndrome/complications , Myocardium/metabolism , Neutrophils/pathology , Organ Specificity , Radiation Tolerance , Signal Transduction , Vasculitis/complications , Vasculitis/pathology
6.
J Leukoc Biol ; 100(4): 747-760, 2016 10.
Article En | MEDLINE | ID: mdl-27256565

Previous studies established that GM-CSF-deficient (Csf2-deficient) mice exhibit profound resistance to experimental autoimmune encephalomyelitis. This study addressed whether the resistance of Csf2-deficient mice was a result of a requirement for GM-CSF in controlling the functional balance between effector and regulatory T cell subsets during experimental autoimmune encephalomyelitis. The main observation was that treatment with the anti-CD25 mAb PC61 rendered Csf2-deficient mice fully susceptible to severe, chronic experimental autoimmune encephalomyelitis, with disease incidences and severities equivalent to that of C57BL/6 mice. When both donors and recipients were treated with PC61 in a passive model of experimental autoimmune encephalomyelitis, adoptive transfer of myelin-specific Csf2-deficient T cells into Csf2-deficient recipients resulted in a nonresolving chronic course of severe paralytic experimental autoimmune encephalomyelitis. The peripheral Csf2-deficient T cell repertoire was marked by elevated CD3+ T cell frequencies that reflected substantial accumulations of naïve CD44null-low CD4+ and CD8+ T cells but essentially normal frequencies of CD4+ CD25+ forkhead box P3+ T cells among the CD3+ T cell pool. These findings suggested that Csf2-deficient mice had secondary deficiencies in peripheral T cell sensitization to environmental antigens. In accordance, myelin oligodendrocyte glycoprotein 35-55/CFA-sensitized Csf2-deficient mice exhibited deficient peripheral sensitization to myelin oligodendrocyte glycoprotein, whereas pretreatment of Csf2-deficient mice with PC61 enabled the robust induction of myelin oligodendrocyte glycoprotein-specific T cell responses in the draining lymphatics. In conclusion, the experimental autoimmune encephalomyelitis resistance of Csf2-deficient mice, at least in part, reflects a deficient induction of effector T cell function that cannot surmount normal regulatory T cell barriers. Experimental autoimmune encephalomyelitis effector responses, however, are unleashed upon depletion of regulatory CD25+ T cells.


CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Lymphocyte Depletion , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/toxicity , Disease Susceptibility , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Granulocytes/immunology , Immunophenotyping , Interleukin-2 Receptor alpha Subunit/analysis , Interleukin-2 Receptor alpha Subunit/immunology , Leukocyte Count , Lymphocyte Count , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/immunology
7.
Arch. bronconeumol. (Ed. impr.) ; 51(7): 344-349, jul. 2015. ilus, tab
Article Es | IBECS | ID: ibc-138231

La proteinosis alveolar pulmonar es una enfermedad rara que se caracteriza por la acumulación del material lipoproteináceo del surfactante en los espacios alveolares y los bronquiolos terminales, lo que puede llegar a producir alteraciones en el intercambio gaseoso. Esta acumulación del surfactante es debida a una disminución en su aclaramiento por parte de los macrófagos alveolares. Su forma primaria, la más frecuente, es considerada actualmente una enfermedad autoinmune. El mayor conocimiento de las causas que la provocan ha conducido a la aparición de tratamientos alternativos al lavado pulmonar total, que sigue siendo considerado de elección. La mayoría de los trabajos están constituidos por series de casos, la mayoría de las veces con pocos pacientes, por lo que el nivel de evidencia es bajo. Dado que la gravedad de su presentación y su curso son variables, no todos los pacientes van a requerir tratamiento. Debido al bajo nivel de evidencia de que se dispone, ya que la mayoría de estudios son series de casos, se han propuesto de manera arbitraria algunos criterios objetivos, basados en la opinión de expertos, que intentan definir en qué pacientes es más adecuado iniciar el tratamiento


Pulmonary alveolar proteinosis (PAP) is a rare disease characterized by the accumulation of surfactantlike lipoproteinaceous material in the distal air spaces and terminal bronchi, which may lead to impaired gas exchange. This accumulation of surfactantis due to decreased clearance by the alveolar macrophages. Its primary, most common form, is currently considered an autoimmune disease. Better knowledge ofthe causes of PAP have led to the emergence of alternatives to whole lung lavage, although this is still considered the treatment of choice. Most studies are case series, often with limited patient numbers, so the level of evidence is low. Since the severity of presentation and clinical course are variable, not all patients will require treatment. Due to the low level of evidence, some objective criteria based on expert opinion have been arbitrarily proposed in an attempt to define in which patients it is best to initiate treatment


Humans , Pulmonary Alveolar Proteinosis/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Antibodies, Monoclonal/therapeutic use , Rare Diseases/therapy , Bronchoalveolar Lavage , Autoimmunity/immunology , Patient Selection , Hematopoietic Stem Cell Transplantation
8.
Arch Bronconeumol ; 51(7): 344-9, 2015 Jul.
Article En, Es | MEDLINE | ID: mdl-25896950

Pulmonary alveolar proteinosis (PAP) is a rare disease characterized by the accumulation of surfactant-like lipoproteinaceous material in the distal air spaces and terminal bronchi, which may lead to impaired gas exchange. This accumulation of surfactant is due to decreased clearance by the alveolar macrophages. Its primary, most common form, is currently considered an autoimmune disease. Better knowledge of the causes of PAP have led to the emergence of alternatives to whole lung lavage, although this is still considered the treatment of choice. Most studies are case series, often with limited patient numbers, so the level of evidence is low. Since the severity of presentation and clinical course are variable, not all patients will require treatment. Due to the low level of evidence, some objective criteria based on expert opinion have been arbitrarily proposed in an attempt to define in which patients it is best to initiate treatment.


Autoimmune Diseases/therapy , Pulmonary Alveolar Proteinosis/therapy , Adult , Allografts , Animals , Antibiotic Prophylaxis , Antibodies, Neutralizing/analysis , Antibodies, Neutralizing/immunology , Autoantibodies/analysis , Autoantibodies/immunology , Autoimmune Diseases/epidemiology , Autoimmune Diseases/immunology , Bronchoalveolar Lavage Fluid/immunology , Clinical Trials as Topic , Combined Modality Therapy , Disease Management , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cell Transplantation , Humans , Immunoglobulin G/analysis , Immunoglobulin G/immunology , Lung Transplantation , Macrophages/pathology , Mice , Mice, Knockout , Oxygen Inhalation Therapy , Plasmapheresis , Prospective Studies , Pulmonary Alveolar Proteinosis/epidemiology , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Alveolar Proteinosis/immunology , Pulmonary Gas Exchange , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Rituximab/therapeutic use , Therapeutic Irrigation
10.
Circ Res ; 116(2): e13-24, 2015 Jan 16.
Article En | MEDLINE | ID: mdl-25348165

RATIONALE: Granulocyte macrophage colony-stimulating factor (GM-CSF, Csf2) is a growth factor for myeloid-lineage cells that has been implicated in the pathogenesis of atherosclerosis and other chronic inflammatory diseases. However, the role of GM-CSF in advanced atherosclerotic plaque progression, the process that gives rise to clinically dangerous plaques, is unknown. OBJECTIVE: To understand the role of GM-CSF in advanced atherosclerotic plaque progression. METHODS AND RESULTS: Ldlr(-/-) mice and Csf2(-/-)Ldlr(-/-) mice were fed a Western-type diet for 12 weeks, and then parameters of advanced plaque progression in the aortic root were quantified. Lesions from the GM-CSF-deficient mice showed a substantial decrease in 2 key hallmarks of advanced atherosclerosis, lesional macrophage apoptosis and plaque necrosis, which indicates that GM-CSF promotes plaque progression. Based on a combination of in vitro and in vivo studies, we show that the mechanism involves GM-CSF-mediated production of interleukin-23, which increases apoptosis susceptibility in macrophages by promoting proteasomal degradation of the cell survival protein Bcl-2 (B-cell lymphoma 2) and by increasing oxidative stress. CONCLUSIONS: In low-density lipoprotein-driven atherosclerosis in mice, GM-CSF promotes advanced plaque progression by increasing macrophage apoptosis susceptibility. This action of GM-CSF is mediated by its interleukin-23-inducing activity rather than its role as a growth factor.


Apoptosis/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Interleukin-23/metabolism , Macrophages/metabolism , Plaque, Atherosclerotic/metabolism , Signal Transduction/physiology , Animals , Intercellular Signaling Peptides and Proteins , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis/metabolism , Necrosis/pathology , Plaque, Atherosclerotic/pathology
11.
J Exp Med ; 211(6): 1243-56, 2014 Jun 02.
Article En | MEDLINE | ID: mdl-24821911

Pneumonia is a major cause of mortality worldwide and a serious problem in critical care medicine, but the immunophysiological processes that confer either protection or morbidity are not completely understood. We show that in response to lung infection, B1a B cells migrate from the pleural space to the lung parenchyma to secrete polyreactive emergency immunoglobulin M (IgM). The process requires innate response activator (IRA) B cells, a transitional B1a-derived inflammatory subset which controls IgM production via autocrine granulocyte/macrophage colony-stimulating factor (GM-CSF) signaling. The strategic location of these cells, coupled with the capacity to produce GM-CSF-dependent IgM, ensures effective early frontline defense against bacteria invading the lungs. The study describes a previously unrecognized GM-CSF-IgM axis and positions IRA B cells as orchestrators of protective IgM immunity.


B-Lymphocyte Subsets/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunoglobulin M/immunology , Pleura/immunology , Pneumonia/immunology , Adult , Animals , B-Lymphocyte Subsets/metabolism , Cell Movement/immunology , Cells, Cultured , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Immunity, Innate/immunology , Immunoglobulin M/deficiency , Immunoglobulin M/genetics , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Pleura/metabolism , Pneumonia/genetics , Pneumonia/metabolism
12.
J Antimicrob Chemother ; 69(4): 1057-64, 2014 Apr.
Article En | MEDLINE | ID: mdl-24222613

OBJECTIVES: Of the non-tuberculous mycobacteria, Mycobacterium abscessus is particularly refractory to antimicrobial therapy and new agents with activity against these pathogens are urgently needed. The screening of candidate antimicrobial agents against M. abscessus requires a relevant and reproducible animal model of chronic infection. Granulocyte-macrophage colony-stimulating factor knockout (GM-CSF KO) mice were used to develop a new animal model of chronic pulmonary M. abscessus infection that can be used for preclinical efficacy testing of antimicrobial drugs. METHODS: GM-CSF KO mice were infected with a clinical isolate of M. abscessus via intrapulmonary aerosol delivery using a microsprayer device. The clinical condition, histology and cfu of M. abscessus-infected GM-CSF KO mice were evaluated over a period of 4 months. Mice were treated with azithromycin (100 mg/kg) by oral gavage and the clinical condition, histology and bacterial burden was determined after 2 weeks of treatment. RESULTS: We show that pulmonary infection of GM-CSF KO mice with M. abscessus results in a chronic pulmonary infection that lends itself to preclinical testing of new antimicrobial drugs against this bacterium. Azithromycin treatment of M. abscessus-infected GM-CSF KO mice resulted in a lower bacterial burden in the lungs and spleen, weight gain and significant improvement in lung pathology. CONCLUSIONS: Intrapulmonary aerosol infection of GM-CSF KO mice with M. abscessus is a useful animal model for studying pathogenesis as well as pre-clinical testing of new compounds against M. abscessus in acute or chronic phases of infection.


Anti-Bacterial Agents/therapeutic use , Azithromycin/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Mycobacterium Infections/drug therapy , Mycobacterium/drug effects , Pneumonia, Bacterial/drug therapy , Animals , Bacterial Load , Chronic Disease , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Histocytochemistry , Lung/microbiology , Lung/pathology , Mice , Mice, Knockout , Mycobacterium Infections/microbiology , Mycobacterium Infections/pathology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Spleen/microbiology
13.
BMC Immunol ; 14: 41, 2013 Sep 17.
Article En | MEDLINE | ID: mdl-24044676

BACKGROUND: Activin A is a pleiotrophic regulatory cytokine, the ablation of which is neonatal lethal. Healthy human alveolar macrophages (AMs) constitutively express activin A, but AMs of patients with pulmonary alveolar proteinosis (PAP) are deficient in activin A. PAP is an autoimmune lung disease characterized by neutralizing autoantibodies to Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF). Activin A can be stimulated, however, by GM-CSF treatment of AMs in vitro. To further explore pulmonary activin A regulation, we examined AMs in bronchoalveolar lavage (BAL) from wild-type C57BL/6 compared to GM-CSF knockout mice which exhibit a PAP-like histopathology. Both human PAP and mouse GM-CSF knockout AMs are deficient in the transcription factor, peroxisome proliferator activated receptor gamma (PPARγ). RESULTS: In sharp contrast to human PAP, activin A mRNA was elevated in mouse GM-CSF knockout AMs, and activin A protein was increased in BAL fluid. Investigation of potential causative factors for activin A upregulation revealed intrinsic overexpression of IFNγ, a potent inducer of the M1 macrophage phenotype, in GM-CSF knockout BAL cells. IFNγ mRNA was not elevated in PAP BAL cells. In vitro studies confirmed that IFNγ stimulated activin A in wild-type AMs while antibody to IFNγ reduced activin A in GM-CSF knockout AMs. Both IFNγ and Activin A were also reduced in GM-CSF knockout mice in vivo after intratracheal instillation of lentivirus-PPARγ compared to control lentivirus vector. Examination of other M1 markers in GM-CSF knockout mice indicated intrinsic elevation of the IFNγ-regulated gene, inducible Nitrogen Oxide Synthetase (iNOS), CCL5, and interleukin (IL)-6 compared to wild-type. The M2 markers, IL-10 and CCL2 were also intrinsically elevated. CONCLUSIONS: Data point to IFNγ as the primary upregulator of activin A in GM-CSF knockout mice which in addition, exhibit a unique mix of M1-M2 macrophage phenotypes.


Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Macrophages, Alveolar/metabolism , Pulmonary Alveolar Proteinosis/metabolism , Activins/genetics , Activins/metabolism , Animals , Bronchoalveolar Lavage Fluid/cytology , Cells, Cultured , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Gene Expression/drug effects , Gene Expression/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Immunohistochemistry , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophages, Alveolar/classification , Macrophages, Alveolar/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Pulmonary Alveolar Proteinosis/genetics , Reverse Transcriptase Polymerase Chain Reaction
14.
J Immunol ; 190(4): 1702-13, 2013 Feb 15.
Article En | MEDLINE | ID: mdl-23325885

GM-CSF is a growth factor that promotes the survival and activation of macrophages and granulocytes, as well as dendritic cell differentiation and survival in vitro. The mechanism by which exogenous GM-CSF ameliorates the severity of Crohn's disease in humans and colitis in murine models has mainly been considered to reflect its activity on myeloid cells. We used GM-CSF-deficient (GM-CSF(-/-)) mice to probe the functional role of endogenous host-produced GM-CSF in a colitis model induced after injury to the colon epithelium. Dextran sodium sulfate (DSS), at doses that resulted in little epithelial damage and mucosal ulceration in wild type mice, caused marked colon ulceration and delayed ulcer healing in GM-CSF(-/-) mice. Colon crypt epithelial cell proliferation in vivo was significantly decreased in GM-CSF(-/-) mice at early times after DSS injury. This was paralleled by decreased expression of crypt epithelial cell genes involved in cell cycle, proliferation, and wound healing. Decreased crypt cell proliferation and delayed ulcer healing in GM-CSF(-/-) mice were rescued by exogenous GM-CSF, indicating the lack of a developmental abnormality in the epithelial cell proliferative response in those mice. Nonhematopoietic cells, and not myeloid cells, produced the GM-CSF important for colon epithelial proliferation after DSS-induced injury, as revealed by bone marrow chimera and dendritic cell-depletion experiments, with colon epithelial cells being the cellular source of GM-CSF. Endogenous epithelial cell-produced GM-CSF has a novel nonredundant role in facilitating epithelial cell proliferation and ulcer healing in response to injury of the colon crypt epithelium.


Bone Marrow Cells/immunology , Cell Proliferation , Colitis, Ulcerative/immunology , Colitis, Ulcerative/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/radiation effects , Colitis, Ulcerative/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Hematopoiesis/genetics , Hematopoiesis/immunology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Radiation Chimera , Time Factors , Wound Healing/genetics , Wound Healing/immunology
15.
Immunity ; 36(6): 1031-46, 2012 Jun 29.
Article En | MEDLINE | ID: mdl-22749353

GM-CSF (Csf-2) is a critical cytokine for the in vitro generation of dendritic cells (DCs) and is thought to control the development of inflammatory DCs and resident CD103(+) DCs in some tissues. Here we showed that in contrast to the current understanding, Csf-2 receptor acts in the steady state to promote the survival and homeostasis of nonlymphoid tissue-resident CD103(+) and CD11b(+) DCs. Absence of Csf-2 receptor on lung DCs abrogated the induction of CD8(+) T cell immunity after immunization with particulate antigens. In contrast, Csf-2 receptor was dispensable for the differentiation and innate function of inflammatory DCs during acute injuries. Instead, inflammatory DCs required Csf-1 receptor for their development. Thus, Csf-2 is important in vaccine-induced CD8(+) T cell immunity through the regulation of nonlymphoid tissue DC homeostasis rather than control of inflammatory DCs in vivo.


Cytokine Receptor Common beta Subunit/physiology , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Inflammation/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Lineage , Cytokine Receptor Common beta Subunit/antagonists & inhibitors , Cytokine Receptor Common beta Subunit/deficiency , Cytokine Receptor Common beta Subunit/genetics , Dendritic Cells/classification , Dendritic Cells/cytology , Encephalomyelitis, Autoimmune, Experimental/immunology , Endotoxemia/immunology , Gene Expression Profiling , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Homeostasis , Lipopolysaccharides/toxicity , Listeriosis/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/transplantation , Organ Specificity , Orthomyxoviridae Infections/immunology , Pneumococcal Infections/immunology , Radiation Chimera , Spleen/immunology , Tamoxifen/pharmacology
16.
J Immunol ; 189(2): 906-15, 2012 Jul 15.
Article En | MEDLINE | ID: mdl-22696442

Pattern recognition receptors for fungi include dectin-1 and mannose receptor, and these mediate phagocytosis, as well as production of cytokines, reactive oxygen species, and the lipid mediator leukotriene B(4) (LTB(4)). The influence of G protein-coupled receptor ligands such as LTB(4) on fungal pattern recognition receptor expression is unknown. In this study, we investigated the role of LTB(4) signaling in dectin-1 expression and responsiveness in macrophages. Genetic and pharmacologic approaches showed that LTB(4) production and signaling through its high-affinity G protein-coupled receptor leukotriene B(4) receptor 1 (BLT1) direct dectin-1-dependent binding, ingestion, and cytokine production both in vitro and in vivo. Impaired responses to fungal glucans correlated with lower dectin-1 expression in macrophages from leukotriene (LT)- and BLT1-deficent mice than their wild-type counterparts. LTB(4) increased the expression of the transcription factor responsible for dectin-1 expression, PU.1, and PU.1 small interfering RNA abolished LTB(4)-enhanced dectin-1 expression. GM-CSF controls PU.1 expression, and this cytokine was decreased in LT-deficient macrophages. Addition of GM-CSF to LT-deficient cells restored expression of dectin-1 and PU.1, as well as dectin-1 responsiveness. In addition, LTB(4) effects on dectin-1, PU.1, and cytokine production were blunted in GM-CSF(-/-) macrophages. Our results identify LTB(4)-BLT1 signaling as an unrecognized controller of dectin-1 transcription via GM-CSF and PU.1 that is required for fungi-protective host responses.


Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Lectins, C-Type/biosynthesis , Leukotriene B4/physiology , Macrophages, Alveolar/immunology , Macrophages, Peritoneal/immunology , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Animals , Candida albicans/immunology , Cells, Cultured , Female , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Leukotriene B4/biosynthesis , Leukotriene B4/deficiency , Macrophages, Alveolar/microbiology , Macrophages, Peritoneal/microbiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/biosynthesis , Receptors, Leukotriene B4/deficiency , Receptors, Leukotriene B4/physiology , Trans-Activators/biosynthesis , Transcription, Genetic/immunology
17.
Mol Immunol ; 52(1): 30-7, 2012 Aug.
Article En | MEDLINE | ID: mdl-22580403

Granulocyte-macrophage colony stimulating factor (GM-CSF) is a cytokine that functions as a hematopoietic growth factor for the generation of white blood cells and is used clinically to stimulate hematopoiesis following chemotherapy. Apart from stimulating production of granulocytes and monocytes/macrophages, GM-CSF has also long been used for in vitro survival/generation of dendritic cells (DCs) from monocytes and bone marrow cells. Evidence has emerged pointing to an additional role for GM-CSF in regulating the function and differential development of several DC subsets. These newly ascribed functions of GM-CSF may underscore its importance in immunity against pathogens as well as initiating/mediating immunopathology in chronic inflammation. Here we summarize recent advances on the role of GM-CSF in regulating the development and function of DC subsets and discuss the biological significance of these new findings.


Dendritic Cells/classification , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Adjuvants, Immunologic/therapeutic use , Animals , Autoimmunity , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Differentiation/physiology , Dendritic Cells/cytology , Dendritic Cells/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Hematopoiesis/drug effects , Hematopoiesis/immunology , Hematopoiesis/physiology , Humans , Inflammation/etiology , Listeria monocytogenes/immunology , Listeria monocytogenes/pathogenicity , Mice , Mice, Knockout , Neoplasms/immunology , Neoplasms/therapy , Recombinant Proteins/pharmacology
18.
Cytokine ; 57(1): 30-1, 2012 Jan.
Article En | MEDLINE | ID: mdl-22129623

Independent studies with GM-CSF-/- mice have concluded that GM-CSF is necessary for normal reproductive outcome and for the maintenance of normal weight. In contrast to the literature we report that GM-CSF-/- and wild type (C57Bl/6) mice over a continuous 12 month period had similar litter size and neonatal survival. Likewise, unlike a literature observation, for the two mouse strains both male and female mice had similar weight gain when fed on a normal chow diet and monitored until 30 weeks of age. It is concluded that GM-CSF is not necessary for an optimal fertility outcome or for normal weight maintenance during development.


Body Weight/physiology , Fertility/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Animals , Animals, Newborn , Female , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Litter Size/physiology , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Obesity/pathology , Survival Analysis
19.
Diabetes ; 60(8): 2134-43, 2011 Aug.
Article En | MEDLINE | ID: mdl-21734017

OBJECTIVE: Previous studies have demonstrated that mice fed a high-fat diet (HFD) develop insulin resistance with proinflammatory macrophage infiltration into white adipose tissue. Concomitantly, adipocytes undergo programmed cell death with the loss of the adipocyte-specific lipid droplet protein perilipin, and the dead/dying adipocytes are surrounded by macrophages that are organized into crown-like structures. This study investigated whether adipocyte cell death provides the driving signal for macrophage inflammation or if inflammation induces adipocyte cell death. RESEARCH DESIGN AND METHODS: Two knockout mouse models were used: granulocyte/monocyte-colony stimulating factor (GM-CSF)-null mice that are protected against HFD-induced adipose tissue inflammation and cyclophilin D (CyP-D)-null mice that are protected against adipocyte cell death. Mice were fed for 4-14 weeks with a 60% HFD, and different markers of cell death and inflammation were analyzed. RESULTS: HFD induced a normal extent of adipocyte cell death in GM-CSF-null mice, despite a marked reduction in adipose tissue inflammation. Similarly, depletion of macrophages by clodronate treatment prevented HFD-induced adipose tissue inflammation without any affect on adipocyte cell death. However, CyP-D deficiency strongly protected adipocytes from HFD-induced cell death, without affecting adipose tissue inflammation. CONCLUSIONS: These data demonstrate that HFD-induced adipocyte cell death is an intrinsic cellular response that is CyP-D dependent but is independent of macrophage infiltration/activation.


Adipocytes/pathology , Cyclophilins/physiology , Dietary Fats/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Macrophages/physiology , 3T3-L1 Cells , Adipose Tissue/pathology , Animals , Peptidyl-Prolyl Isomerase F , Cyclophilins/deficiency , Dietary Fats/adverse effects , Inflammation/physiopathology , Insulin Resistance/physiology , Mice , Mice, Knockout , Signal Transduction/drug effects
20.
J Clin Invest ; 121(6): 2436-46, 2011 Jun.
Article En | MEDLINE | ID: mdl-21537082

Lung cancer is the leading cause of cancer death worldwide. Both principal factors known to cause lung cancer, cigarette smoke and asbestos, induce pulmonary inflammation, and pulmonary inflammation has recently been implicated in several murine models of lung cancer. To further investigate the role of inflammation in the development of lung cancer, we generated mice with combined loss of IFN-γ and the ß-common cytokines GM-CSF and IL-3. These immunodeficient mice develop chronic pulmonary inflammation and lung tumors at a high frequency. Examination of the relationship between these tumors and their inflammatory microenvironment revealed a dual role for the immune system in tumor development. The inflammatory cytokine IL-6 promoted optimal tumor growth, yet wild-type mice rejected transplanted tumors through the induction of adaptive immunity. These findings suggest a model whereby cytokine deficiency leads to oncogenic inflammation that combines with defective antitumor immunity to promote lung tumor formation, representing a unique system for studying the role of the immune system in lung tumor development.


Adenocarcinoma/etiology , Inflammation/complications , Lung Neoplasms/etiology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Animals , Autocrine Communication , Disease Models, Animal , Graft Rejection/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Hematopoietic Stem Cell Transplantation , Immunocompetence , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/genetics , Inflammation/immunology , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interleukin-3/deficiency , Interleukin-3/genetics , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/physiology , Lung Neoplasms/genetics , Lung Neoplasms/immunology , MAP Kinase Signaling System , Mice , Mice, Inbred BALB C , Mice, Knockout , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Neoplasm Transplantation , Radiation Chimera/immunology , STAT3 Transcription Factor/physiology
...