Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 13: 885101, 2022.
Article in English | MEDLINE | ID: mdl-35619690

ABSTRACT

IgG antibodies form immune complexes (IC) that propagate inflammation and tissue damage in autoimmune diseases such as systemic lupus erythematosus. IgG IC engage Fcγ receptors (FcγR) on mononuclear phagocytes (MNP), leading to widespread changes in gene expression that mediate antibody effector function. Bromodomain and extra-terminal domain (BET) proteins are involved in governing gene transcription. We investigated the capacity of BET protein inhibitors (iBET) to alter IgG FcγR-mediated MNP activation. We found that iBET dampened IgG IC-induced pro-inflammatory gene expression and decreased activating FcγR expression on MNPs, reducing their ability to respond to IgG IC. Despite FcγR downregulation, iBET-treated macrophages demonstrated increased phagocytosis of protein antigen, IgG IC, and apoptotic cells. iBET also altered cell morphology, generating more amoeboid MNPs with reduced adhesion. iBET treatment impaired chemotaxis towards a CCL19 gradient in IC-stimulated dendritic cells (DC) in vitro, and inhibited IC-induced DC migration to draining lymph nodes in vivo, in a DC-intrinsic manner. Altogether, our data show that iBET modulates FcγR-mediated MNP activation and migration, revealing the therapeutic potential of BET protein inhibition in antibody-mediated diseases.


Subject(s)
Chemotaxis , Receptors, IgG , Antigen-Antibody Complex , Immunoglobulin G , Macrophages , Receptors, IgG/metabolism
2.
Cell Rep ; 32(1): 107857, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32640223

ABSTRACT

Macrophages play a central role in intestinal immunity, but inappropriate macrophage activation is associated with inflammatory bowel disease (IBD). Here, we identify granulocyte-macrophage colony stimulating factor (GM-CSF) as a critical regulator of intestinal macrophage activation in patients with IBD and mice with dextran sodium sulfate (DSS)-induced colitis. We find that GM-CSF drives the maturation and polarization of inflammatory intestinal macrophages, promoting anti-microbial functions while suppressing wound-healing transcriptional programs. Group 3 innate lymphoid cells (ILC3s) are a major source of GM-CSF in intestinal inflammation, with a strong positive correlation observed between ILC or CSF2 transcripts and M1 macrophage signatures in IBD mucosal biopsies. Furthermore, GM-CSF-dependent macrophage polarization results in a positive feedback loop that augmented ILC3 activation and type 17 immunity. Together, our data reveal an important role for GM-CSF-mediated ILC-macrophage crosstalk in calibrating intestinal macrophage phenotype to enhance anti-bacterial responses, while inhibiting pro-repair functions associated with fibrosis and stricturing, with important clinical implications.


Subject(s)
Enterobacteriaceae Infections/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Inflammation/pathology , Intestines/pathology , Macrophages/pathology , Wound Healing , Animals , Cell Polarity , Citrobacter rodentium/physiology , Colitis/complications , Colitis/immunology , Colitis/pathology , Humans , Immunity, Innate , Lymphocytes/immunology , Macrophage Activation , Mice, Inbred C57BL , Phenotype
3.
Proc Natl Acad Sci U S A ; 117(26): 15160-15171, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32541026

ABSTRACT

IgG antibodies cause inflammation and organ damage in autoimmune diseases such as systemic lupus erythematosus (SLE). We investigated the metabolic profile of macrophages isolated from inflamed tissues in immune complex (IC)-associated diseases, including SLE and rheumatoid arthritis, and following IgG Fcγ receptor cross-linking. We found that human and mouse macrophages undergo a switch to glycolysis in response to IgG IC stimulation, mirroring macrophage metabolic changes in inflamed tissue in vivo. This metabolic reprogramming was required to generate a number of proinflammatory mediators, including IL-1ß, and was dependent on mTOR and hypoxia-inducible factor (HIF)1α. Inhibition of glycolysis, or genetic depletion of HIF1α, attenuated IgG IC-induced activation of macrophages in vitro, including primary human kidney macrophages. In vivo, glycolysis inhibition led to a reduction in kidney macrophage IL-1ß and reduced neutrophil recruitment in a murine model of antibody-mediated nephritis. Together, our data reveal the molecular mechanisms underpinning FcγR-mediated metabolic reprogramming in macrophages and suggest a therapeutic strategy for autoantibody-induced inflammation, including lupus nephritis.


Subject(s)
Cellular Reprogramming/physiology , Lupus Nephritis/metabolism , Animals , Cells, Cultured , Dinoprostone/genetics , Dinoprostone/metabolism , Energy Metabolism , Gene Expression Regulation , Glycolysis/physiology , Humans , Immunoglobulin G/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Kidney/cytology , Macrophages , Mice , Mice, Inbred C57BL , Mice, Knockout , Reactive Oxygen Species , Receptors, IgG/genetics , Receptors, IgG/metabolism
4.
Immunity ; 50(4): 1099-1114.e10, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30876876

ABSTRACT

Inflammatory bowel disease is a chronic, relapsing condition with two subtypes, Crohn's disease (CD) and ulcerative colitis (UC). Genome-wide association studies (GWASs) in UC implicate a FCGR2A variant that alters the binding affinity of the antibody receptor it encodes, FcγRIIA, for immunoglobulin G (IgG). Here, we aimed to understand the mechanisms whereby changes in FcγRIIA affinity would affect inflammation in an IgA-dominated organ. We found a profound induction of anti-commensal IgG and a concomitant increase in activating FcγR signaling in the colonic mucosa of UC patients. Commensal-IgG immune complexes engaged gut-resident FcγR-expressing macrophages, inducing NLRP3- and reactive-oxygen-species-dependent production of interleukin-1ß (IL-1ß) and neutrophil-recruiting chemokines. These responses were modulated by the FCGR2A genotype. In vivo manipulation of macrophage FcγR signal strength in a mouse model of UC determined the magnitude of intestinal inflammation and IL-1ß-dependent type 17 immunity. The identification of an important contribution of IgG-FcγR-dependent inflammation to UC has therapeutic implications.


Subject(s)
Antibodies, Bacterial/immunology , Colitis, Ulcerative/immunology , Gastrointestinal Microbiome/immunology , Immunoglobulin G/immunology , Interleukin-1beta/immunology , Th17 Cells/immunology , Animals , Colitis/chemically induced , Colitis/immunology , Colitis/microbiology , Colitis/pathology , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Dextran Sulfate/toxicity , Gene Expression Regulation , Genotype , Humans , Inflammation , Interleukin-8/biosynthesis , Interleukin-8/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Macrophages/immunology , Mice , Phagocytes/immunology , RNA, Messenger/biosynthesis , Reactive Oxygen Species , Receptors, IgG/biosynthesis , Receptors, IgG/genetics , Receptors, IgG/immunology
5.
Cell ; 170(5): 860-874.e19, 2017 Aug 24.
Article in English | MEDLINE | ID: mdl-28803730

ABSTRACT

Lower urinary tract infections are among the most common human bacterial infections, but extension to the kidneys is rare. This has been attributed to mechanical forces, such as urine flow, that prevent the ascent of bladder microbes. Here, we show that the regional hypersalinity, required for the kidney's urine-concentrating function, instructs epithelial cells to produce chemokines that localize monocyte-derived mononuclear phagocytes (MNPs) to the medulla. This hypersaline environment also increases the intrinsic bactericidal and neutrophil chemotactic activities of MNPs to generate a zone of defense. Because MNP positioning and function are dynamically regulated by the renal salt gradient, we find that patients with urinary concentrating defects are susceptible to kidney infection. Our work reveals a critical accessory role for the homeostatic function of a vital organ in optimizing tissue defense.


Subject(s)
Kidney/immunology , Phagocytes/immunology , Animals , Cell Line , Chemokine CCL2/metabolism , Chemokines/immunology , Diabetes Insipidus , Humans , Kidney/cytology , Kidney Medulla/immunology , Lipopolysaccharide Receptors/metabolism , Mice , Mice, Inbred C57BL , Monocytes/cytology , Salinity , Sodium/metabolism , Transcription Factors/genetics , Urinary Tract Infections/immunology , Urinary Tract Infections/microbiology , Urine/chemistry , Uropathogenic Escherichia coli/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...