Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Clin Immunol ; 42(5): 1093-1105, 2022 07.
Article in English | MEDLINE | ID: mdl-35486340

ABSTRACT

Common variable immunodeficiency disorders (CVID), the most common primary immune deficiency, includes heterogeneous syndromes characterized by hypogammaglobulinemia and impaired antibody responses. CVID patients frequently suffer from recurrent infections and inflammatory conditions. Currently, immunoglobulin replacement therapy (IgRT) is the first-line treatment to prevent infections and aminorate immune alterations in CVID patients. Intravenous Immunoglobulin (IVIg), a preparation of highly purified poly-specific IgG, is used for treatment of immunodeficiencies as well as for autoimmune and inflammatory disorders, as IVIg exerts immunoregulatory and anti-inflammatory actions on innate and adaptive immune cells. To determine the mechanism of action of IVIg in CVID in vivo, we determined the effect of IVIg infusion on the transcriptome of peripheral blood mononuclear cells from CVID patients, and found that peripheral blood monocytes are primary targets of IVIg in vivo, and that IVIg triggers the acquisition of an anti-inflammatory gene profile in human monocytes. Moreover, IVIg altered the relative proportions of peripheral blood monocyte subsets and enhanced the proportion of CD14+ cells with a transcriptional, phenotypic, and functional profile that resembles that of monocytic myeloid-derived suppressor cells (MDSC). Therefore, our results indicate that CD14 + MDSC-like cells might contribute to the immunoregulatory effects of IVIg in CVID and other inflammatory disorders.


Subject(s)
Common Variable Immunodeficiency , Myeloid-Derived Suppressor Cells , Common Variable Immunodeficiency/drug therapy , Humans , Immunoglobulins, Intravenous , Leukocytes, Mononuclear , Monocytes
2.
Eur J Immunol ; 51(4): 824-834, 2021 04.
Article in English | MEDLINE | ID: mdl-33169838

ABSTRACT

CD28 expression is generally considered to be T lymphocyte specific. We have previously shown CD28 mRNA expression in M-CSF-dependent anti-inflammatory monocyte-derived macrophages (M-MØ), and now demonstrate that CD28 cell surface expression is higher in M-MØ than in GM-CSF-dependent macrophages, and that macrophage CD28 expression is regulated by MAFB and activin A. In vivo, CD28 was found in tumor-associated macrophages and, to a lower extent, in pro-inflammatory synovial fluid macrophages from rheumatoid arthritis patients. Analysis of mouse macrophages confirmed Cd28 expression in bone-marrow derived M-MØ. Indeed, anti-CD28 antibodies triggered ERK1/2 phosphorylation in mouse M-MØ. At the functional level, Cd28KO M-MØ exhibited a significantly higher capacity to activate the OVA-specific proliferation of OT-II CD4+ T cells than WT M-MØ, as well as enhanced LPS-induced IL-6 production. Besides, the Cd28KO M-MØ transcriptome was significantly different from WT M-MØ regarding the expression IFN response, inflammatory response, and TGF-ß signaling related gene sets. Therefore, defective CD28 expression in mouse macrophages associates to changes in gene expression profile, what might contribute to the altered functionality displayed by Cd28KO M-MØ. Thus, CD28 expression appears as a hallmark of anti-inflammatory macrophages and might be a target for immunotherapy.


Subject(s)
CD28 Antigens/immunology , Inflammation/immunology , Lymphocyte Activation/immunology , Macrophages/immunology , T-Lymphocytes/immunology , Activins/genetics , Activins/immunology , Activins/metabolism , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , CD28 Antigens/genetics , CD28 Antigens/metabolism , Cells, Cultured , Gene Expression/immunology , Gene Expression Profiling/methods , Humans , Inflammation/genetics , Inflammation/metabolism , Lymphocyte Activation/genetics , Macrophages/metabolism , MafB Transcription Factor/genetics , MafB Transcription Factor/immunology , MafB Transcription Factor/metabolism , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
3.
J Immunol ; 205(3): 776-788, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32591394

ABSTRACT

Growth hormone (GH), a pleiotropic hormone secreted by the pituitary gland, regulates immune and inflammatory responses. In this study, we show that GH regulates the phenotypic and functional plasticity of macrophages both in vitro and in vivo. Specifically, GH treatment of GM-CSF-primed monocyte-derived macrophages promotes a significant enrichment of anti-inflammatory genes and dampens the proinflammatory cytokine profile through PI3K-mediated downregulation of activin A and upregulation of MAFB, a critical transcription factor for anti-inflammatory polarization of human macrophages. These in vitro data correlate with improved remission of inflammation and mucosal repair during recovery in the acute dextran sodium sulfate-induced colitis model in GH-overexpressing mice. In this model, in addition to the GH-mediated effects on other immune cells, we observed that macrophages from inflamed gut acquire an anti-inflammatory/reparative profile. Overall, these data indicate that GH reprograms inflammatory macrophages to an anti-inflammatory phenotype and improves resolution during pathologic inflammatory responses.


Subject(s)
Cellular Reprogramming/immunology , Colitis/immunology , Gene Expression Regulation/immunology , Growth Hormone/immunology , Macrophages/immunology , MafB Transcription Factor/immunology , Animals , Cattle , Cellular Reprogramming/genetics , Colitis/chemically induced , Colitis/genetics , Dextran Sulfate/toxicity , Disease Models, Animal , Growth Hormone/genetics , MafB Transcription Factor/genetics , Mice , Mice, Transgenic
4.
J Immunol ; 201(1): 41-52, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29743313

ABSTRACT

IVIg is an approved therapy for immunodeficiency and for several autoimmune and inflammatory diseases. However, the molecular basis for the IVIg anti-inflammatory activity remains to be fully explained and cannot be extrapolated from studies on animal models of disease. We now report that IVIg impairs the generation of human monocyte-derived anti-inflammatory macrophages by inducing JNK activation and activin A production and limits proinflammatory macrophage differentiation by inhibiting GM-CSF-driven STAT5 activation. In vivo, IVIg provokes a rapid increase in peripheral blood activin A, CCL2, and IL-6 levels, an effect that can be recapitulated in vitro on human monocytes. On differentiating monocytes, IVIg promotes the acquisition of altered transcriptional and cytokine profiles, reduces TLR expression and signaling, and upregulates negative regulators of TLR-initiated intracellular signaling. In line with these effects, in vivo IVIg infusion induces a state tolerant toward subsequent stimuli that results in reduced inflammatory cytokine production after LPS challenge in human peripheral blood and significant protection from LPS-induced death in mice. Therefore, IVIg conditions human macrophages toward the acquisition of a state of cross-tolerance against inflammatory stimuli, an effect that correlates with the net anti-inflammatory action of IVIg in vivo.


Subject(s)
Anti-Inflammatory Agents/immunology , Immune Tolerance/immunology , Immunoglobulins, Intravenous/immunology , Immunoglobulins, Intravenous/pharmacology , Macrophages/immunology , STAT5 Transcription Factor/metabolism , Activins/blood , Animals , Cells, Cultured , Chemokine CCL2/blood , Enzyme Activation , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Inflammation/immunology , Interleukin-6/blood , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/immunology , Mice , Monocytes/cytology , Monocytes/immunology
5.
J Immunol ; 198(5): 2070-2081, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28093525

ABSTRACT

Macrophage phenotypic and functional heterogeneity derives from tissue-specific transcriptional signatures shaped by the local microenvironment. Most studies addressing the molecular basis for macrophage heterogeneity have focused on murine cells, whereas the factors controlling the functional specialization of human macrophages are less known. M-CSF drives the generation of human monocyte-derived macrophages with a potent anti-inflammatory activity upon stimulation. We now report that knockdown of MAFB impairs the acquisition of the anti-inflammatory profile of human macrophages, identify the MAFB-dependent gene signature in human macrophages and illustrate the coexpression of MAFB and MAFB-target genes in CD163+ tissue-resident and tumor-associated macrophages. The contribution of MAFB to the homeostatic/anti-inflammatory macrophage profile is further supported by the skewed polarization of monocyte-derived macrophages from multicentric carpotarsal osteolysis (Online Mendelian Inheritance in Man #166300), a pathology caused by mutations in the MAFB gene. Our results demonstrate that MAFB critically determines the acquisition of the anti-inflammatory transcriptional and functional profiles of human macrophages.


Subject(s)
Cell Differentiation , Hajdu-Cheney Syndrome/immunology , Macrophages/physiology , MafB Transcription Factor/metabolism , Monocytes/physiology , Animals , Anti-Inflammatory Agents , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cell Differentiation/genetics , Cells, Cultured , Cellular Microenvironment , Cytokines/metabolism , Gene Knockdown Techniques , Gene Ontology , Hajdu-Cheney Syndrome/genetics , Homeostasis , Humans , Macrophage Colony-Stimulating Factor/metabolism , MafB Transcription Factor/genetics , Mice , Mutation/genetics , Receptors, Cell Surface/metabolism , Th2 Cells/immunology , Transcriptome
6.
Ann Rheum Dis ; 77(5): 752-759, 2018 05.
Article in English | MEDLINE | ID: mdl-29431121

ABSTRACT

OBJECTIVES: Methotrexate (MTX) is the anchor drug for treatment of rheumatoid arthritis (RA), but the mechanism of its anti-inflammatory action is not fully understood. In RA, macrophages display a proinflammatory polarisation profile that resembles granulocyte-macrophage colony-stimulating factor (GM-CSF)-differentiated macrophages and the response to MTX is only observed in thymidylate synthase+ GM-CSF-dependent macrophages. To determine the molecular basis for the MTX anti-inflammatory action, we explored toll-like receptor (TLR), RA synovial fluid (RASF) and tumour necrosis factor receptor (TNFR)-initiated signalling in MTX-exposed GM-CSF-primed macrophages. METHODS: Intracellular responses to TLR ligands, TNFα or RASF stimulation in long-term low-dose MTX-exposed human macrophages were determined through quantitative real-time PCR, western blot, ELISA and siRNA-mediated knockdown approaches. The role of MTX in vivo was assessed in patients with arthritis under MTX monotherapy and in a murine sepsis model. RESULTS: MTX conditioned macrophages towards a tolerant state, diminishing interleukin (IL)-6 and IL-1ß production in LPS, LTA, TNFα or RASF-challenged macrophages. MTX attenuated LPS-induced MAPK and NF-κB activation, and toll/IL-1R domain-containing adaptor inducing IFN-beta (TRIF1)-dependent signalling. Conversely, MTX increased the expression of the NF-κB suppressor A20 (TNFAIP3), itself a RA-susceptibility gene. Mechanistically, MTX-induced macrophage tolerance was dependent on A20, as siRNA-mediated knockdown of A20 reversed the MTX-induced reduction of IL-6 expression. In vivo, TNFAIP3 expression was significantly higher in peripheral blood cells of MTX-responsive individuals from a cohort of patients with arthritis under MTX monotherapy, whereas MTX-treated mice exhibited reduced inflammatory responses to LPS. CONCLUSIONS: MTX impairs macrophage proinflammatory responses through upregulation of A20 expression. The A20-mediated MTX-induced innate tolerance might limit inflammation in the RA synovial context, and positions A20 as a potential MTX-response biomarker.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Granulocyte-Macrophage Colony-Stimulating Factor/drug effects , Methotrexate/pharmacology , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Animals , Arthritis, Rheumatoid/metabolism , Humans , Inflammation/metabolism , Mice , Signal Transduction/drug effects , Synovial Fluid/metabolism , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
J Immunol ; 196(3): 1327-37, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26729812

ABSTRACT

Human CD14(++)CD16(-) and CD14(+/lo)CD16(+) monocyte subsets comprise 85 and 15% of blood monocytes, respectively, and are thought to represent distinct stages in the monocyte differentiation pathway. However, the differentiation fates of both monocyte subsets along the macrophage (Mϕ) lineage have not yet been elucidated. We have now evaluated the potential of CD14(++) CD16(-) and CD16(+) monocytes to differentiate and to be primed toward pro- or anti-inflammatory Mϕs upon culture with GM-CSF or M-CSF, respectively (subsequently referred to as GM14, M14, GM16, or M16). Whereas GM16 and GM14 were phenotypic and functionally analogous, M16 displayed a more proinflammatory profile than did M14. Transcriptomic analyses evidenced that genes associated with M-CSF-driven Mϕ differentiation (including FOLR2, IL10, IGF1, and SERPINB2) are underrepresented in M16 with respect to M14. The preferential proinflammatory skewing of M16 relative to M14 was found to be mediated by the secretion of activin A and the low levels of IL-10 produced by M16. In fact, activin A receptor blockade during the M-CSF-driven differentiation of CD16(+) monocytes, or addition of IL-10-containing M14-conditioned medium, significantly enhanced their expression of anti-inflammatory-associated molecules while impairing their acquisition of proinflammatory-related markers. Thus, we propose that M-CSF drives CD14(++)CD16- monocyte differentiation into bona fide anti-inflammatory Mϕs in a self-autonomous manner, whereas M-CSF-treated CD16(+) monocytes generate Mϕs with a skewed proinflammatory profile by virtue of their high activin A expression unless additional anti-inflammatory stimuli such as IL-10 are provided.


Subject(s)
Activins/biosynthesis , Cell Differentiation/immunology , Interleukin-10/biosynthesis , Macrophages/cytology , Monocytes/immunology , Activins/immunology , Blotting, Western , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique , Humans , Inflammation/immunology , Interleukin-10/immunology , Macrophages/immunology , Monocytes/cytology , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Receptors, IgG/immunology
8.
J Immunol ; 192(8): 3858-67, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24639350

ABSTRACT

The CCL2 chemokine mediates monocyte egress from bone marrow and recruitment into inflamed tissues through interaction with the CCR2 chemokine receptor, and its expression is upregulated by proinflammatory cytokines. Analysis of the gene expression profile in GM-CSF- and M-CSF-polarized macrophages revealed that a high CCL2 expression characterizes macrophages generated under the influence of M-CSF, whereas CCR2 is expressed only by GM-CSF-polarized macrophages. Analysis of the factors responsible for this differential expression identified activin A as a critical factor controlling the expression of the CCL2/CCR2 pair in macrophages, as activin A increased CCR2 expression but inhibited the acquisition of CCL2 expression by M-CSF-polarized macrophages. CCL2 and CCR2 were found to determine the extent of macrophage polarization because CCL2 enhances the LPS-induced production of IL-10, whereas CCL2 blockade leads to enhanced expression of M1 polarization-associated genes and cytokines, and diminished expression of M2-associated markers in human macrophages. Along the same line, Ccr2-deficient bone marrow-derived murine macrophages displayed an M1-skewed polarization profile at the transcriptomic level and exhibited a significantly higher expression of proinflammatory cytokines (TNF-α, IL-6) in response to LPS. Therefore, the CCL2-CCR2 axis regulates macrophage polarization by influencing the expression of functionally relevant and polarization-associated genes and downmodulating proinflammatory cytokine production.


Subject(s)
Chemokine CCL2/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Activins/pharmacology , Animals , Chemokine CCL2/metabolism , Chemokine CCL8/genetics , Chemokine CCL8/metabolism , Cluster Analysis , Gene Expression Regulation/drug effects , Humans , Lipopolysaccharides/immunology , Macrophages/immunology , Mice , Mice, Knockout , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Transcriptome
9.
J Immunol ; 193(10): 5181-9, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25326025

ABSTRACT

Intravenous Igs (IVIg) therapy is widely used as an immunomodulatory strategy in inflammatory pathologies and is suggested to promote cancer regression. Because progression of tumors depends on their ability to redirect the polarization state of tumor-associated macrophages (from M1/immunogenic/proinflammatory to M2/anti-inflammatory), we have evaluated whether IVIg limits tumor progression and dissemination through modulation of macrophage polarization. In vitro, IVIg inhibited proinflammatory cytokine production from M1 macrophages and induced a M2-to-M1 polarization switch on human and murine M2 macrophages. In vivo, IVIg modified the polarization of tumor-associated myeloid cells in a Fcεr1γ chain-dependent manner, modulated cytokine blood levels in tumor-bearing animals, and impaired tumor progression via FcγRIII (CD16), FcγRIV, and FcRγ engagement, the latter two effects being macrophage mediated. Therefore, IVIg immunomodulatory activity is dependent on the polarization state of the responding macrophages, and its ability to trigger a M2-to-M1 macrophage polarization switch might be therapeutically useful in cancer, in which proinflammatory or immunogenic functions should be promoted.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Immunoglobulins, Intravenous/pharmacology , Immunologic Factors/pharmacology , Lung Neoplasms/drug therapy , Macrophages/drug effects , Melanoma, Experimental/drug therapy , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Humans , Lung/drug effects , Lung/immunology , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Macrophages/classification , Macrophages/immunology , Macrophages/pathology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Neoplasm Transplantation , Receptors, IgE/genetics , Receptors, IgE/immunology , Receptors, IgG/genetics , Receptors, IgG/immunology , Signal Transduction , Tumor Burden/drug effects
10.
Blood ; 121(15): e108-17, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23430108

ABSTRACT

Dendritic cells (DCs) promote tolerance or immunity depending on their maturation state, which is enhanced or accelerated upon MEK-ERK signaling pathway inhibition. We have determined the contribution of MEK-ERK activation to the profile of gene expression of human immature monocyte-derived dendritic cells (MDDCs) and peripheral blood myeloid DCs. ERK inhibition altered the expression of genes that mediate Chemokine (C-C motif) ligand 19 (CCL19)-directed migration (CCR7) and low-density lipoprotein (LDL) binding (CD36, SCARB1, OLR1, CXCL16) by immature DCs. In addition, ERK upregulated CCL2 expression while impairing the expression of DC maturation markers (RUNX3, ITGB7, IDO1). MEK-ERK-regulated genes exhibited an overrepresentation of cognate sequences for the aryl hydrocarbon receptor (AhR) transcription factor, whose transcriptional and DNA-binding activities increased in MDDCs upon exposure to the MEK1/2 inhibitor U0126. Therefore, the MEK-ERK signaling pathway regulates antigen capture, lymph node homing, and acquisition of maturation-associated genes, and its contribution to the maintenance of the immature state of MDDCs and myeloid DCs is partly dependent on the activity of AhR. Since pharmacologic modulation of the MEK-ERK signaling pathway has been proposed as a potential therapeutic strategy for cancer, our findings indicate that ERK inhibitors might influence antitumor responses through regulation of critical DC effector functions.


Subject(s)
Dendritic Cells/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Blotting, Western , Butadienes/pharmacology , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Dendritic Cells/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression/drug effects , Gene Expression Profiling , Hep G2 Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Monocytes/drug effects , Monocytes/metabolism , Nitriles/pharmacology , Oligonucleotide Array Sequence Analysis , Polychlorinated Dibenzodioxins/pharmacology , Receptors, Aryl Hydrocarbon/genetics , Receptors, CCR7/genetics , Receptors, CCR7/metabolism
11.
J Immunol ; 190(5): 2301-10, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23355731

ABSTRACT

Besides its role as a neurotransmitter, serotonin (5-hydroxytryptamine, 5HT) regulates inflammation and tissue repair via a set of receptors (5HT(1-7)) whose pattern of expression varies among cell lineages. Considering the importance of macrophage polarization plasticity for inflammatory responses and tissue repair, we evaluated whether 5HT modulates human macrophage polarization. 5HT inhibited the LPS-induced release of proinflammatory cytokines without affecting IL-10 production, upregulated the expression of M2 polarization-associated genes (SERPINB2, THBS1, STAB1, COL23A1), and reduced the expression of M1-associated genes (INHBA, CCR2, MMP12, SERPINE1, CD1B, ALDH1A2). Whereas only 5HT(7) mediated the inhibitory action of 5HT on the release of proinflammatory cytokines, both 5HT(2B) and 5HT(7) receptors mediated the pro-M2 skewing effect of 5HT. In fact, blockade of both receptors during in vitro monocyte-to-macrophage differentiation preferentially modulated the acquisition of M2 polarization markers. 5HT(2B) was found to be preferentially expressed by anti-inflammatory M2(M-CSF) macrophages and was detected in vivo in liver Kupffer cells and in tumor-associated macrophages. Therefore, 5HT modulates macrophage polarization and contributes to the maintenance of an anti-inflammatory state via 5HT(2B) and 5HT(7), whose identification as functionally relevant markers for anti-inflammatory/homeostatic human M2 macrophages suggests their potential therapeutic value in inflammatory pathologies.


Subject(s)
Biomarkers/metabolism , Cell Differentiation/drug effects , Macrophages/drug effects , Receptor, Serotonin, 5-HT2B/immunology , Receptors, Serotonin/immunology , Serotonin/pharmacology , Animals , Cell Lineage , Cells, Cultured , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Genes, Reporter , Humans , Inflammation/chemically induced , Inflammation/immunology , Inflammation/metabolism , Interleukin-10/biosynthesis , Interleukin-10/immunology , Kupffer Cells/cytology , Kupffer Cells/drug effects , Kupffer Cells/immunology , Lipopolysaccharides , Luciferases , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Receptor, Serotonin, 5-HT2B/genetics , Receptors, Serotonin/genetics , Serotonin/immunology , Signal Transduction/drug effects
14.
J Immunol ; 186(4): 2192-200, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21239715

ABSTRACT

Dendritic cell-specific ICAM-3-grabbing nonintegrin (DC-SIGN; CD209) is a human pathogen-attachment C-type lectin with no obvious murine ortholog and for which ligation leads to enhanced anti-inflammatory cytokine release and altered proinflammatory cytokine production. Although induced by IL-4 in monocytes and considered as a DC marker, DC-SIGN expression on human APCs under homeostatic conditions is so far unexplained. We report in this study that M-CSF enhances DC-SIGN expression on in vitro derived anti-inflammatory macrophages and that M-CSF mediates the induction of DC-SIGN by fibroblast- and tumor cell-conditioned media. The M-CSF-inducible DC-SIGN expression along monocyte-to-macrophage differentiation is dependent on JNK and STAT3 activation, potentiated by STAT3-activating cytokines (IL-6, IL-10), and abrogated by the M1-polarizing cytokine GM-CSF. In pathological settings, DC-SIGN expression is detected in tumor tissues and on ex vivo-isolated CD14(+) CD163(+) IL-10-producing tumor-associated macrophages. Importantly, DC-SIGN Abs reduced the release of IL-10 from macrophages exposed to Lewis(x)-expressing SKBR3 tumor cells. These results indicate that DC-SIGN is expressed on both wound-healing (IL-4-dependent) and regulatory (M-CSF-dependent) alternative (M2) macrophages and that DC-SIGN expression on tumor-associated macrophages might help tumor progression by contributing to the maintenance of an immunosuppressive environment.


Subject(s)
Adenocarcinoma/immunology , Breast Neoplasms/immunology , Cell Adhesion Molecules/biosynthesis , Cell Polarity/immunology , Dendritic Cells/immunology , Interleukin-10/physiology , Interleukin-6/physiology , Lectins, C-Type/biosynthesis , Macrophage Colony-Stimulating Factor/physiology , Melanoma, Experimental/immunology , Receptors, Cell Surface/biosynthesis , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion Molecules/genetics , Cell Differentiation/immunology , Cells, Cultured , Culture Media, Conditioned/pharmacology , Dendritic Cells/metabolism , Disease Progression , Gene Expression Regulation/immunology , Humans , Immunocompromised Host/genetics , Immunocompromised Host/immunology , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Lectins, C-Type/genetics , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Neoplasm Proteins/physiology , Receptors, Cell Surface/genetics , Tumor Cells, Cultured
15.
J Innate Immun ; 15(1): 517-530, 2023.
Article in English | MEDLINE | ID: mdl-37040733

ABSTRACT

Toll-like receptor 7 (TLR7) is an endosomal pathogen-associated molecular pattern (PAMP) receptor that senses single-stranded RNA (ssRNA) and whose engagement results in the production of type I IFN and pro-inflammatory cytokines upon viral exposure. Recent genetic studies have established that a dysfunctional TLR7-initiated signaling is directly linked to the development of inflammatory responses. We present evidence that TLR7 is preferentially expressed by monocyte-derived macrophages generated in the presence of M-CSF (M-MØ). We now show that TLR7 activation in M-MØ triggers a weak MAPK, NFκB, and STAT1 activation and results in low production of type I IFN. Of note, TLR7 engagement reprograms MAFB+ M-MØ towards a pro-inflammatory transcriptional profile characterized by the expression of neutrophil-attracting chemokines (CXCL1-3, CXCL5, CXCL8), whose expression is dependent on the transcription factors MAFB and AhR. Moreover, TLR7-activated M-MØ display enhanced pro-inflammatory responses and a stronger production of neutrophil-attracting chemokines upon secondary stimulation. As aberrant TLR7 signaling and enhanced pulmonary neutrophil/lymphocyte ratio associate with impaired resolution of virus-induced inflammatory responses, these results suggest that targeting macrophage TLR7 might be a therapeutic strategy for viral infections where monocyte-derived macrophages exhibit a pathogenic role.


Subject(s)
Monocytes , Toll-Like Receptor 7 , Humans , Toll-Like Receptor 7/metabolism , Monocytes/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Neutrophil Infiltration , Cytokines/metabolism , Macrophages/metabolism , Chemokines/metabolism
16.
JCI Insight ; 8(24)2023 Dec 22.
Article in English | MEDLINE | ID: mdl-37917179

ABSTRACT

Monocyte-derived macrophages, the major source of pathogenic macrophages in COVID-19, are oppositely instructed by macrophage CSF (M-CSF) or granulocyte macrophage CSF (GM-CSF), which promote the generation of antiinflammatory/immunosuppressive MAFB+ (M-MØ) or proinflammatory macrophages (GM-MØ), respectively. The transcriptional profile of prevailing macrophage subsets in severe COVID-19 led us to hypothesize that MAFB shapes the transcriptome of pulmonary macrophages driving severe COVID-19 pathogenesis. We have now assessed the role of MAFB in the response of monocyte-derived macrophages to SARS-CoV-2 through genetic and pharmacological approaches, and we demonstrate that MAFB regulated the expression of the genes that define pulmonary pathogenic macrophages in severe COVID-19. Indeed, SARS-CoV-2 potentiated the expression of MAFB and MAFB-regulated genes in M-MØ and GM-MØ, where MAFB upregulated the expression of profibrotic and neutrophil-attracting factors. Thus, MAFB determines the transcriptome and functions of the monocyte-derived macrophage subsets that underlie pulmonary pathogenesis in severe COVID-19 and controls the expression of potentially useful biomarkers for COVID-19 severity.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/metabolism , COVID-19/metabolism , Macrophages/metabolism , Macrophages, Alveolar/metabolism , Biomarkers/metabolism , MafB Transcription Factor/genetics , MafB Transcription Factor/metabolism
17.
J Innate Immun ; : 1-14, 2022 Nov 14.
Article in English | MEDLINE | ID: mdl-36380627

ABSTRACT

Methotrexate (MTX) is an antifolate drug used as a chemotherapeutic agent for acute lymphoblastic leukemia, where MTX improves patients' prognosis. Macrophage reprogramming is being increasingly assessed as an antitumor therapeutic strategy. However, and although MTX limits the pathogenic action of macrophages in chronic inflammatory diseases, its effects on tumor-promoting macrophages have not been previously explored. We now report that MTX shapes the transcriptional and functional profile of M-CSF-dependent human macrophages, whose transcriptome is highly enriched in the gene signature that defines pathogenic tumor-associated macrophages ("large TAM"). Specifically, MTX prompted the acquisition of the gene signature of antitumoral "small TAM" and skewed macrophages toward an IL-6high IFNß1high IL-10low phenotype upon subsequent stimulation. Mechanistically, the MTX-induced macrophage reprogramming effect correlated with a reduction of the M-CSF receptor CSF1R expression and function, as well as a diminished expression of MAF and MAFB transcription factors, primary determinants of pro-tumoral macrophages whose transcriptional activity is dependent on GSK3ß. Indeed, the ability of MTX to transcriptionally reprogram macrophages toward an antitumoral phenotype was abrogated by inhibition of GSK3ß. Globally, our results establish MTX as a macrophage reprogramming drug and indicate that its ability to modulate macrophage polarization may also underlie its therapeutic benefits. Since GSK3ß inhibition abrogates the reprogramming action of MTX, our results suggest that the GSK3ß-MAFB/MAF axis constitutes a target for the macrophage-centered antitumor strategies.

18.
J Reprod Immunol ; 154: 103735, 2022 12.
Article in English | MEDLINE | ID: mdl-36063657

ABSTRACT

Recurrent pregnancy loss (RPL) and recurrent implantation failure (RIF) are two well-defined clinical entities, but the role of the monocytes in their pathophysiology needs to be clarified. This study aimed to evaluate the role of the three monocyte subsets (classical, intermediate, and non-classical) and relevant cytokines/chemokines in a cohort of RPL and RIF women to better characterize a baseline proinflammatory profile that could define inflammatory pathophysiology in these two different conditions. We evaluated 108 non-pregnant women: 53 RPL, 24 RIF, and 31 fertile healthy controls (HC). Multiparametric flow cytometry was used to quantify the frequency of surface chemokine receptors (CCR2, CCR5, and CX3CR1) on the monocyte subsets. Cytokines were assessed in plasma samples using a multiplex assay. The CX3CR1+ and CCR5+ intermediate monocytes were significantly higher in RPL and RIF compared to HC. A significant positive correlation was observed between CX3CR1+ intermediate monocytes and IL-17A (P = .03, r = 0.43). The Boruta algorithm followed by a multivariate logistic regression model was used to select the most relevant variables that could help define RPL and RIF: in RPL were CX3CR1 non-classical monocytes, TGF-ß1, and CCR5 intermediate monocytes; in RIF: CCR5 intermediate monocytes and TGF-ß3. The combination of these variables could predict RPL and RIF with 90 % and 82 %, respectively. Our study suggests that a combination of specific blood monocyte subsets and cytokines could aid in identifying RPL and RIF women with a pro-inflammatory profile. These findings could provide a more integrated understanding of these pathologies. Further investigation and validation in independent cohorts are warranted.


Subject(s)
Abortion, Habitual , Monocytes , Pregnancy , Humans , Female , Immunophenotyping , Flow Cytometry , Cytokines
19.
J Innate Immun ; 14(3): 243-256, 2022.
Article in English | MEDLINE | ID: mdl-34670213

ABSTRACT

During inflammatory responses, monocytes are recruited into inflamed tissues, where they become monocyte-derived macrophages and acquire pro-inflammatory and tissue-damaging effects in response to the surrounding environment. In fact, monocyte-derived macrophage subsets are major pathogenic cells in inflammatory pathologies. Strikingly, the transcriptome of pathogenic monocyte-derived macrophage subsets resembles the gene profile of macrophage colony-stimulating factor (M-CSF)-primed monocyte-derived human macrophages (M-MØ). As M-MØ display a characteristic cytokine profile after activation (IL10high TNFlow IL23low IL6low), we sought to determine the transcriptional signature of M-MØ upon exposure to pathogenic stimuli. Activation of M-MØ led to the acquisition of a distinctive transcriptional profile characterized by the induction of a group of genes (Gene set 1) highly expressed by pathogenic monocyte-derived macrophages in COVID-19 and whose presence in tumor-associated macrophages (TAM) correlates with the expression of macrophage-specific markers (CD163, SPI1) and IL10. Indeed, Gene set 1 expression was primarily dependent on ERK/p38 and STAT3 activation, and transcriptional analysis and neutralization experiments revealed that IL-10 is not only required for the expression of a subset of genes within Gene set 1 but also significantly contributes to the idiosyncratic gene signature of activated M-MØ. Our results indicate that activation of M-CSF-dependent monocyte-derived macrophages induces a distinctive gene expression profile, which is partially dependent on IL-10, and identifies a gene set potentially helpful for macrophage-centered therapeutic strategies.


Subject(s)
COVID-19 , Macrophage Colony-Stimulating Factor , Cell Differentiation , Cells, Cultured , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Monocytes/metabolism
20.
Hepatology ; 49(1): 287-96, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19111020

ABSTRACT

UNLABELLED: Human LSECtin (liver and lymph node sinusoidal endothelial cell C-type lectin, CLEC4G) is a C-type lectin encoded within the L-SIGN/DC-SIGN/CD23 gene cluster. LSECtin acts as a pathogen attachment factor for Ebolavirus and the SARS coronavirus, and its expression can be induced by interleukin-4 on monocytes and macrophages. Although reported as a liver and lymph node sinusoidal endothelial cell-specific molecule, LSECtin could be detected in the MUTZ-3 dendritic-like cell line at the messenger RNA (mRNA) and protein level, and immunohistochemistry analysis on human liver revealed its presence in Kupffer cells coexpressing the myeloid marker CD68. The expression of LSECtin in myeloid cells was further corroborated through the analysis of the proximal regulatory region of the human LSECtin gene, whose activity was maximal in LSECtin+ myeloid cells, and which contains a highly conserved PU.1-binding site. PU.1 transactivated the LSECtin regulatory region in collaboration with hematopoietic-restricted transcription factors (Myb, RUNX3), and was found to bind constitutively to the LSECtin proximal promoter. Moreover, knockdown of PU.1 through the use of small interfering RNA led to a decrease in LSECtin mRNA levels in THP-1 and monocyte-derived dendritic cells, thus confirming the involvement of PU.1 in the myeloid expression of the lectin. CONCLUSION: LSECtin is expressed by liver myeloid cells, and its expression is dependent on the PU.1 transcription factor.


Subject(s)
Kupffer Cells/metabolism , Lectins, C-Type/biosynthesis , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Animals , Humans , Liver/cytology , Mice , Myeloid Cells/metabolism , NIH 3T3 Cells , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL