Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 264
Filter
1.
Nat Immunol ; 14(1): 41-51, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23179077

ABSTRACT

Coordinated navigation within tissues is essential for cells of the innate immune system to reach the sites of inflammatory processes, but the signals involved are incompletely understood. Here we demonstrate that NG2(+) pericytes controlled the pattern and efficacy of the interstitial migration of leukocytes in vivo. In response to inflammatory mediators, pericytes upregulated expression of the adhesion molecule ICAM-1 and released the chemoattractant MIF. Arteriolar and capillary pericytes attracted and interacted with myeloid leukocytes after extravasating from postcapillary venules, 'instructing' them with pattern-recognition and motility programs. Inhibition of MIF neutralized the migratory cues provided to myeloid leukocytes by NG2(+) pericytes. Hence, our results identify a previously unknown role for NG2(+) pericytes as an active component of innate immune responses, which supports the immunosurveillance and effector function of extravasated neutrophils and macrophages.


Subject(s)
Intercellular Adhesion Molecule-1/metabolism , Intramolecular Oxidoreductases/metabolism , Leukocytes/immunology , Macrophage Migration-Inhibitory Factors/metabolism , Pericytes/immunology , Receptors, Pattern Recognition/immunology , Antibodies, Blocking/pharmacology , Arterioles/immunology , Capillaries/immunology , Cell Communication/drug effects , Cell Movement/drug effects , Cell Movement/immunology , Cells, Cultured , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Intercellular Adhesion Molecule-1/genetics , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Leukocytes/drug effects , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/immunology , Neutrophil Activation/drug effects , Up-Regulation/drug effects , Venules/immunology
2.
Nat Immunol ; 14(6): 554-63, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23624557

ABSTRACT

Microenvironment-based alterations in phenotypes of mast cells influence the susceptibility to anaphylaxis, yet the mechanisms underlying proper maturation of mast cells toward an anaphylaxis-sensitive phenotype are incompletely understood. Here we report that PLA2G3, a mammalian homolog of anaphylactic bee venom phospholipase A2, regulates this process. PLA2G3 secreted from mast cells is coupled with fibroblastic lipocalin-type PGD2 synthase (L-PGDS) to provide PGD2, which facilitates mast-cell maturation via PGD2 receptor DP1. Mice lacking PLA2G3, L-PGDS or DP1, mast cell-deficient mice reconstituted with PLA2G3-null or DP1-null mast cells, or mast cells cultured with L-PGDS-ablated fibroblasts exhibited impaired maturation and anaphylaxis of mast cells. Thus, we describe a lipid-driven PLA2G3-L-PGDS-DP1 loop that drives mast cell maturation.


Subject(s)
Group III Phospholipases A2/immunology , Mast Cells/immunology , Paracrine Communication/immunology , Prostaglandin D2/immunology , Receptors, Prostaglandin/immunology , Animals , Blotting, Western , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Gene Expression Profiling , Group III Phospholipases A2/genetics , Group III Phospholipases A2/metabolism , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Intramolecular Oxidoreductases/metabolism , Lipocalins/genetics , Lipocalins/immunology , Lipocalins/metabolism , Mast Cells/metabolism , Mast Cells/ultrastructure , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Oligonucleotide Array Sequence Analysis , Paracrine Communication/genetics , Prostaglandin D2/metabolism , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Reverse Transcriptase Polymerase Chain Reaction
3.
Trends Immunol ; 40(5): 447-462, 2019 05.
Article in English | MEDLINE | ID: mdl-30962001

ABSTRACT

Produced by many cell types, macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine with critical and supporting roles in many disease states and conditions. Its disease associations, myriad functions, receptors, and downstream signaling have been the subject of considerable research, yet many questions remain. Moreover, the relevance of MIF's partially functionally redundant family member, D-dopachrome tautomerase (D-DT), also remains to be further characterized. Here, we discuss recent discoveries demonstrating direct roles of MIF in supporting NLR Family Pyrin Domain-Containing 3 (NRLP3) inflammasome activation, as well as acting as a molecular chaperone for intracellular proteins. These findings may offer new clues to understanding MIF's multiple functions, and assist the development of putative MIF-targeting therapeutics for a variety of pathologies.


Subject(s)
Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Humans , Inflammasomes/immunology , Intramolecular Oxidoreductases/biosynthesis , Macrophage Migration-Inhibitory Factors/biosynthesis , NLR Family, Pyrin Domain-Containing 3 Protein/immunology
4.
J Biol Chem ; 295(3): 850-867, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31811089

ABSTRACT

Human macrophage migration-inhibitory factor (MIF) is an evolutionarily-conserved protein that has both extracellular immune-modulating and intracellular cell-regulatory functions. MIF plays a role in various diseases, including inflammatory diseases, atherosclerosis, autoimmunity, and cancer. It serves as an inflammatory cytokine and chemokine, but also exhibits enzymatic activity. Secreted MIF binds to cell-surface immune receptors such as CD74 and CXCR4. Plants possess MIF orthologs but lack the associated receptors, suggesting functional diversification across kingdoms. Here, we characterized three MIF orthologs (termed MIF/d-dopachrome tautomerase-like proteins or MDLs) of the model plant Arabidopsis thaliana Recombinant Arabidopsis MDLs (AtMDLs) share similar secondary structure characteristics with human MIF, yet only have minimal residual tautomerase activity using either p-hydroxyphenylpyruvate or dopachrome methyl ester as substrate. Site-specific mutagenesis suggests that this is due to a distinct amino acid difference at the catalytic cavity-defining residue Asn-98. Surprisingly, AtMDLs bind to the human MIF receptors CD74 and CXCR4. Moreover, they activate CXCR4-dependent signaling in a receptor-specific yeast reporter system and in CXCR4-expressing human HEK293 transfectants. Notably, plant MDLs exert dose-dependent chemotactic activity toward human monocytes and T cells. A small molecule MIF inhibitor and an allosteric CXCR4 inhibitor counteract this function, revealing its specificity. Our results indicate cross-kingdom conservation of the receptor signaling and leukocyte recruitment capacities of human MIF by its plant orthologs. This may point toward a previously unrecognized interplay between plant proteins and the human innate immune system.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/genetics , Histocompatibility Antigens Class II/genetics , Immunity, Innate/genetics , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Receptors, CXCR4/genetics , Antigens, Differentiation, B-Lymphocyte/chemistry , Arabidopsis/genetics , Arabidopsis/immunology , Chemotaxis/genetics , Chemotaxis/immunology , Conserved Sequence/genetics , Conserved Sequence/immunology , Cytokines/genetics , Cytokines/immunology , HEK293 Cells , Histocompatibility Antigens Class II/chemistry , Humans , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/chemistry , Macrophage Migration-Inhibitory Factors/immunology , Monocytes/chemistry , Monocytes/metabolism , Protein Binding/genetics , Receptors, CXCR4/chemistry , Sequence Homology , T-Lymphocytes/chemistry , T-Lymphocytes/metabolism
5.
Cell Immunol ; 370: 104439, 2021 12.
Article in English | MEDLINE | ID: mdl-34607646

ABSTRACT

Macrophage migration inhibitory factor (MIF-1) and its homologue d-dopachrome tautomerase (MIF-2) share the common CD74 receptor and function innately to enhance severity of multiple sclerosis (MS) as well as the experimental autoimmune encephalomyelitis (EAE) model for MS. We previously demonstrated that genetically high-MIF-expressing male subjects with relapsing MS had a significantly greater risk of conversion to progressive MS (PMS) than lower-MIF-expressing males. To expand on this observation, we utilized MIF-1, MIF-2, and MIF-1/2-DUAL-deficient male mice to discern if there would be a greater contribution of these inflammatory factors in EAE mice with severe vs. moderate clinical disease signs. As shown previously, mice deficient in either MIF-1 or MIF-2 each had a ∼25% reduction of moderate EAE compared to WT mice, with significant differences in disease onset and trajectory. However, EAE induction in mice deficient in both MIF-1 and MIF-2 genes did not result in a further reduction in EAE severity. This result suggests that the two MIF homologues were likely affecting the same pathogenic pathways such that each could partially compensate for the other but not in an additive or synergistic manner. However, MIF-1-KO, MIF-2-KO, and MIF-1/2-DUAL-KO mice with severe EAE did not exhibit a significant reduction in cumulative EAE scores compared with WT mice, but the MIF-1-KO and, to a lesser extent, MIF-1/2-DUAL-KO mice did show a significant reduction in daily EAE scores over the last 3 days of observation, and MIF-2-KO mice showed a more modest but still consistent reduction over the same span. Furthermore, deletion of MIF-1 resulted in a massive reduction in the expression of EAE- and Complete Freund's Adjuvant-associated inflammatory factors, suggesting delayed involvement of the MIF/CD74 axis in promoting disease expression. To further explore modulation of MIF-1 and MIF-2 effects on EAE, we treated WT mice with moderate EAE using DRα1-mMOG-35-55, an inhibitor of CD74 that blocks both MIF-1 and MIF-2 action. This treatment reduced ongoing moderate EAE severity in excess of 25%, suggesting efficient blockade of the MIF/CD74 axis in disease-enhancing pathways. Moreover, DRα1-mMOG-35-55 treatment of mice with severe EAE strongly reversed EAE- and CFA-associated expression of inflammatory cytokines and chemokines including Tnf, Ccr7, Ccr6, Ccl8, Cxcr3, and Ccl19 in MIF-deficient mouse genotypes, and also exceeded innate MIF-1 and MIF-2 EAE enhancing effects, especially in MIF-1-KO mice. These results illustrate the therapeutic potential of targeting the disease-enhancing MIF/CD74 pathway in male mice with moderate and severe EAE, with implications for treatment of high-MIF-expressing RRMS human males at risk of conversion to progressive MS as well as those that have already transitioned to PMS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Neuroprotective Agents/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Br J Haematol ; 191(5): 784-795, 2020 12.
Article in English | MEDLINE | ID: mdl-32558939

ABSTRACT

An increase in immunosuppressive myeloid-derived suppressor cells (MDSCs) is associated with disease progression and treatment resistance in multiple myeloma (MM). We investigated the mechanisms underlying MDSC induction, and sought to discover a strategy for prevention of MDSC induction in MM. Using a transwell co-culture system, four of nine examined human myeloma-derived cell lines (HMCLs) were potent in inducing monocytic (M)-MDSCs from normal peripheral blood mononuclear cells (PBMCs). As the results, we identified that secretion of C-C motif chemokine ligand 5 (CCL5) and macrophage migration inhibitory factor (MIF) by myeloma cells is a prerequisite for induction of MDSCs in MM. The immunomodulatory drug (IMiD) compounds, such as lenalidomide (LEN) and pomalidomide (POM), were identified as potent inhibitors of MDSC induction through bidirectional molecular effects of cereblon (CRBN)-dependent and -independent downregulation of CCL5 and MIF in myeloma cells; and downregulation of C-C motif chemokine receptor 5, a receptor for CCL5, and induction of interferon regulatory factor 8, a critical transcription factor for monocytic differentiation, in PBMCs. In the present study of the molecular mechanisms underlying MDSC induction, we identified a novel effect of LEN and POM of inhibiting MDSC induction via overlapping regulatory effects in myeloma cells and normal PBMCs.


Subject(s)
Lenalidomide/pharmacology , Multiple Myeloma/immunology , Myeloid-Derived Suppressor Cells/immunology , Thalidomide/analogs & derivatives , Cell Line, Tumor , Chemokine CCL5/immunology , Coculture Techniques , Humans , Interferon Regulatory Factors/immunology , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Myeloid-Derived Suppressor Cells/pathology , Neoplasm Proteins/immunology , Thalidomide/pharmacology
7.
Cytokine ; 133: 155121, 2020 09.
Article in English | MEDLINE | ID: mdl-32417648

ABSTRACT

Human macrophage migration inhibition factor (MIF) is a protein with cytokine and chemokine properties that regulates a diverse range of physiological functions related to innate immunity and inflammation. Most research has focused on the role of MIF in different inflammatory diseases. D-dopachrome tautomerase (DDT), a different molecule with structural similarities to MIF, which shares receptors and biological functions, has recently been reported, but little is known about its roles and mechanisms. In this review, we sought to understand the similarities and differences between these molecules by summarizing what is known about their different structures, receptors and mechanisms regulating their expression and biological activities with an emphasis on immunological aspects.


Subject(s)
Immunologic Factors/immunology , Immunomodulation/immunology , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Animals , Humans
8.
Cytokine ; 136: 155283, 2020 12.
Article in English | MEDLINE | ID: mdl-32947151

ABSTRACT

Toxoplasma gondii (T. gondii) is an intracellular parasite responsible for causing toxoplasmosis. When infection occurs during pregnancy, it can produce severe congenital infection with ocular and neurologic damage to the infant. From the oral infection parasite reaches the intestine, causing inflammatory response, damage in tissue architecture and systemic dissemination. Macrophage migration inhibition factor (MIF) is a cytokine secreted from both immune and non-immune cells, including gut epithelial cells. MIF is described to promote inflammatory responses, to be associated in colitis pathogenesis and also to play role in maintaining the intestinal barrier. The aim of the present study was to evaluate the influence of the pregnancy and MIF deficiency on T. gondii infection in the intestinal microenvironment and to address how these factors can impact on the intestinal architecture and local cytokine profile. For this purpose, small intestine of pregnant and non-pregnant C57BL/6 MIF deficient mice (MIF-/-) and Wild-type (WT) orally infected with 5 cysts of ME-49 strain of T. gondii were collected on day 8th of infection. Intestines were processed for morphological and morphometric analyses, parasite quantification and for cytokines mensuration. Our results showed that the absence of MIF and pregnancy caused an increase in T. gondii infection index. T. gondii immunolocalization demonstrated that segments preferentially infected with T. gondii were duodenum and ileum. The infection caused a reduction in the size of the intestinal villi, whereas, infection associated with pregnancy caused an increase in villi size due to edema caused by the infection. Also, the goblet cell number was increased in the ileum of MIF-/- mice, when compared to the corresponding WT group. Analyses of cytokine production in the small intestine showed that MIF was up regulated in the gut of pregnant WT mice due to infection. Also, infection provoked an intense Th1 response that was more exacerbated in pregnant MIF-/- mice. We also detected that the Th2/Treg response was more pronounced in MIF-/- mice. Altogether, our results demonstrated that pregnancy and MIF deficiency interferes in the balance of the intestinal cytokines and favors a Th1-immflamatory profile, which in turn, impact in the development of pathology caused by T. gondii infection in the intestinal microenvironment.


Subject(s)
Duodenum/immunology , Ileum/immunology , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Pregnancy Complications, Parasitic/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Animals , Female , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Mice , Mice, Knockout , Pregnancy , Pregnancy Complications, Parasitic/genetics , Toxoplasmosis/genetics
9.
Immunity ; 34(4): 514-26, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21497116

ABSTRACT

Particulates such as silica crystal (silica) and aluminum salts (alum) activate the inflammasome and induce the secretion of proinflammatory cytokines in macrophages. These particulates also induce the production of immunoglobulin E via a T helper 2 (Th2) cell-associated mechanism. However, the mechanism involved in the induction of type 2 immunity has not been elucidated. Here, we showed that silica and alum induced lipopolysaccharide-primed macrophages to produce the lipid mediator prostaglandin E2 (PGE2) and interleukin-1ß (IL-1ß). Macrophages deficient in the inflammasome components caspase 1, NALP3, and ASC revealed that PGE2 production was independent of the NALP3 inflammasome. PGE2 expression was markedly reduced in PGE synthase-deficient (Ptges⁻/⁻) macrophages, and Ptges⁻/⁻ mice displayed reduced antigen-specific serum IgE concentrations after immunization with alum or silica. Our results indicate that silica and alum regulate the production of PGE2 and that the induction of PGE2 by particulates controls the immune response in vivo.


Subject(s)
Aluminum/pharmacology , Carrier Proteins/immunology , Inflammasomes/immunology , Macrophages/drug effects , Macrophages/immunology , Prostaglandins/biosynthesis , Silicon Dioxide/pharmacology , Animals , Caspase 1/metabolism , Cells, Cultured , Crystallization , Intramolecular Oxidoreductases/deficiency , Intramolecular Oxidoreductases/immunology , Macrophages/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Phagosomes/immunology , Prostaglandin-E Synthases , Prostaglandins/immunology
10.
Molecules ; 25(2)2020 Jan 10.
Article in English | MEDLINE | ID: mdl-31936865

ABSTRACT

Recent preclinical and clinical observations have offered relevant insights on the etiopathogenesis of late onset Alzheimer's disease (AD) and upregulated immunoinflammatory events have been described as underlying mechanisms involved in the development of AD. Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine produced by several cells of the innate and adaptive immune system, as well as non-immune cells. In the present review, we highlight experimental, genetic, and clinical studies on MIF in rodent models of AD and AD patients, and we discuss emerging therapeutic opportunities for tailored modulation of the activity of MIF, that may potentially be applied to AD patients. Dismantling the exact role of MIF and its receptors in AD may offer novel diagnostic and therapeutic opportunities in AD.


Subject(s)
Alzheimer Disease , Intramolecular Oxidoreductases , Macrophage Migration-Inhibitory Factors , Receptors, Immunologic , Up-Regulation/immunology , Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/immunology , Macrophages , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Rodentia
11.
FASEB J ; 32(6): 3411-3422, 2018 06.
Article in English | MEDLINE | ID: mdl-29401625

ABSTRACT

Sepsis-leading to septic shock-is the leading cause of death in intensive care units. The systemic inflammatory response to infection, which is initiated by activated myeloid cells, plays a key role in the lethal outcome. Macrophage migration inhibitory factor (MIF) is an upstream immunoregulatory mediator, released by myeloid cells, that underlies a common genetic susceptibility to different infections and septic shock. Accordingly, strategies that are aimed at inhibiting the action of MIF have therapeutic potential. Here, we report the isolation and characterization of tailorable, small, affinity-matured nanobodies (Nbs; single-domain antigen-binding fragments derived from camelid heavy-chain Abs) directed against MIF. Of importance, these bioengineered Nbs bind both human and mouse MIFs with nanomolar affinity. NbE5 and NbE10 inhibit key MIF functions that can exacerbate septic shock, such as the tautomerase activity of MIF (by blocking catalytic pocket residues that are critical for MIF's conformation and receptor binding), the TNF-inducing potential, and the ability of MIF to antagonize glucocorticoid action. A lead NbE10, tailored to be a multivalent, half-life extended construct (NbE10-NbAlb8-NbE10), attenuated lethality in murine endotoxemia when administered via single injection, either prophylactically or therapeutically. Hence, Nbs, with their structural and pharmacologic advantages over currently available inhibitors, may be an effective, novel approach to interfere with the action of MIF in septic shock and other conditions of inflammatory end-organ damage.-Sparkes, A., De Baetselier, P., Brys, L., Cabrito, I., Sterckx, Y. G.-J., Schoonooghe, S., Muyldermans, S., Raes, G., Bucala, R., Vanlandschoot, P., Van Ginderachter, J. A., Stijlemans, B. Novel half-life extended anti-MIF nanobodies protect against endotoxic shock.


Subject(s)
Intramolecular Oxidoreductases/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Shock, Septic/drug therapy , Single-Domain Antibodies/pharmacology , Animals , Female , Half-Life , Humans , Intramolecular Oxidoreductases/immunology , Lipopolysaccharides/toxicity , Macrophage Migration-Inhibitory Factors/immunology , Mice , Shock, Septic/chemically induced , Shock, Septic/immunology , Shock, Septic/pathology , Single-Domain Antibodies/immunology
12.
Proc Natl Acad Sci U S A ; 113(8): E997-1005, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26858459

ABSTRACT

The vulnerability to infection of newborns is associated with a limited ability to mount efficient immune responses. High concentrations of adenosine and prostaglandins in the fetal and neonatal circulation hamper the antimicrobial responses of newborn immune cells. However, the existence of mechanisms counterbalancing neonatal immunosuppression has not been investigated. Remarkably, circulating levels of macrophage migration inhibitory factor (MIF), a proinflammatory immunoregulatory cytokine expressed constitutively, were 10-fold higher in newborns than in children and adults. Newborn monocytes expressed high levels of MIF and released MIF upon stimulation with Escherichia coli and group B Streptococcus, the leading pathogens of early-onset neonatal sepsis. Inhibition of MIF activity or MIF expression reduced microbial product-induced phosphorylation of p38 and ERK1/2 mitogen-activated protein kinases and secretion of cytokines. Recombinant MIF used at newborn, but not adult, concentrations counterregulated adenosine and prostaglandin E2-mediated inhibition of ERK1/2 activation and TNF production in newborn monocytes exposed to E. coli. In agreement with the concept that once infection is established high levels of MIF are detrimental to the host, treatment with a small molecule inhibitor of MIF reduced systemic inflammatory response, bacterial proliferation, and mortality of septic newborn mice. Altogether, these data provide a mechanistic explanation for how newborns may cope with an immunosuppressive environment to maintain a certain threshold of innate defenses. However, the same defense mechanisms may be at the expense of the host in conditions of severe infection, suggesting that MIF could represent a potential attractive target for immune-modulating adjunctive therapies for neonatal sepsis.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Innate/physiology , Intramolecular Oxidoreductases/immunology , MAP Kinase Signaling System/immunology , Macrophage Migration-Inhibitory Factors/immunology , Monocytes/immunology , Adult , Animals , Escherichia coli/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Female , Humans , Infant, Newborn , Male , Mice , Streptococcus agalactiae/immunology
13.
Proc Natl Acad Sci U S A ; 113(13): 3597-602, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26976591

ABSTRACT

Pneumococcal meningitis is the most frequent and critical type of bacterial meningitis. Because cytokines play an important role in the pathogenesis of bacterial meningitis, we examined whether functional polymorphisms of the proinflammatory cytokine macrophage migration inhibitory factor (MIF) were associated with morbidity and mortality of pneumococcal meningitis. Two functional MIF promoter polymorphisms, a microsatellite (-794 CATT5-8; rs5844572) and a single-nucleotide polymorphism (-173 G/C; rs755622) were genotyped in a prospective, nationwide cohort of 405 patients with pneumococcal meningitis and in 329 controls matched for age, gender, and ethnicity. Carriages of the CATT7 and -173 C high-expression MIF alleles were associated with unfavorable outcome (P= 0.005 and 0.003) and death (P= 0.03 and 0.01). In a multivariate logistic regression model, shock [odds ratio (OR) 26.0, P= 0.02] and carriage of the CATT7 allele (OR 5.12,P= 0.04) were the main predictors of mortality. MIF levels in the cerebrospinal fluid were associated with systemic complications and death (P= 0.0002). Streptococcus pneumoniae strongly up-regulated MIF production in whole blood and transcription activity of high-expression MIF promoter Luciferase reporter constructs in THP-1 monocytes. Consistent with these findings, treatment with anti-MIF immunoglogulin G (IgG) antibodies reduced bacterial loads and improved survival in a mouse model of pneumococcal pneumonia and sepsis. The present study provides strong evidence that carriage of high-expression MIF alleles is a genetic marker of morbidity and mortality of pneumococcal meningitis and also suggests a potential role for MIF as a target of immune-modulating adjunctive therapy.


Subject(s)
Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Meningitis, Pneumococcal/genetics , Polymorphism, Genetic , Adult , Aged , Animals , Antibodies, Neutralizing/administration & dosage , Case-Control Studies , Cell Line , Disease Models, Animal , Female , Genetic Predisposition to Disease , Humans , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/cerebrospinal fluid , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/cerebrospinal fluid , Macrophage Migration-Inhibitory Factors/immunology , Male , Meningitis, Pneumococcal/cerebrospinal fluid , Meningitis, Pneumococcal/mortality , Mice , Mice, Inbred BALB C , Microsatellite Repeats , Middle Aged , Netherlands/epidemiology , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Prospective Studies , Streptococcus pneumoniae/pathogenicity
14.
Biochemistry ; 57(9): 1523-1532, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29412660

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a pro-inflammatory and tumor-promoting cytokine that occurs in two redox-dependent immunologically distinct conformational isoforms. The disease-related structural isoform of MIF (oxMIF) can be specifically and predominantly detected in the circulation of patients with inflammatory diseases and in tumor tissue, whereas the ubiquitously expressed isoform of MIF (redMIF) is abundantly expressed in healthy and diseased subjects. In this article, we report that cysteine 81 within MIF serves as a "switch cysteine" for the conversion of redMIF to oxMIF. Modulating cysteine 81 by thiol reactive agents leads to significant structural rearrangements of the protein, resulting in a decreased ß-sheet content and an increased random coil content, but maintaining the trimeric quaternary structure. This conformational change in the MIF molecule enables binding of oxMIF-specific antibodies BaxB01 and BaxM159, which showed beneficial activity in animal models of inflammation and cancer. Crystal structure analysis of the MIF-derived EPCALCS peptide, bound in its oxMIF-like conformation by the Fab fragment of BaxB01, revealed that this peptide adopts a curved conformation, making the central thiol protein oxidoreductase motif competent to undergo disulfide shuffling. We conclude that redMIF might reflect a latent zymogenic form of MIF, and formation of oxMIF leads to a physiologically relevant, i.e., enzymatically active, state.


Subject(s)
Cysteine/chemistry , Cysteine/metabolism , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/chemistry , Macrophage Migration-Inhibitory Factors/metabolism , Antibody Specificity , Circular Dichroism , Cysteine/immunology , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Glutathione Disulfide/chemistry , Glutathione Disulfide/metabolism , Humans , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Models, Molecular , Oxidation-Reduction , Protein Conformation , Structure-Activity Relationship
15.
Cell Immunol ; 324: 42-49, 2018 02.
Article in English | MEDLINE | ID: mdl-29397904

ABSTRACT

Macrophage migration Inhibitory Factor (MIF) is a cytokine associated with the pathogenesis of autoimmune inflammatory diseases. There is evidence that MIF functions in a positive-feedback-loop with proinflammatory cytokines and could perpetuate the inflammatory process in Systemic Lupus Erythematosus (SLE).The aim of this study was to assess the effect of recombinant-human-MIF (rhMIF) on the expression of Th1, Th2 and Th17 cytokines in Peripheral Blood Mononuclear Cells (PBMC) from Healthy Subjects (HS) and SLE patients. The PBMC were isolated from SLE patients classified according to the 1997 SLE ACR criteria and HS donors; all subjects included were women from an unrelated Mexican-Mestizo population. The PBMC isolated were stimulated with rhMIF, LPS and ISO-1 in different combinations; Th1, Th2 and Th17cytokine profiles levels were determined by MAGPIX Bio-plex assay in supernatants from cell cultures. We observed in supernatants of PBMCs from HS treated with rhMIF a predominance of Th17 cytokine profile with an increase of IL-17A, IL-17F and IL-21 versus PBMCs from SLE patients, which showed an inflammatory profile represented by increase of IL-6 cytokine. According to SLE remission/activity presented at enrollment in the study (Mex-SLEDAI index), the PBMC from active SLE patients showed higher levels of TNF-α and IL-6 versus PBMC from remission SLE patients. In conclusion, our results suggest that MIF can induce a differential inflammatory response in physiological and pathological conditions with a predominance of a Th17 cytokine profile in PBMC from HS and an increase in TNF-α and IL-6 expression in PBMC from active SLE patients.


Subject(s)
Interleukin-6/immunology , Intramolecular Oxidoreductases/immunology , Lupus Erythematosus, Systemic/immunology , Macrophage Migration-Inhibitory Factors/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Tumor Necrosis Factor-alpha/immunology , Adult , Case-Control Studies , Cytokines/immunology , Female , Humans , Interleukin-17/immunology , Interleukin-6/blood , Intramolecular Oxidoreductases/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lupus Erythematosus, Systemic/blood , Macrophage Migration-Inhibitory Factors/pharmacology , Middle Aged , Primary Cell Culture , Recombinant Proteins/pharmacology , Th1 Cells/drug effects , Th17 Cells/drug effects , Th2 Cells/drug effects , Tumor Necrosis Factor-alpha/blood
16.
Cytokine ; 102: 34-40, 2018 02.
Article in English | MEDLINE | ID: mdl-29275011

ABSTRACT

Red blood cells are widely accepted to be inert carriers of oxygen and haemoglobin, but there is growing evidence that they play a much more critical role in immune function. Macrophage migration inhibitory factor (MIF) is a key cytokine in disease with additional oxido-reductase activity, which aids in managing oxidative stress. Although two studies have reported the presence of MIF in red blood cells, no study has quantified the levels of this protein. In this study, freshly isolated plasma, platelets, leukocytes, and red blood cells from healthy individuals were collected and the concentration of MIF was determined using an enzyme linked immunosorbent assay. This analysis demonstrated that MIF in red blood cells was present at 25 µg per millilitre of whole blood, which is greater than99% of the total MIF and 1000-fold higher concentration than plasma. This result was supported by electrophoresis and Western blot analysis, which identified MIF in its monomer structural form following sample processing. Furthermore, by assessing the level of tautomerase activity in red blood cell fractions in the presence of a MIF inhibitor, it was determined that the red blood cell-derived MIF was also functionally active. Together, these findings have implications on the effect of haemolysis during sample preparation and provide some clue into the inflammatory processes that occur following haemolysis in vivo. These results support the hypothesis that red blood cells are a major reservoir of this inflammatory protein and may play a role in inflammation.


Subject(s)
Erythrocytes/metabolism , Intramolecular Oxidoreductases/blood , Macrophage Migration-Inhibitory Factors/blood , Adult , Female , Humans , Intramolecular Oxidoreductases/analysis , Intramolecular Oxidoreductases/immunology , Leukocytes/metabolism , Macrophage Migration-Inhibitory Factors/analysis , Macrophage Migration-Inhibitory Factors/immunology , Male , Middle Aged
17.
Mol Ther ; 25(7): 1665-1675, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28284981

ABSTRACT

Exosomes are small membrane-bound vesicular particles generated by most cells for intercellular communication and regulation. During biogenesis, specific lipids, RNAs, proteins, and carbohydrates are enriched and packaged into the vesicles so that the exosomal contents reflect not only the source but also the physiological conditions of the parental cells. These exosomes transport materials or signals to the target cells for diverse physiological purposes. Our study focused on the exosomes derived from M1-polarized, proinflammatory macrophages for the possibility of using M1 exosomes as an immunopotentiator for a cancer vaccine. The M1 exosomes displayed a tropism toward lymph nodes after subcutaneous injection, primarily taken up by the local macrophages and dendritic cells, and they induced the release of a pool of Th1 cytokines. We found that M1, but not M2, exosomes enhanced activity of lipid calcium phosphate (LCP) nanoparticle-encapsulated Trp2 vaccine, and they induced a stronger antigen-specific cytotoxic T cell response. The M1 exosomes proved to be a more potent immunopotentiator than CpG oligonucleotide when used with LCP nanoparticle vaccine in a melanoma growth inhibition study. Thus, our study indicated that exosomes derived from M1-polarized macrophages could be used as a vaccine adjuvant.


Subject(s)
Cancer Vaccines/administration & dosage , Exosomes/chemistry , Immunologic Factors/pharmacology , Lymph Nodes/drug effects , Melanoma, Experimental/therapy , Skin Neoplasms/therapy , Adjuvants, Immunologic/pharmacology , Animals , Calcium Phosphates/chemistry , Drug Carriers , Drug Compounding , Exosomes/immunology , Female , Gene Expression , Immunologic Factors/chemistry , Inflammation/chemically induced , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Macrophages/immunology , Macrophages/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Oligodeoxyribonucleotides/pharmacology , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
18.
Semin Immunol ; 26(5): 409-14, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24768088

ABSTRACT

Traumatic spinal cord injury (SCI) activates the hypothalamic-pituitary-adrenal (HPA) axis, a potent neuroendocrine regulator of stress and inflammation. SCI also elicits a profound and sustained intraspinal and systemic inflammatory response. Together, stress hormones and inflammatory mediators will affect the growth and survival of neural and non-neural cells and ultimately neurologic recovery after SCI. Glucocorticoids (GCs) are endogenous anti-inflammatory steroids that are synthesized in response to stress or injury, in part to regulate inflammation. Exogenous synthetic GCs are often used for similar purposes in various diseases; however, their safety and efficacy in pre-clinical and clinical SCI is controversial. The relatively recent discovery that macrophage migration inhibitory factor (MIF) is produced throughout the body and can override the anti-inflammatory effects of GCs may provide unique insight to the importance of endogenous and exogenous GCs after SCI. Here, we review both GCs and MIF and discuss the potential relevance of their interactions after SCI, especially their role in regulating maladaptive mechanisms of plasticity and repair that may contribute to the onset and maintenance of neuropathic pain.


Subject(s)
Glucocorticoids/metabolism , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Neuralgia/metabolism , Receptors, Glucocorticoid/metabolism , Spinal Cord Injuries/metabolism , Animals , Gene Expression Regulation , Glucocorticoids/genetics , Glucocorticoids/immunology , Humans , Hypothalamo-Hypophyseal System/immunology , Hypothalamo-Hypophyseal System/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/immunology , Neuralgia/genetics , Neuralgia/immunology , Neuralgia/pathology , Pituitary-Adrenal System/immunology , Pituitary-Adrenal System/metabolism , Protein Interaction Mapping , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/immunology , Signal Transduction , Spinal Cord Injuries/genetics , Spinal Cord Injuries/immunology , Spinal Cord Injuries/pathology
19.
J Immunol ; 195(8): 3537-45, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26342029

ABSTRACT

Aspirin-exacerbated respiratory disease (AERD), a severe eosinophilic inflammatory disorder of the airways, involves overproduction of cysteinyl leukotrienes (cysLTs), activation of airway mast cells (MCs), and bronchoconstriction in response to nonselective cyclooxygenase inhibitors that deplete homeostatic PGE2. The mechanistic basis for MC activation in this disorder is unknown. We now demonstrate that patients with AERD have markedly increased epithelial expression of the alarmin-like cytokine IL-33 in nasal polyps, as compared with polyps from aspirin-tolerant control subjects. The murine model of AERD, generated by dust mite priming of mice lacking microsomal PGE2 synthase (ptges(-/-) mice), shows a similar upregulation of IL-33 protein in the airway epithelium, along with marked eosinophilic bronchovascular inflammation. Deletion of leukotriene C4 synthase, the terminal enzyme needed to generate cysLTs, eliminates the increased IL-33 content of the ptges(-/-) lungs and sharply reduces pulmonary eosinophilia and basal secretion of MC products. Challenges of dust mite-primed ptges(-/-) mice with lysine aspirin induce IL-33-dependent MC activation and bronchoconstriction. Thus, IL-33 is a component of a cysLT-driven innate type 2 immune response that drives pathogenic MC activation and contributes substantially to AERD pathogenesis.


Subject(s)
Asthma, Aspirin-Induced/immunology , Immunity, Innate , Interleukin-33/immunology , Leukotrienes/immunology , Mast Cells/immunology , Adolescent , Adult , Aged , Animals , Asthma, Aspirin-Induced/genetics , Asthma, Aspirin-Induced/pathology , Dinoprostone/genetics , Dinoprostone/immunology , Female , Humans , Interleukin-33/genetics , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Leukotrienes/genetics , Male , Mast Cells/pathology , Mice , Mice, Knockout , Middle Aged , Prostaglandin-E Synthases , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/pathology
20.
J Immunol ; 194(12): 5937-47, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25972487

ABSTRACT

The development of effective cancer vaccines remains an urgent, but as yet unmet, clinical need. This deficiency is in part due to an incomplete understanding of how to best invoke dendritic cells (DC) that are crucial for the induction of tumor-specific CD8(+) T cells capable of mediating durable protective immunity. In this regard, elevated expression of the transcription factor X box-binding protein 1 (XBP1) in DC appears to play a decisive role in promoting the ability of DC to cross-present Ags to CD8(+) T cells in the therapeutic setting. Delivery of DNA vaccines encoding XBP1 and tumor Ag to skin DC resulted in increased IFN-α production by plasmacytoid DC (pDC) from skin/tumor draining lymph nodes and the cross-priming of Ag-specific CD8(+) T cell responses associated with therapeutic benefit. Antitumor protection was dependent on cross-presenting Batf3(+) DC, pDC, and CD8(+) T cells. CD103(+) DC from the skin/tumor draining lymph nodes of the immunized mice appeared responsible for activation of Ag-specific naive CD8(+) T cells, but were dependent on pDC for optimal effectiveness. Similarly, human XBP1 improved the capacity of human blood- and skin-derived DC to activate human T cells. These data support an important intrinsic role for XBP1 in DC for effective cross-priming and orchestration of Batf3(+) DC-pDC interactions, thereby enabling effective vaccine induction of protective antitumor immunity.


Subject(s)
Cancer Vaccines/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Neoplasms/immunology , Vaccines, DNA/immunology , Animals , Antigen Presentation , Antigens, CD/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Basic-Leucine Zipper Transcription Factors/metabolism , Bone Marrow/immunology , Bone Marrow/pathology , Cancer Vaccines/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Female , HSP72 Heat-Shock Proteins/genetics , HSP72 Heat-Shock Proteins/immunology , Humans , Integrin alpha Chains/metabolism , Interferon-alpha/biosynthesis , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lymphocyte Activation , Melanoma, Experimental , Mice , Mice, Knockout , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , Phenotype , Rats , Regulatory Factor X Transcription Factors , Repressor Proteins/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Toll-Like Receptor 3/metabolism , Transcription Factors/genetics , Transcription Factors/immunology , X-Box Binding Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL