Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Nat Immunol ; 22(11): 1382-1390, 2021 11.
Article in English | MEDLINE | ID: mdl-34663978

ABSTRACT

Intergenerational inheritance of immune traits linked to epigenetic modifications has been demonstrated in plants and invertebrates. Here we provide evidence for transmission of trained immunity across generations to murine progeny that survived a sublethal systemic infection with Candida albicans or a zymosan challenge. The progeny of trained mice exhibited cellular, developmental, transcriptional and epigenetic changes associated with the bone marrow-resident myeloid effector and progenitor cell compartment. Moreover, the progeny of trained mice showed enhanced responsiveness to endotoxin challenge, alongside improved protection against systemic heterologous Escherichia coli and Listeria monocytogenes infections. Sperm DNA of parental male mice intravenously infected with the fungus C. albicans showed DNA methylation differences linked to immune gene loci. These results provide evidence for inheritance of trained immunity in mammals, enhancing protection against infections.


Subject(s)
Candida albicans/immunology , Candidiasis/immunology , Escherichia coli Infections/immunology , Escherichia coli/immunology , Heredity , Immunity, Innate/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Myeloid Cells/immunology , Animals , Candida albicans/pathogenicity , Candidiasis/genetics , Candidiasis/metabolism , Candidiasis/microbiology , Cells, Cultured , DNA Methylation , Disease Models, Animal , Epigenesis, Genetic , Escherichia coli/pathogenicity , Escherichia coli Infections/genetics , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Listeria monocytogenes/pathogenicity , Listeriosis/genetics , Listeriosis/metabolism , Listeriosis/microbiology , Male , Mice, Transgenic , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Spermatozoa/immunology , Spermatozoa/metabolism , Transcription, Genetic
4.
Nat Immunol ; 16(9): 970-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26168081

ABSTRACT

Interleukin 17-producing helper T cells (T(H)17 cells) have a major role in protection against infections and in mediating autoimmune diseases, yet the mechanisms involved are incompletely understood. We found that interleukin 26 (IL-26), a human T(H)17 cell-derived cytokine, is a cationic amphipathic protein that kills extracellular bacteria via membrane-pore formation. Furthermore, T(H)17 cell-derived IL-26 formed complexes with bacterial DNA and self-DNA released by dying bacteria and host cells. The resulting IL-26-DNA complexes triggered the production of type I interferon by plasmacytoid dendritic cells via activation of Toll-like receptor 9, but independently of the IL-26 receptor. These findings provide insights into the potent antimicrobial and proinflammatory function of T(H)17 cells by showing that IL-26 is a natural human antimicrobial that promotes immune sensing of bacterial and host cell death.


Subject(s)
DNA, Bacterial/immunology , DNA/immunology , Immunity, Innate/immunology , Interleukins/immunology , Th17 Cells/immunology , Toll-Like Receptor 9/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Interferon Type I/immunology , Interferon Type I/metabolism , Mice , Psoriasis/immunology , Receptors, Interleukin/immunology , Receptors, Interleukin/metabolism
5.
Eur J Immunol ; 52(10): 1676-1679, 2022 10.
Article in English | MEDLINE | ID: mdl-35689332

ABSTRACT

We used unsupervised immunophenotyping of blood leukocytes and measured cytokine production by innate immune cell exposed to LPS and R848. We show that COVID-19 induces a rapid, transient upregulation of myeloid-derived suppressor cells (MDSCs) accompanied by a rapid, sustained (up to 3 months) hyporesponsiveness of dendritic cells and monocytes. Blood MDSCs may represent biomarkers and targets for intervention strategies in COVID-19 patients.


Subject(s)
COVID-19 , Immune System Diseases , Myeloid-Derived Suppressor Cells , Biomarkers , Cytokines/pharmacology , Humans , Immunity, Innate , Lipopolysaccharides
6.
FASEB J ; 35(5): e21418, 2021 05.
Article in English | MEDLINE | ID: mdl-33774873

ABSTRACT

Constitutively expressed by innate immune cells, the cytokine macrophage migration inhibitory factor (MIF) initiates host immune responses and drives pathogenic responses in infectious, inflammatory, and autoimmune diseases. Dendritic cells (DCs) express high levels of MIF, but the role of MIF in DC function remains poorly characterized. As migration is critical for DC immune surveillance, we investigated whether MIF promoted the migration of DCs. In classical transwell experiments, MIF-/- bone marrow-derived DCs (BMDCs) or MIF+/+ BMDCs treated with ISO-1, an inhibitor of MIF, showed markedly reduced spontaneous migration and chemotaxis. CD74-/- BMDCs that are deficient in the ligand-binding component of the cognate MIF receptor exhibited a migration defect similar to that of MIF-/- BMDCs. Adoptive transfer experiments of LPS-matured MIF+/+ and MIF-/- and of CD74+/+ and CD74-/- BMDCs injected into the hind footpads of homologous or heterologous mice showed that the autocrine and paracrine MIF activity acting via CD74 contributed to the recruitment of DCs to the draining lymph nodes. Mechanistically, MIF activated the Src/PI3K signaling pathway and myosin II complexes, which were required for the migration of BMDCs. Altogether, these data show that the cytokine MIF exerts chemokine-like activity for DC motility and trafficking.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/physiology , Chemotaxis , Dendritic Cells/physiology , Histocompatibility Antigens Class II/physiology , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Myosin Type II/metabolism , Phosphatidylinositol 3-Kinases/metabolism , src-Family Kinases/metabolism , Animals , Cells, Cultured , Chemokines/metabolism , Dendritic Cells/cytology , Immunity , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myosin Type II/genetics , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction , src-Family Kinases/genetics
7.
J Surg Res ; 272: 132-138, 2022 04.
Article in English | MEDLINE | ID: mdl-34973547

ABSTRACT

BACKGROUND: Biological xenografts using tubulized porcine pericardium are an alternative to replace infected prosthetic graft. We recently reported an innovative technique using a stapled porcine pericardial bioconduit for immediate vascular reconstruction in emergency. The objective of this study is to compare the growth and adherence to grafts of bacteria and yeast incubated with stapled porcine pericardium, sutured or naked pericardium. MATERIAL AND METHODS: One square centimeter of porcine pericardial patches, with or without staples or sutures, was incubated with 105 colony forming units of Escherichia coli, Staphylococcus aureus, Staphylococcus epidermidis, and Candida albicans for 1, 6, and 24 h. The medium was collected to quantify planktonic microorganisms, while grafts were sonicated to quantify adherent microorganisms. Dacron and Dacron Silver were analyzed in parallel as synthetic reference prostheses. RESULTS: Stapled porcine pericardium reduced the growth and the adherence of E coli (2- to 30-fold; P < 0.0005), S aureus (11- to 1000-fold; P < 0.0006), S epidermidis (>500-fold; P < 0.0001), and C albicans (12- to 50-fold; P < 0.0001) when compared to medium alone (growth) and pericardium or Dacron (adherence). Native and sutured porcine pericardium interfered with the growth and the adherence of E coli and C albicans, and Dacron with that of S epidermidis. As expected, Dacron Silver was robustly bactericidal. CONCLUSIONS: Stapled porcine pericardium exhibited a lower susceptibility to infection by bacteria and yeasts in vitro when compared to the native and sutured porcine pericardium. Stapled porcine pericardium might be a good option for rapid vascular grafting without increasing infectivity.


Subject(s)
Blood Vessel Prosthesis , Polyethylene Terephthalates , Animals , Escherichia coli , Humans , Pericardium , Silver , Staphylococcus aureus , Staphylococcus epidermidis , Swine
8.
J Infect Dis ; 222(11): 1869-1881, 2020 11 09.
Article in English | MEDLINE | ID: mdl-31889191

ABSTRACT

BACKGROUND: The innate immune system recalls a challenge to adapt to a secondary challenge, a phenomenon called trained immunity. Training involves cellular metabolic, epigenetic and functional reprogramming, but how broadly trained immunity protects from infections is unknown. For the first time, we addressed whether trained immunity provides protection in a large panel of preclinical models of infections. METHODS: Mice were trained and subjected to systemic infections, peritonitis, enteritis, and pneumonia induced by Staphylococcus aureus, Listeria monocytogenes, Escherichia coli, Citrobacter rodentium, and Pseudomonas aeruginosa. Bacteria, cytokines, leukocytes, and hematopoietic precursors were quantified in blood, bone marrow, and organs. The role of monocytes/macrophages, granulocytes, and interleukin 1 signaling was investigated using depletion or blocking approaches. RESULTS: Induction of trained immunity protected mice in all preclinical models, including when training and infection were initiated in distant organs. Trained immunity increased bone marrow hematopoietic progenitors, blood Ly6Chigh inflammatory monocytes and granulocytes, and sustained blood antimicrobial responses. Monocytes/macrophages and interleukin 1 signaling were required to protect trained mice from listeriosis. Trained mice were efficiently protected from peritonitis and listeriosis for up to 5 weeks. CONCLUSIONS: Trained immunity confers broad-spectrum protection against lethal bacterial infections. These observations support the development of trained immunity-based strategies to improve host defenses.


Subject(s)
Bacterial Infections/immunology , Immunity, Innate , Animals , Bacterial Infections/microbiology , Bone Marrow , Citrobacter rodentium , Cytokines/metabolism , Escherichia coli , Female , Interleukin-1/metabolism , Listeria monocytogenes , Listeriosis/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Pseudomonas aeruginosa , Sepsis/immunology , Signal Transduction , Staphylococcal Infections/immunology , Staphylococcus aureus
9.
EMBO Rep ; 19(12)2018 12.
Article in English | MEDLINE | ID: mdl-30337494

ABSTRACT

Chitin is the second most abundant polysaccharide in nature and linked to fungal infection and asthma. However, bona fide immune receptors directly binding chitin and signaling immune activation and inflammation have not been clearly identified because polymeric crude chitin with unknown purity and molecular composition has been used. By using defined chitin (N-acetyl-glucosamine) oligomers, we here identify six-subunit-long chitin chains as the smallest immunologically active motif and the innate immune receptor Toll-like receptor (TLR2) as a primary fungal chitin sensor on human and murine immune cells. Chitin oligomers directly bind TLR2 with nanomolar affinity, and this fungal TLR2 ligand shows overlapping and distinct signaling outcomes compared to known mycobacterial TLR2 ligands. Unexpectedly, chitin oligomers composed of five or less subunits are inactive, hinting to a size-dependent system of immuno-modulation that appears conserved in plants and humans. Since blocking of the chitin-TLR2 interaction effectively prevents chitin-mediated inflammation in vitro and in vivo, our study highlights the chitin-TLR2 interaction as a potential target for developing novel therapies in chitin-related pathologies and fungal disease.


Subject(s)
Chitin/chemistry , Chitin/metabolism , Fungi/metabolism , Inflammation/metabolism , Inflammation/pathology , Toll-Like Receptor 2/metabolism , Animals , Cell Wall/drug effects , Cell Wall/metabolism , Chitinases/metabolism , Female , Humans , Hydrophobic and Hydrophilic Interactions , Immunologic Factors/pharmacology , Ligands , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice, Inbred C57BL , Mice, Knockout , THP-1 Cells , Toll-Like Receptor 1/agonists , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/chemistry , Zymosan/metabolism
10.
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
11.
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
12.
J Toxicol Environ Health A ; 78(13-14): 871-85, 2015.
Article in English | MEDLINE | ID: mdl-26167753

ABSTRACT

Chronic inhalation of grain dust is associated with asthma and chronic bronchitis in grain worker populations. Exposure to fungal particles was postulated to be an important etiologic agent of these pathologies. Fusarium species frequently colonize grain and straw and produce a wide array of mycotoxins that impact human health, necessitating an evaluation of risk exposure by inhalation of Fusarium and its consequences on immune responses. Data showed that Fusarium culmorum is a frequent constituent of aerosols sampled during wheat harvesting in the Vaud region of Switzerland. The aim of this study was to examine cytokine/chemokine responses and innate immune sensing of F. culmorum in bone-marrow-derived dendritic cells and macrophages. Overall, dendritic cells and macrophages responded to F. culmorum spores but not to its secreted components (i.e., mycotoxins) by releasing large amounts of macrophage inflammatory protein (MIP)-1α, MIP-1ß, MIP-2, monocyte chemoattractant protein (MCP)-1, RANTES, and interleukin (IL)-12p40, intermediate amounts of tumor necrosis factor (TNF), IL-6, IL-12p70, IL-33, granulocyte colony-stimulating factor (G-CSF), and interferon gamma-induced protein (IP-10), but no detectable amounts of IL-4 and IL-10, a pattern of mediators compatible with generation of Th1 or Th17 antifungal protective immune responses rather than with Th2-dependent allergic responses. The sensing of F. culmorum spores by dendritic cells required dectin-1, the main pattern recognition receptor involved in ß-glucans detection, but likely not MyD88 and TRIF-dependent Toll-like receptors. Taken together, our results indicate that F. culmorum stimulates potently innate immune cells in a dectin-1-dependent manner, suggesting that inhalation of F. culmorum from grain dust may promote immune-related airway diseases in exposed worker populations.


Subject(s)
Air Microbiology , Dendritic Cells/immunology , Fusarium/physiology , Immunity, Innate , Aerosols/analysis , Air Pollutants/analysis , Animals , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/microbiology , Female , Macrophages/immunology , Mice , Mice, Inbred C57BL , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/metabolism , Specific Pathogen-Free Organisms , Switzerland , Triticum
13.
Biochim Biophys Acta ; 1833(6): 1498-510, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23499872

ABSTRACT

Sirtuins (SIRT1-7) are NAD(+)-dependent histone deacetylases (HDACs) that play an important role in the control of metabolism and proliferation and the development of age-associated diseases like oncologic, cardiovascular and neurodegenerative diseases. Cambinol was originally described as a compound inhibiting the activity of SIRT1 and SIRT2, with efficient anti-tumor activity in vivo. Here, we studied the effects of cambinol on microbial sensing by mouse and human immune cells and on host innate immune responses in vivo. Cambinol inhibited the expression of cytokines (TNF, IL-1ß, IL-6, IL-12p40, and IFN-γ), NO and CD40 by macrophages, dendritic cells, splenocytes and whole blood stimulated with a broad range of microbial and inflammasome stimuli. Sirtinol, an inhibitor of SIRT1 and SIRT2 structurally related to cambinol, also decreased macrophage response to TLR stimulation. On the contrary, selective inhibitors of SIRT1 (EX-527 and CHIC-35) and SIRT2 (AGK2 and AK-7) used alone or in combination had no inhibitory effect, suggesting that cambinol and sirtinol act by targeting more than just SIRT1 and SIRT2. Cambinol and sirtinol at anti-inflammatory concentrations also did not inhibit SIRT6 activity in in vitro assay. At the molecular level, cambinol impaired stimulus-induced phosphorylation of MAPKs and upstream MEKs. Going well along with its powerful anti-inflammatory activity, cambinol reduced TNF blood levels and bacteremia and improved survival in preclinical models of endotoxic shock and septic shock. Altogether, our data suggest that pharmacological inhibitors of sirtuins structurally related to cambinol may be of clinical interest to treat inflammatory diseases.


Subject(s)
Immunity, Innate/immunology , Inflammation/prevention & control , Macrophages/drug effects , Naphthalenes/pharmacology , Pyrimidinones/pharmacology , Shock, Septic/prevention & control , Sirtuins/antagonists & inhibitors , Animals , Apoptosis , Benzamides/pharmacology , Blotting, Western , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Flow Cytometry , Humans , Inflammation/immunology , Inflammation/microbiology , Klebsiella Infections/immunology , Klebsiella Infections/microbiology , Klebsiella Infections/prevention & control , Klebsiella pneumoniae , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Naphthols/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Shock, Septic/immunology , Shock, Septic/microbiology
14.
J Infect Dis ; 207(2): 331-9, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23125447

ABSTRACT

The cytokine macrophage migration inhibitory factor (MIF) is an important component of the early proinflammatory response of the innate immune system. However, the antimicrobial defense mechanisms mediated by MIF remain fairly mysterious. In the present study, we examined whether MIF controls bacterial uptake and clearance by professional phagocytes, using wild-type and MIF-deficient macrophages. MIF deficiency did not affect bacterial phagocytosis, but it strongly impaired the killing of gram-negative bacteria by macrophages and host defenses against gram-negative bacterial infection, as shown by increased mortality in a Klebsiella pneumonia model. Consistent with MIF's regulatory role of Toll-like 4 expression in macrophages, MIF-deficient cells stimulated with lipopolysaccharide or Escherichia coli exhibited reduced nuclear factor κB activity and tumor necrosis factor (TNF) production. Addition of recombinant MIF or TNF corrected the killing defect of MIF-deficient macrophages. Together, these data show that MIF is a key mediator of host responses against gram-negative bacteria, acting in part via a modulation of bacterial killing by macrophages.


Subject(s)
Gram-Negative Bacteria/immunology , Klebsiella Infections/immunology , Klebsiella pneumoniae/pathogenicity , Macrophage Migration-Inhibitory Factors/deficiency , Macrophages/immunology , Animals , Cell Line , Cells, Cultured , Humans , Klebsiella Infections/microbiology , Klebsiella pneumoniae/immunology , Macrophage Migration-Inhibitory Factors/immunology , Macrophage Migration-Inhibitory Factors/metabolism , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Phagocytosis/immunology
15.
Cells ; 13(4)2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38391927

ABSTRACT

Patients admitted to the intensive care unit (ICU) often experience endotoxemia, nosocomial infections and sepsis. Polymorphonuclear and monocytic myeloid-derived suppressor cells (PMN-MDSCs and M-MDSCs) can have an important impact on the development of infectious diseases, but little is known about their potential predictive value in critically ill patients. Here, we used unsupervised flow cytometry analyses to quantify MDSC-like cells in healthy subjects challenged with endotoxin and in critically ill patients admitted to intensive care units and at risk of developing infections. Cells phenotypically similar to PMN-MDSCs and M-MDSCs increased after endotoxin challenge. Similar cells were elevated in patients at ICU admission and normalized at ICU discharge. A subpopulation of M-MDSC-like cells expressing intermediate levels of CD15 (CD15int M-MDSCs) was associated with overall mortality (p = 0.02). Interestingly, the high abundance of PMN-MDSCs and CD15int M-MDSCs was a good predictor of mortality (p = 0.0046 and 0.014), with area under the ROC curve for mortality of 0.70 (95% CI = 0.4-1.0) and 0.86 (0.62-1.0), respectively. Overall, our observations support the idea that MDSCs represent biomarkers for sepsis and that flow cytometry monitoring of MDSCs may be used to risk-stratify ICU patients for targeted therapy.


Subject(s)
Endotoxemia , Myeloid-Derived Suppressor Cells , Humans , Critical Illness , Prognosis , Critical Care , Endotoxins
16.
J Biol Chem ; 287(10): 7446-55, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22238348

ABSTRACT

The macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine that recently emerged as an attractive therapeutic target for a variety of diseases. A diverse panel of fully human anti-MIF antibodies was generated by selection from a phage display library and extensively analyzed in vitro. Epitope mapping studies identified antibodies specific for linear as well as structural epitopes. Experimental animal studies revealed that only those antibodies binding epitopes within amino acids 50-68 or 86-102 of the MIF molecule exerted protective effects in models of sepsis or contact hypersensitivity. Within the MIF protein, these two binding regions form a ß-sheet structure that includes the MIF oxidoreductase motif. We therefore conclude that this ß-sheet structure is a crucial region for MIF activity and a promising target for anti-MIF antibody therapy.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Intramolecular Oxidoreductases/chemistry , Macrophage Migration-Inhibitory Factors/chemistry , Amino Acid Motifs , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Dermatitis, Contact/drug therapy , Dermatitis, Contact/immunology , Disease Models, Animal , Humans , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Mice , Sepsis/drug therapy , Sepsis/immunology
17.
Blood ; 117(4): 1205-17, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-20956800

ABSTRACT

Regulated by histone acetyltransferases and deacetylases (HDACs), histone acetylation is a key epigenetic mechanism controlling chromatin structure, DNA accessibility, and gene expression. HDAC inhibitors induce growth arrest, differentiation, and apoptosis of tumor cells and are used as anticancer agents. Here we describe the effects of HDAC inhibitors on microbial sensing by macrophages and dendritic cells in vitro and host defenses against infection in vivo. HDAC inhibitors down-regulated the expression of numerous host defense genes, including pattern recognition receptors, kinases, transcription regulators, cytokines, chemokines, growth factors, and costimulatory molecules as assessed by genome-wide microarray analyses or innate immune responses of macrophages and dendritic cells stimulated with Toll-like receptor agonists. HDAC inhibitors induced the expression of Mi-2ß and enhanced the DNA-binding activity of the Mi-2/NuRD complex that acts as a transcriptional repressor of macrophage cytokine production. In vivo, HDAC inhibitors increased the susceptibility to bacterial and fungal infections but conferred protection against toxic and septic shock. Thus, these data identify an essential role for HDAC inhibitors in the regulation of the expression of innate immune genes and host defenses against microbial pathogens.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Immunity, Innate/drug effects , Infections/immunology , Toll-Like Receptors/agonists , Animals , Cells, Cultured , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Evaluation, Preclinical , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Immunity, Innate/genetics , Infections/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Microarray Analysis
18.
J Infect Dis ; 205(6): 944-54, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22315281

ABSTRACT

BACKGROUND: Aspergillus fumigatus causes invasive aspergillosis, a potentially fatal infection in oncohematological patients. Innate immune detection of A. fumigatus involves Toll-like receptor (TLR) 4 and TLR2, which forms a heterodimer with either TLR1 or TLR6. The role of those coreceptors in Aspergillus sensing is unknown. METHODS: Cytokine production was measured in bone marrow-derived macrophages (BMDMs) from wild-type (WT) and TLR-deficient mice after incubation with a WT and an immunogenic RodA-deficient (ΔrodA-47) strain of A. fumigatus and in lungs from these mice after intranasal mold inoculation. Aspergillus fumigatus-mediated NF-κB activation was measured in HEK293T cells transfected with plasmids expressing mouse or human TLRs. RESULTS: Bone marrow-derived macrophages from TLR1- and TLR6-deficient mice produced lower amounts of interleukin 12p40, CXCL2, interleukin 6, and tumor necrosis factor α than BMDMs from WT mice after stimulation with A. fumigatus. Lungs from TLR1- and TLR6-deficient mice had diminished CXCL1 and CXCL2 production and increased fungal burden after intranasal inoculation of ΔrodA A. fumigatus compared with lungs from WT mice. ΔrodA strain-mediated NF-κB activation was observed in HEK293T cells expressing mouse TLR2/1, mouse TLR2/6, and human TLR2/1 but not human TLR2/6. CONCLUSIONS: Innate immune detection of A. fumigatus is mediated by TLR4 and TLR2 together with TLR1 or TLR6 in mice and TLR1 but not TLR6 in humans.


Subject(s)
Aspergillus fumigatus/pathogenicity , Gene Deletion , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 6/metabolism , Animals , Aspergillosis/genetics , Chemokine CXCL2/metabolism , Female , HEK293 Cells , Humans , Immunity, Innate , Interleukin-12 Subunit p40/biosynthesis , Interleukin-6/biosynthesis , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Species Specificity , Tumor Necrosis Factor-alpha/metabolism
19.
J Immunother Cancer ; 11(1)2023 01.
Article in English | MEDLINE | ID: mdl-36593065

ABSTRACT

BACKGROUND: Agonistic anti-CD40 monoclonal antibodies (mAbs) have emerged as promising immunotherapeutic compounds with impressive antitumor effects in mouse models. However, preclinical and clinical studies faced dose-limiting toxicities mediated by necroinflammatory liver disease. An effective prophylactic treatment for liver immune-related adverse events that does not suppress specific antitumor immunity remains to be found. METHODS: We used different mouse models and time-resolved single-cell RNA-sequencing to characterize the pathogenesis of anti-CD40 mAb induced liver toxicity. Subsequently, we developed an antibody-based treatment protocol to selectively target red blood cells (RBCs) for erythrophagocytosis in the liver, inducing an anti-inflammatory liver macrophage reprogramming. RESULTS: We discovered that CD40 signaling in Clec4f+ Kupffer cells is the non-redundant trigger of anti-CD40 mAb-induced liver toxicity. Taking advantage of the highly specific functionality of liver macrophages to clear antibody-tagged RBCs from the blood, we hypothesized that controlled erythrophagocytosis and the linked anti-inflammatory signaling by the endogenous metabolite heme could be exploited to reprogram liver macrophages selectively. Repeated low-dose administration of a recombinant murine Ter119 antibody directed RBCs for selective phagocytosis in the liver and skewed the phenotype of liver macrophages into a Hmoxhigh/Marcohigh/MHCIIlow anti-inflammatory phenotype. This unique mode of action prevented necroinflammatory liver disease following high-dose administration of anti-CD40 mAbs. In contrast, extrahepatic inflammation, antigen-specific immunity, and antitumor activity remained unaffected in Ter119 treated animals. CONCLUSIONS: Our study offers a targeted approach to uncouple CD40-augmented antitumor immunity in peripheral tissues from harmful inflammatoxicity in the liver.


Subject(s)
Antineoplastic Agents , Neoplasms , Mice , Animals , Kupffer Cells/metabolism , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Immunotherapy/methods , Liver
20.
Proc Natl Acad Sci U S A ; 106(7): 2348-52, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19181857

ABSTRACT

Toll-like receptor 4 (TLR4), the signal-transducing molecule of the LPS receptor complex, plays a fundamental role in the sensing of LPS from gram-negative bacteria. Activation of TLR4 signaling pathways by LPS is a critical upstream event in the pathogenesis of gram-negative sepsis, making TLR4 an attractive target for novel antisepsis therapy. To validate the concept of TLR4-targeted treatment strategies in gram-negative sepsis, we first showed that TLR4(-/-) and myeloid differentiation primary response gene 88 (MyD88)(-/-) mice were fully resistant to Escherichia coli-induced septic shock, whereas TLR2(-/-) and wild-type mice rapidly died of fulminant sepsis. Neutralizing anti-TLR4 antibodies were then generated using a soluble chimeric fusion protein composed of the N-terminal domain of mouse TLR4 (amino acids 1-334) and the Fc portion of human IgG1. Anti-TLR4 antibodies inhibited intracellular signaling, markedly reduced cytokine production, and protected mice from lethal endotoxic shock and E. coli sepsis when administered in a prophylactic and therapeutic manner up to 13 h after the onset of bacterial sepsis. These experimental data provide strong support for the concept of TLR4-targeted therapy for gram-negative sepsis.


Subject(s)
Gene Expression Regulation, Bacterial , Gram-Negative Bacteria/metabolism , Sepsis/microbiology , Toll-Like Receptor 4/physiology , Animals , Escherichia coli/metabolism , Humans , Immunoglobulin G/chemistry , Lipopolysaccharides/metabolism , Mice , Mice, Inbred C57BL , Models, Genetic , Myeloid Differentiation Factor 88/genetics , Recombinant Fusion Proteins/metabolism , Sepsis/genetics , Sepsis/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL