Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Immunity ; 53(4): 793-804.e9, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32910906

ABSTRACT

Allergies are considered to represent mal-directed type 2 immune responses against mostly innocuous exogenous compounds. Immunoglobulin E (IgE) antibodies are a characteristic feature of allergies and mediate hypersensitivity against allergens through activation of effector cells, particularly mast cells (MCs). Although the physiological functions of this dangerous branch of immunity have remained enigmatic, recent evidence shows that allergic immune reactions can help to protect against the toxicity of venoms. Because bacteria are a potent alternative source of toxins, we assessed the possible role of allergy-like type 2 immunity in antibacterial host defense. We discovered that the adaptive immune response against Staphylococcus aureus (SA) skin infection substantially improved systemic host defense against secondary SA infections in mice. Moreover, this acquired protection depended on IgE effector mechanisms and MCs. Importantly, our results reveal a previously unknown physiological function of allergic immune responses, IgE antibodies, and MCs in host defense against a pathogenic bacterium.


Subject(s)
Adaptive Immunity/immunology , Immunoglobulin E/immunology , Mast Cells/immunology , Staphylococcal Infections/immunology , Staphylococcal Skin Infections/immunology , Staphylococcus aureus/immunology , Allergens/immunology , Animals , Female , Hypersensitivity/immunology , Hypersensitivity/microbiology , Mast Cells/microbiology , Mice , Mice, Inbred C57BL , Skin/immunology , Skin/microbiology , Staphylococcal Infections/microbiology , Staphylococcal Skin Infections/microbiology
2.
Nat Immunol ; 17(12): 1361-1372, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27798618

ABSTRACT

Hemolysis drives susceptibility to bacterial infections and predicts poor outcome from sepsis. These detrimental effects are commonly considered to be a consequence of heme-iron serving as a nutrient for bacteria. We employed a Gram-negative sepsis model and found that elevated heme levels impaired the control of bacterial proliferation independently of heme-iron acquisition by pathogens. Heme strongly inhibited phagocytosis and the migration of human and mouse phagocytes by disrupting actin cytoskeletal dynamics via activation of the GTP-binding Rho family protein Cdc42 by the guanine nucleotide exchange factor DOCK8. A chemical screening approach revealed that quinine effectively prevented heme effects on the cytoskeleton, restored phagocytosis and improved survival in sepsis. These mechanistic insights provide potential therapeutic targets for patients with sepsis or hemolytic disorders.


Subject(s)
Gram-Negative Bacterial Infections/immunology , Guanine Nucleotide Exchange Factors/metabolism , Heme/metabolism , Hemolysis/immunology , Macrophages/immunology , Phagocytosis , Sepsis/immunology , Animals , Anti-Bacterial Agents/therapeutic use , Cytoskeleton/metabolism , Female , Gram-Negative Bacterial Infections/drug therapy , Guanine Nucleotide Exchange Factors/genetics , Heme Oxygenase-1/genetics , Hemolysis/drug effects , Humans , Immune Evasion , Macrophages/drug effects , Macrophages/microbiology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/drug effects , Quinine/therapeutic use , RAW 264.7 Cells , Sepsis/drug therapy , cdc42 GTP-Binding Protein/metabolism
4.
PLoS Pathog ; 17(4): e1009487, 2021 04.
Article in English | MEDLINE | ID: mdl-33905460

ABSTRACT

Lipocalin 2 (LCN2) is a secreted glycoprotein with roles in multiple biological processes. It contributes to host defense by interference with bacterial iron uptake and exerts immunomodulatory functions in various diseases. Here, we aimed to characterize the function of LCN2 in lung macrophages and dendritic cells (DCs) using Lcn2-/- mice. Transcriptome analysis revealed strong LCN2-related effects in CD103+ DCs during homeostasis, with differential regulation of antigen processing and presentation and antiviral immunity pathways. We next validated the relevance of LCN2 in a mouse model of influenza infection, wherein LCN2 protected from excessive weight loss and improved survival. LCN2-deficiency was associated with enlarged mediastinal lymph nodes and increased lung T cell numbers, indicating a dysregulated immune response to influenza infection. Depletion of CD8+ T cells equalized weight loss between WT and Lcn2-/- mice, proving that LCN2 protects from excessive disease morbidity by dampening CD8+ T cell responses. In vivo T cell chimerism and in vitro T cell proliferation assays indicated that improved antigen processing by CD103+ DCs, rather than T cell intrinsic effects of LCN2, contribute to the exacerbated T cell response. Considering the antibacterial potential of LCN2 and that commensal microbes can modulate antiviral immune responses, we speculated that LCN2 might cause the observed influenza phenotype via the microbiome. Comparing the lung and gut microbiome of WT and Lcn2-/- mice by 16S rRNA gene sequencing, we observed profound effects of LCN2 on gut microbial composition. Interestingly, antibiotic treatment or co-housing of WT and Lcn2-/- mice prior to influenza infection equalized lung CD8+ T cell counts, suggesting that the LCN2-related effects are mediated by the microbiome. In summary, our results highlight a novel regulatory function of LCN2 in the modulation of antiviral immunity.


Subject(s)
Influenza, Human/immunology , Lipocalin-2/metabolism , Microbiota/immunology , Transcriptome , Animals , Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Female , Gastrointestinal Microbiome , Homeostasis , Humans , Immunity , Influenza, Human/virology , Lipocalin-2/genetics , Lung/immunology , Lung/virology , Lymphocyte Activation , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Specific Pathogen-Free Organisms
5.
Am J Respir Cell Mol Biol ; 66(1): 64-75, 2022 01.
Article in English | MEDLINE | ID: mdl-34586974

ABSTRACT

Tissue-resident macrophages are of vital importance as they preserve tissue homeostasis in all mammalian organs. Nevertheless, appropriate cell culture models are still limited. Here, we propose a novel culture model to study and expand murine primary alveolar macrophages (AMs), the tissue-resident macrophages of the lung, in vitro over several months. By providing a combination of granulocyte-macrophage colony-stimulating factor, TGFß, and the PPARγ activator rosiglitazone, we maintain and expand mouse ex vivo cultured AMs (mexAMs) over several months. MexAMs maintain typical morphologic features and stably express primary AM surface markers throughout in vitro culture. They respond to microbial ligands and exhibit an AM-like transcriptional profile, including the expression of AM-specific transcription factors. Furthermore, when transferred into AM-deficient mice, mexAMs efficiently engraft in the lung and fulfill key macrophage functions, leading to a significantly reduced surfactant load in those mice. Altogether, mexAMs provide a novel, simple, and versatile tool to study AM behavior in homeostasis and disease settings.


Subject(s)
Macrophages, Alveolar/metabolism , Animals , Animals, Newborn , Cells, Cultured , Disease Models, Animal , Liver/metabolism , Lung/pathology , Lung/physiopathology , Macrophages, Alveolar/pathology , Mice, Inbred C57BL , Phenotype , Pulmonary Alveolar Proteinosis/metabolism , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Alveolar Proteinosis/physiopathology , Transcription, Genetic
6.
Eur Respir J ; 59(2)2022 02.
Article in English | MEDLINE | ID: mdl-34244315

ABSTRACT

RATIONALE: Lung transplantation is the ultimate treatment option for patients with end-stage respiratory diseases but bears the highest mortality rate among all solid organ transplantations due to chronic lung allograft dysfunction (CLAD). The mechanisms leading to CLAD remain elusive due to an insufficient understanding of the complex post-transplant adaptation processes. OBJECTIVES: To better understand these lung adaptation processes after transplantation and to investigate their association with future changes in allograft function. METHODS: We performed an exploratory cohort study of bronchoalveolar lavage samples from 78 lung recipients and donors. We analysed the alveolar microbiome using 16S rRNA sequencing, the cellular composition using flow cytometry, as well as metabolome and lipidome profiling. MEASUREMENTS AND MAIN RESULTS: We established distinct temporal dynamics for each of the analysed data sets. Comparing matched donor and recipient samples, we revealed that recipient-specific as well as environmental factors, rather than the donor microbiome, shape the long-term lung microbiome. We further discovered that the abundance of certain bacterial strains correlated with underlying lung diseases even after transplantation. A decline in forced expiratory volume during the first second (FEV1) is a major characteristic of lung allograft dysfunction in transplant recipients. By using a machine learning approach, we could accurately predict future changes in FEV1 from our multi-omics data, whereby microbial profiles showed a particularly high predictive power. CONCLUSION: Bronchoalveolar microbiome, cellular composition, metabolome and lipidome show specific temporal dynamics after lung transplantation. The lung microbiome can predict future changes in lung function with high precision.


Subject(s)
Lung Transplantation , Microbiota , Allografts , Cohort Studies , Humans , Lung , RNA, Ribosomal, 16S/genetics , Retrospective Studies
7.
Eur J Immunol ; 46(9): 2175-86, 2016 09.
Article in English | MEDLINE | ID: mdl-27312374

ABSTRACT

Protecting the integrity of the lung epithelial barrier is essential to ensure respiration and proper oxygenation in patients suffering from various types of lung inflammation. Type I interferon (IFN-I) has been associated with pulmonary epithelial barrier function, however, the mechanisms and involved cell types remain unknown. We aimed to investigate the importance of IFN-I with respect to its epithelial barrier strengthening function to better understand immune-modulating effects in the lung with potential medical implications. Using a mouse model of pneumococcal pneumonia, we revealed that IFN-I selectively protects alveolar epithelial type II cells (AECII) from inflammation-induced cell death. Mechanistically, signaling via the IFN-I receptor on AECII is sufficient to promote AECII survival. The net effects of IFN-I are barrier protection, together with diminished tissue damage, inflammation, and bacterial loads. Importantly, we found that the protective role of IFN-I can also apply to sterile acute lung injury, in which loss of IFN-I signaling leads to a significant reduction in barrier function caused by AECII cell death. Our data suggest that IFN-I is an important mediator in lung inflammation that plays a protective role by antagonizing inflammation-associated cell obstruction, thereby strengthening the integrity of the epithelial barrier.


Subject(s)
Alveolar Epithelial Cells/metabolism , Cell Survival , Interferon Type I/metabolism , Lung Injury/etiology , Lung Injury/metabolism , Pneumonia, Pneumococcal/etiology , Pneumonia, Pneumococcal/metabolism , Animals , Disease Models, Animal , Female , Immunomodulation , Lung Injury/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Mice, Knockout , Pneumonia, Pneumococcal/pathology , Receptor, Interferon alpha-beta/metabolism , Signal Transduction , Streptococcus pneumoniae
8.
PLoS Pathog ; 10(6): e1004167, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24945405

ABSTRACT

Phagocytosis and inflammation within the lungs is crucial for host defense during bacterial pneumonia. Triggering receptor expressed on myeloid cells (TREM)-2 was proposed to negatively regulate TLR-mediated responses and enhance phagocytosis by macrophages, but the role of TREM-2 in respiratory tract infections is unknown. Here, we established the presence of TREM-2 on alveolar macrophages (AM) and explored the function of TREM-2 in the innate immune response to pneumococcal infection in vivo. Unexpectedly, we found Trem-2(-/-) AM to display augmented bacterial phagocytosis in vitro and in vivo compared to WT AM. Mechanistically, we detected that in the absence of TREM-2, pulmonary macrophages selectively produced elevated complement component 1q (C1q) levels. We found that these increased C1q levels depended on peroxisome proliferator-activated receptor-δ (PPAR-δ) activity and were responsible for the enhanced phagocytosis of bacteria. Upon infection with S. pneumoniae, Trem-2(-/-) mice exhibited an augmented bacterial clearance from lungs, decreased bacteremia and improved survival compared to their WT counterparts. This work is the first to disclose a role for TREM-2 in clinically relevant respiratory tract infections and demonstrates a previously unknown link between TREM-2 and opsonin production within the lungs.


Subject(s)
Complement C1q/metabolism , Disease Models, Animal , Lung/immunology , Macrophages, Alveolar/immunology , Membrane Glycoproteins/metabolism , Pneumonia, Pneumococcal/immunology , Receptors, Immunologic/metabolism , Respiratory Mucosa/immunology , Animals , Apoptosis , Cell Line, Transformed , Cells, Cultured , Complement C1q/genetics , Cytokines/metabolism , Female , Lung/cytology , Lung/metabolism , Lung/pathology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Male , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , PPAR gamma/metabolism , Phagocytosis , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/pathology , Receptors, Immunologic/genetics , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Survival Analysis
9.
FASEB J ; 29(4): 1247-57, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25477281

ABSTRACT

During infections, TLR-mediated responses require tight regulation to allow for pathogen removal, while preventing overwhelming inflammation and immunopathology. The triggering receptor expressed on myeloid cells (TREM)-2 negatively regulates inflammation by macrophages and impacts on phagocytosis, but the function of endogenous TREM-2 during infections is poorly understood. We investigated TREM-2's role in regulating TLR4-mediated inflammation by studying wild-type and TREM-2(-/-) mice challenged with LPS and found TREM-2 to dampen early inflammation. Augmented early inflammation in TREM-2(-/-) animals was followed by an accelerated resolution and ultimately improved survival, associated with the induction of the negative regulator A20. Upon infection with Escherichia coli, the otherwise beneficial effect of an exaggerated early immune response in TREM-2(-/-) animals was counteracted by a 50% reduction in bacterial phagocytosis. In line with this, TREM-2(-/-) peritoneal macrophages (PMs) exhibited augmented inflammation following TLR4 stimulation, demonstrating the presence and negative regulatory functionality of TREM-2 on primary PMs. Significantly, we identified a high turnover rate because TREM-2 RNA is 25-fold down-regulated and the protein proteasomally degraded upon LPS encounter, thus ensuring a tightly regulated and versatile system that modulates inflammation. Our results illustrate TREM-2's effects on infection-triggered inflammation and identify TREM-2 as a potential target to prevent overwhelming inflammation while preserving antibacterial-effector functions.


Subject(s)
Gram-Negative Bacterial Infections/immunology , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Sepsis/immunology , Animals , Bacterial Load , Down-Regulation , Endotoxemia/etiology , Endotoxemia/immunology , Escherichia coli Infections/etiology , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Female , Gram-Negative Bacterial Infections/etiology , Gram-Negative Bacterial Infections/microbiology , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Macrophages, Peritoneal/immunology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritonitis/etiology , Peritonitis/immunology , Peritonitis/microbiology , Phagocytosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Sepsis/etiology , Toll-Like Receptor 4/metabolism
10.
J Immunol ; 190(11): 5640-8, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23610144

ABSTRACT

CD36 is a scavenger receptor that exhibits pleiotropic functions, including adhesion to thrombospondin, inhibition of angiogenesis, transport of long-chain fatty acids, and clearance of apoptotic cells. In addition, it has been implicated in the host immune response because it acts as a coreceptor for TLR2 and plays a role in Staphylococcus aureus infection. However, its role in other Gram-positive bacterial infections is unclear. In this study, using mice deficient in CD36, we sought to examine the role of CD36 in pneumococcal pneumonia, a major cause of morbidity and mortality worldwide. We show that CD36 is expressed on both alveolar macrophages and respiratory epithelial cells. Early in infection, CD36(-/-) mice have an exaggerated inflammatory response compared with wild-type littermate controls. In vitro studies using CD36(-/-) primary cells confirm the enhanced early inflammation in response to S. pneumoniae and its lipoteichoic acid, demonstrate that S. pneumoniae binds to cells via its phosphocholine residues, and suggest a role for CD36 in reducing inflammation induced by the phosphocholine residues of pneumococcal lipoteichoic acid. Later in infection, although CD36(-/-) mice exhibit impaired bacterial clearance, owing to a decreased capacity of CD36(-/-) macrophages to phagocytose S. pneumoniae, minor effects on mortality occur, in comparison with those in wild-type littermate control mice. These data show that CD36 contributes to the pulmonary host response during S. pneumoniae infection by virtue of its ability to act as a phagocytic receptor and as a modulator of the early innate immune response.


Subject(s)
CD36 Antigens/metabolism , Phagocytosis/immunology , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/metabolism , Streptococcus pneumoniae/immunology , Animals , CD36 Antigens/genetics , Disease Models, Animal , Female , Immunity, Innate , Inflammation/immunology , Inflammation/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Mice, Knockout , Phosphorylcholine/immunology , Pneumonia, Pneumococcal/genetics , Pneumonia, Pneumococcal/mortality , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Streptococcus pneumoniae/chemistry
11.
Eur J Pediatr ; 173(4): 469-76, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24221605

ABSTRACT

UNLABELLED: In a prospective surveillance study covering all pediatric wards in Austria, 308 cases of invasive pneumococcal disease (IPD) were reported in hospitalized children <5 years of age between 2002 and 2012. Incidence was 7.1 per 100,000 per year for IPD with a case fatality rate of 3 %, and 1.9 per 100,000 per year for pneumococcal meningitis with a case fatality rate of 9 %. At hospital discharge, 17 % of the children were not fully recovered and suffered from problems such as hearing or motor deficits. Persistent sequelae 6 months after hospital discharge were present in 13 % of the children, a finding that emphasizes the seriousness of IPD. From 2007 onwards, we observed a shift of pneumococcal serotypes from those covered by the heptavalent vaccine to serotypes consequently added to 10- and 13-valent vaccines, particularly regarding serotype 19A. Among antimicrobial resistances detected, macrolide resistance was predominant; however, between 2002 and 2012, we saw an overall decrease of resistance rates. CONCLUSION: Considering this change of serotypes and the high rate of permanent sequelae after IPD, our data show the importance of pediatric pneumococcal vaccination and the relevance of continuous monitoring of circulating serotypes. By the end of 2012, which was the first year of universal mass vaccination against pneumococcal disease in Austria, no change in the incidence of invasive pneumococcal disease was observed yet.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Pneumococcal Infections/epidemiology , Pneumococcal Vaccines/immunology , Streptococcus pneumoniae/isolation & purification , Austria/epidemiology , Child , Child, Hospitalized , Child, Preschool , Female , Humans , Incidence , Infant , Male , Pneumococcal Infections/drug therapy , Pneumococcal Infections/prevention & control , Population Surveillance , Prospective Studies , Streptococcus pneumoniae/immunology , Survival Rate
12.
Elife ; 112022 09 30.
Article in English | MEDLINE | ID: mdl-36178806

ABSTRACT

Sepsis is a life-threatening condition characterized by uncontrolled systemic inflammation and coagulation, leading to multiorgan failure. Therapeutic options to prevent sepsis-associated immunopathology remain scarce. Here, we established a mouse model of long-lasting disease tolerance during severe sepsis, manifested by diminished immunothrombosis and organ damage in spite of a high pathogen burden. We found that both neutrophils and B cells emerged as key regulators of tissue integrity. Enduring changes in the transcriptional profile of neutrophils include upregulated Cxcr4 expression in protected, tolerant hosts. Neutrophil Cxcr4 upregulation required the presence of B cells, suggesting that B cells promoted disease tolerance by improving tissue damage control via the suppression of neutrophils' tissue-damaging properties. Finally, therapeutic administration of a Cxcr4 agonist successfully promoted tissue damage control and prevented liver damage during sepsis. Our findings highlight the importance of a critical B-cell/neutrophil interaction during sepsis and establish neutrophil Cxcr4 activation as a potential means to promote disease tolerance during sepsis.


Subject(s)
Bacterial Infections , Sepsis , Animals , Bacterial Infections/metabolism , Disease Models, Animal , Mice , Multiple Organ Failure/metabolism , Multiple Organ Failure/pathology , Neutrophils/metabolism , Sepsis/metabolism
13.
Mucosal Immunol ; 15(5): 896-907, 2022 05.
Article in English | MEDLINE | ID: mdl-35856089

ABSTRACT

Environmental microbial triggers shape the development and functionality of the immune system. Alveolar macrophages (AMs), tissue-resident macrophages of the lungs, are in constant and direct contact with inhaled particles and microbes. Such exposures likely impact AM reactivity to subsequent challenges by immunological imprinting mechanisms referred to as trained immunity. Here, we investigated whether a ubiquitous microbial compound has the potential to induce AM training in vivo. We discovered that intranasal exposure to ambient amounts of lipopolysaccharide (LPS) induced a pronounced AM memory response, characterized by enhanced reactivity upon pneumococcal challenge. Exploring the mechanistic basis of AM training, we identified a critical role of type 1 interferon signaling and found that inhibition of fatty acid oxidation and glutaminolysis significantly attenuated the training effect. Notably, adoptive transfer of trained AMs resulted in increased bacterial loads and tissue damage upon subsequent pneumococcal infection. In contrast, intranasal pre-exposure to LPS promoted bacterial clearance, highlighting the complexity of stimulus-induced immune responses, which likely involve multiple cell types and may depend on the local immunological and metabolic environment. Collectively, our findings demonstrate the profound impact of ambient microbial exposure on pulmonary immune memory and reveal tissue-specific features of trained immunity.


Subject(s)
Interferon Type I , Macrophages, Alveolar , Interferon Type I/metabolism , Lipopolysaccharides , Lung , Signal Transduction
14.
Elife ; 112022 01 13.
Article in English | MEDLINE | ID: mdl-35023830

ABSTRACT

Despite tremendous progress in the understanding of COVID-19, mechanistic insight into immunological, disease-driving factors remains limited. We generated maVie16, a mouse-adapted SARS-CoV-2, by serial passaging of a human isolate. In silico modeling revealed how only three Spike mutations of maVie16 enhanced interaction with murine ACE2. maVie16 induced profound pathology in BALB/c and C57BL/6 mice, and the resulting mouse COVID-19 (mCOVID-19) replicated critical aspects of human disease, including early lymphopenia, pulmonary immune cell infiltration, pneumonia, and specific adaptive immunity. Inhibition of the proinflammatory cytokines IFNγ and TNF substantially reduced immunopathology. Importantly, genetic ACE2-deficiency completely prevented mCOVID-19 development. Finally, inhalation therapy with recombinant ACE2 fully protected mice from mCOVID-19, revealing a novel and efficient treatment. Thus, we here present maVie16 as a new tool to model COVID-19 for the discovery of new therapies and show that disease severity is determined by cytokine-driven immunopathology and critically dependent on ACE2 in vivo.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/virology , Interferon-gamma/pharmacology , SARS-CoV-2/pathogenicity , Adaptive Immunity/immunology , Animals , Disease Models, Animal , Interferon-gamma/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptidyl-Dipeptidase A/genetics , Spike Glycoprotein, Coronavirus/genetics
15.
Diabetes ; 70(9): 2042-2057, 2021 09.
Article in English | MEDLINE | ID: mdl-33627323

ABSTRACT

Obesity-induced white adipose tissue (WAT) hypertrophy is associated with elevated adipose tissue macrophage (ATM) content. Overexpression of the triggering receptor expressed on myeloid cells 2 (TREM2) reportedly increases adiposity, worsening health. Paradoxically, using insulin resistance, elevated fat mass, and hypercholesterolemia as hallmarks of unhealthy obesity, a recent report demonstrated that ATM-expressed TREM2 promoted health. Here, we identified that in mice, TREM2 deficiency aggravated diet-induced insulin resistance and hepatic steatosis independently of fat and cholesterol levels. Metabolomics linked TREM2 deficiency with elevated obesity-instigated serum ceramides that correlated with impaired insulin sensitivity. Remarkably, while inhibiting ceramide synthesis exerted no influences on TREM2-dependent ATM remodeling, inflammation, or lipid load, it restored insulin tolerance, reversing adipose hypertrophy and secondary hepatic steatosis of TREM2-deficient animals. Bone marrow transplantation experiments revealed unremarkable influences of immune cell-expressed TREM2 on health, instead demonstrating that WAT-intrinsic mechanisms impinging on sphingolipid metabolism dominate in the systemic protective effects of TREM2 on metabolic health.


Subject(s)
Adipose Tissue/metabolism , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Obesity/metabolism , Receptors, Immunologic/metabolism , Animals , Diet, High-Fat , Inflammation/metabolism , Insulin Resistance/physiology , Lipid Metabolism/physiology , Mice , Up-Regulation
16.
Sci Rep ; 7(1): 11289, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900132

ABSTRACT

Lipid metabolism is increasingly being appreciated to affect immunoregulation, inflammation and pathology. In this study we found that mice infected with lymphocytic choriomeningitis virus (LCMV) exhibit global perturbations of circulating serum lipids. Mice lacking the lipid-sensing surface receptor triggering receptor expressed on myeloid cells 2 (Trem2 -/-) were protected from LCMV-induced hepatitis and showed improved virus control despite comparable virus-specific T cell responses. Non-hematopoietic expression of TREM2 was found to be responsible for aggravated hepatitis, indicating a novel role for TREM2 in the non-myeloid compartment. These results suggest a link between virus-perturbed lipids and TREM2 that modulates liver pathogenesis upon viral infection. Targeted interventions of this immunoregulatory axis may ameliorate tissue pathology in hepatitis.


Subject(s)
Hepatitis/metabolism , Hepatitis/virology , Lipid Metabolism , Lymphocytic choriomeningitis virus/physiology , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Animals , Cytokines , Disease Models, Animal , Hepatitis/pathology , Membrane Glycoproteins/genetics , Metabolome , Metabolomics/methods , Mice , Mice, Knockout , Protein Aggregates , Receptors, Immunologic/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Viral Load
17.
Cell Rep ; 18(8): 1893-1905, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228256

ABSTRACT

From birth onward, the lungs are exposed to the external environment and therefore harbor a complex immunological milieu to protect this organ from damage and infection. We investigated the homeostatic role of the epithelium-derived alarmin interleukin-33 (IL-33) in newborn mice and discovered the immediate upregulation of IL-33 from the first day of life, closely followed by a wave of IL-13-producing type 2 innate lymphoid cells (ILC2s), which coincided with the appearance of alveolar macrophages (AMs) and their early polarization to an IL-13-dependent anti-inflammatory M2 phenotype. ILC2s contributed to lung quiescence in homeostasis by polarizing tissue resident AMs and induced an M2 phenotype in transplanted macrophage progenitors. ILC2s continued to maintain the M2 AM phenotype during adult life at the cost of a delayed response to Streptococcus pneumoniae infection in mice. These data highlight the homeostatic role of ILC2s in setting the activation threshold in the lung and underline their implications in anti-bacterial defenses.


Subject(s)
Animals, Newborn/immunology , Homeostasis/immunology , Immunity, Innate/immunology , Interleukin-13/immunology , Lung/immunology , Animals , Lymphocytes/immunology , Macrophages/immunology , Macrophages, Alveolar/immunology , Mice , Mice, Inbred C57BL , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , Up-Regulation/immunology
18.
J Clin Invest ; 123(7): 3014-24, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23934128

ABSTRACT

Clearance of invading pathogens is essential to preventing overwhelming inflammation and sepsis that are symptomatic of bacterial peritonitis. Macrophages participate in this innate immune response by engulfing and digesting pathogens, a process called phagocytosis. Oxidized phospholipids (OxPL) are danger-associated molecular patterns (DAMPs) generated in response to infection that can prevent the phagocytic clearance of bacteria. We investigated the mechanism underlying OxPL action in macrophages. Exposure to OxPL induced alterations in actin polymerization, resulting in spreading of peritoneal macrophages and diminished uptake of E. coli. Pharmacological and cell-based studies showed that an anchored pool of PKA mediates the effects of OxPL. Gene silencing approaches identified the A-kinase anchoring protein (AKAP) WAVE1 as an effector of OxPL action in vitro. Chimeric Wave1(-/-) mice survived significantly longer after infection with E. coli and OxPL treatment in vivo. Moreover, we found that endogenously generated OxPL in human peritoneal dialysis fluid from end-stage renal failure patients inhibited phagocytosis via WAVE1. Collectively, these data uncover an unanticipated role for WAVE1 as a critical modulator of the innate immune response to severe bacterial infections.


Subject(s)
Escherichia coli Infections/immunology , Macrophages, Peritoneal/immunology , Peritonitis/immunology , Phagocytosis , Phospholipids/physiology , Wiskott-Aldrich Syndrome Protein Family/metabolism , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Dimyristoylphosphatidylcholine/pharmacology , Enzyme Activation , Escherichia coli/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Humans , Immunity, Innate , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/therapy , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Peritoneal Dialysis , Peritonitis/metabolism , Peritonitis/microbiology , Phosphatidylcholines/pharmacology , Phosphatidylcholines/physiology , Wiskott-Aldrich Syndrome Protein Family/genetics
19.
J Clin Invest ; 123(8): 3363-72, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23863624

ABSTRACT

Macrophages play a key role in responding to pathogens and initiate an inflammatory response to combat microbe multiplication. Deactivation of macrophages facilitates resolution of the inflammatory response. Deactivated macrophages are characterized by an immunosuppressive phenotype, but the lack of unique markers that can reliably identify these cells explains the poorly defined biological role of this macrophage subset. We identified lipocalin 2 (LCN2) as both a marker of deactivated macrophages and a macrophage deactivator. We show that LCN2 attenuated the early inflammatory response and impaired bacterial clearance, leading to impaired survival of mice suffering from pneumococcal pneumonia. LCN2 induced IL-10 formation by macrophages, skewing macrophage polarization in a STAT3-dependent manner. Pulmonary LCN2 levels were tremendously elevated during bacterial pneumonia in humans, and high LCN2 levels were indicative of a detrimental outcome from pneumonia with Gram-positive bacteria. Our data emphasize the importance of macrophage deactivation for the outcome of pneumococcal infections and highlight the role of LCN2 and IL-10 as determinants of macrophage performance in the respiratory tract.


Subject(s)
Acute-Phase Proteins/immunology , Lipocalins/immunology , Macrophages, Alveolar/immunology , Oncogene Proteins/immunology , Pneumonia, Pneumococcal/immunology , Proto-Oncogene Proteins/immunology , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Adult , Aged , Animals , Female , Humans , Immune Tolerance , Interleukin-10/biosynthesis , Lipocalin-2 , Lipocalins/genetics , Lung/immunology , Macrophage Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Pneumonia, Pneumococcal/etiology , Transplantation Chimera/immunology
SELECTION OF CITATIONS
SEARCH DETAIL