Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Cutan Pathol ; 50(6): 544-551, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36562598

ABSTRACT

INTRODUCTION: The implications of infiltrative compared to non-infiltrative growth of cutaneous basal cell carcinoma (BCC) on the tumor stroma and immune cell landscape are unknown. This is of clinical importance, because infiltrative BCCs, in contrast to other BCC subtypes, are more likely to relapse after surgery and radiotherapy. MATERIALS AND METHODS: This descriptive cross-sectional study analyzed 38 BCCs collected from 2018 to 2021. In the first cohort (n = 28), immune cells were characterized by immunohistochemistry and multiplex immunofluorescence staining for CD3, CD8, CD68, Foxp3, and α-SMA protein expression. In the second cohort (n = 10) with matched characteristics (age, sex, location, and BCC subtype), inflammatory parameters, including TGF-ß1, TGF-ß2, ACTA2, IL-10, IL-12A, and Foxp3, were quantified via RT-qPCR after isolating mRNA from BCC tissue samples and perilesional skin. RESULTS: Infiltrative BCCs showed significantly increased levels of α-SMA expression in fibroblasts (p = 0.0001) and higher levels of Foxp3+ (p = 0.0023) and CD3+ (p = 0.0443) T-cells compared to non-infiltrative BCCs. CD3+ (p = 0.0171) and regulatory T-cells (p = 0.0026) were significantly increased in α-SMA-positive tumor stroma, whereas CD8+ T-cells (p = 0.1329) and CD68+ myeloid cells (p = 0.2337) were not affected. TGF-ß1 and TGF-ß2 correlated significantly with ACTA2/α-SMA mRNA expression (p = 0.020, p = 0.005). CONCLUSION: Infiltrative growth of BCCs shows a myofibroblastic stroma differentiation and is accompanied by an immunosuppressive tumor microenvironment.


Subject(s)
Carcinoma, Basal Cell , Skin Neoplasms , Humans , Skin Neoplasms/pathology , Transforming Growth Factor beta1 , Transforming Growth Factor beta2 , T-Lymphocytes, Regulatory/pathology , Cross-Sectional Studies , Myofibroblasts/pathology , Neoplasm Recurrence, Local , Carcinoma, Basal Cell/pathology , Cell Differentiation , Forkhead Transcription Factors , Tumor Microenvironment
2.
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
3.
Cell Rep ; 38(8): 110420, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35196494

ABSTRACT

Dendritic cells (DCs) induce peripheral T cell tolerance, but cell-intrinsic signaling cascades governing their stable tolerogenesis remain poorly defined. Janus Kinase 1 (JAK1) transduces cytokine-receptor signaling, and JAK inhibitors (Jakinibs), including JAK1-specific filgotinib, break inflammatory cycles in autoimmunity. Here, we report in heterogeneous DC populations of multiple secondary lymphoid organs that JAK1 promotes peripheral T cell tolerance during experimental autoimmune encephalomyelitis (EAE). Mice harboring DC-specific JAK1 deletion exhibit elevated peripheral CD4+ T cell expansion, less regulatory T cells (Tregs), and worse EAE outcomes, whereas adoptive DC transfer ameliorates EAE pathogenesis by inducing peripheral Tregs, programmed cell death ligand 1 (PD-L1) dependently. This tolerogenic program is substantially reduced upon the transfer of JAK1-deficient DCs. DC-intrinsic IFN-γ-JAK1-STAT1 signaling induces PD-L1, which is required for DCs to convert CD4+ T cells into Tregs in vitro and attenuated upon JAK1 deficiency and filgotinib treatment. Thus, DC-intrinsic JAK1 promotes peripheral tolerance, suggesting potential unwarranted DC-mediated effects of Jakinibs in autoimmune diseases.


Subject(s)
B7-H1 Antigen , Encephalomyelitis, Autoimmune, Experimental , Janus Kinase 1 , T-Lymphocytes, Regulatory , Animals , Autoimmunity , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Dendritic Cells/metabolism , Immune Tolerance , Janus Kinase 1/immunology , Janus Kinase 1/metabolism , Mice , Peripheral Tolerance
4.
J Invest Dermatol ; 142(6): 1737-1748.e5, 2022 06.
Article in English | MEDLINE | ID: mdl-34922948

ABSTRACT

Patients suffering from large scars such as burn victims not only encounter aesthetic challenges but also ongoing itching or pain that substantially deteriorates their quality of life. Skin appendages such as hair follicles rarely regenerate within the healing wound. Because they are crucial for skin homeostasis and the lack thereof constitutes one of the main limitations to scarless wound healing, their regeneration represents a major objective for regenerative medicine. Fibroblasts, the main resident cell type of the skin dermis, mediate embryonic hair follicle morphogenesis and are particularly involved in wound healing because they orchestrate extracellular matrix remodeling and collagen deposition in the wound bed. Importantly, dermal fibroblasts originate from two distinct developmental lineages with unique functions that differently mediate the response to epidermal signals such as Hedgehog signaling. In this study, we show that Hedgehog signaling in the reticular fibroblast lineage promotes the initial phase of wound repair, possibly by modulating angiogenesis and fibroblast proliferation, whereas Hedgehog signaling in papillary fibroblasts is essential to induce de novo hair follicle formation within the healing wound.


Subject(s)
Hair Follicle , Hedgehog Proteins , Regeneration , Signal Transduction , Wound Healing , Dermis/metabolism , Fibroblasts/metabolism , Hair Follicle/growth & development , Hedgehog Proteins/physiology , Humans , Quality of Life , Regeneration/physiology , Wound Healing/physiology
5.
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
6.
Nat Metab ; 2(12): 1427-1442, 2020 12.
Article in English | MEDLINE | ID: mdl-33199895

ABSTRACT

Adipose tissue macrophages (ATMs) display tremendous heterogeneity depending on signals in their local microenvironment and contribute to the pathogenesis of obesity. The phosphoinositide 3-kinase (PI3K) signalling pathway, antagonized by the phosphatase and tensin homologue (PTEN), is important for metabolic responses to obesity. We hypothesized that fluctuations in macrophage-intrinsic PI3K activity via PTEN could alter the trajectory of metabolic disease by driving distinct ATM populations. Using mice harbouring macrophage-specific PTEN deletion or bone marrow chimeras carrying additional PTEN copies, we demonstrate that sustained PI3K activity in macrophages preserves metabolic health in obesity by preventing lipotoxicity. Myeloid PI3K signalling promotes a beneficial ATM population characterized by lipid uptake, catabolism and high expression of the scavenger macrophage receptor with collagenous structure (MARCO). Dual MARCO and myeloid PTEN deficiencies prevent the generation of lipid-buffering ATMs, reversing the beneficial actions of elevated myeloid PI3K activity in metabolic disease. Thus, macrophage-intrinsic PI3K signalling boosts metabolic health by driving ATM programmes associated with MARCO-dependent lipid uptake.


Subject(s)
Adipose Tissue/metabolism , Lipid Metabolism/genetics , Macrophages/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Bone Marrow Transplantation , Cell Differentiation , Chimera , Glucose Tolerance Test , Lipidomics , Macrophages/pathology , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Obesity/metabolism , Obesity/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Receptors, Immunologic/genetics , Signal Transduction/genetics
7.
J Vis Exp ; (147)2019 05 07.
Article in English | MEDLINE | ID: mdl-31132050

ABSTRACT

Fibroblasts are a highly heterogeneous cell population implicated in the pathogenesis of many human diseases. In human skin dermis, fibroblasts have traditionally been attributed to the superficial papillary or lower reticular dermis according to their histological localization. In mouse dermis, papillary and reticular fibroblasts originate from two different lineages with diverging functions regarding physiological and pathological processes and a distinct cell surface marker expression profile by which they can be distinguished. Importantly, evidence from explant cultures from superficial and lower dermal layers suggest that at least two functionally distinct dermal fibroblasts lineages exist in human skin dermis as well. However, unlike for mouse skin, cell surface markers enabling the discrimination of different fibroblast subsets have not yet been established for human skin. We developed a novel protocol for the isolation of human papillary and reticular fibroblast populations via fluorescence-activated cell sorting (FACS) using the two cell surface markers Fibroblast Activation Protein (FAP) and Thymocyte antigen 1 (Thy1)/CD90. This method enables the isolation of pure fibroblast subsets without in vitro manipulation, which was shown to affect gene expression, thus permitting accurate functional analysis of human dermal fibroblast subsets in regard to tissue homeostasis or disease pathology.


Subject(s)
Fibroblasts/metabolism , Flow Cytometry/methods , Skin/metabolism , Cells, Cultured , Humans , Skin/cytology
8.
Gut ; 68(3): 533-546, 2019 03.
Article in English | MEDLINE | ID: mdl-29374630

ABSTRACT

OBJECTIVE: Liver injury impacts hepatic inflammation in part via Toll-like receptor (TLR) signalling. Triggering receptor expressed on myeloid cells 2 (TREM-2) modulates TLR4-mediated inflammation in bone marrow (BM)-derived macrophages but its function in liver injury is unknown. Here we hypothesised that the anti-inflammatory effects of TREM-2 on TLR signalling may limit hepatic injury. DESIGN: TREM-2 expression was analysed in livers of humans with various forms of liver injury compared with control individuals. Acute and chronic liver injury models were performed in wild type and Trem-2-/- mice. Primary liver cells from both genotypes of mice were isolated for in vitro experiments. RESULTS: TREM-2 was expressed on non-parenchymal hepatic cells and induced during liver injury in mice and man. Mice lacking TREM-2 exhibited heightened liver damage and inflammation during acute and repetitive carbon tetrachloride and acetaminophen (APAP) intoxication, the latter of which TREM-2 deficiency was remarkably associated with worsened survival. Liver damage in Trem-2-/- mice following chronic injury and APAP challenge was associated with elevated hepatic lipid peroxidation and macrophage content. BM transplantation experiments and cellular reactive oxygen species assays revealed effects of TREM-2 in the context of chronic injury depended on both immune and resident TREM-2 expression. Consistent with effects of TREM-2 on inflammation-associated injury, primary hepatic macrophages and hepatic stellate cells lacking TREM-2 exhibited augmented TLR4-driven proinflammatory responses. CONCLUSION: Our data indicate that by acting as a natural brake on inflammation during hepatocellular injury, TREM-2 is a critical regulator of diverse types of hepatotoxic injury.


Subject(s)
Liver Cirrhosis/metabolism , Liver/metabolism , Membrane Glycoproteins/physiology , Receptors, Immunologic/physiology , Acetaminophen , Aged , Animals , Carbon Tetrachloride , Case-Control Studies , Female , Hematopoietic Stem Cells/metabolism , Hepatocytes/metabolism , Humans , Inflammation Mediators/metabolism , Kupffer Cells/metabolism , Lipid Peroxidation/physiology , Liver Cirrhosis/etiology , Liver Cirrhosis/immunology , Liver Cirrhosis, Experimental/immunology , Liver Cirrhosis, Experimental/metabolism , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, Knockout , Middle Aged , Reactive Oxygen Species/metabolism , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Toll-Like Receptor 4/physiology , Up-Regulation/physiology
9.
J Invest Dermatol ; 139(2): 342-351, 2019 02.
Article in English | MEDLINE | ID: mdl-30179601

ABSTRACT

Human skin dermis is composed of the superficial papillary dermis and the reticular dermis in the lower layers, which can easily be distinguished histologically. In vitro analyses of fibroblasts from explant cultures from superficial and lower dermal layers suggest that human skin comprises at least two fibroblast lineages with distinct morphology, expression profiles, and functions. However, while for mouse skin cell surface markers have been identified, allowing the isolation of pure populations of one lineage or the other via FACS, this has not been achieved for human skin fibroblasts. We have now discovered two cell surface markers that discriminate between papillary and reticular fibroblasts. While FAP+CD90- cells display increased proliferative potential, express PDPN and NTN1, and cannot be differentiated into adipocytes, FAP-CD90+ fibroblasts express high levels of ACTA2, MGP, PPARγ, and CD36 and readily undergo adipogenic differentiation, a hallmark of reticular fibroblasts. Flow cytometric analysis of fibroblasts isolated from superficial and lower layers of human dermis showed that FAP+CD90- cells are enriched in the papillary dermis. Altogether, functional analysis and expression profiling confirms that FAP+CD90- cells represent papillary fibroblasts, whereas FAP-CD90+ fibroblasts derive from the reticular lineage. Although papillary and reticular fibroblasts are enriched in the upper or lower dermis, respectively, they are not spatially restricted, and the microenvironment seems to affect their function.


Subject(s)
Cell Differentiation , Dermis/cytology , Fibroblasts/physiology , Adipocytes/physiology , Adult , Biomarkers/metabolism , Cell Separation , Cells, Cultured , Endopeptidases , Female , Flow Cytometry , Gelatinases/metabolism , Humans , Male , Membrane Proteins/metabolism , Middle Aged , Primary Cell Culture , Serine Endopeptidases/metabolism , Thy-1 Antigens/metabolism
11.
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
12.
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
13.
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
14.
EMBO Mol Med ; 6(10): 1312-27, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25216727

ABSTRACT

Several subtypes of APCs are found in psoriasis patients, but their involvement in disease pathogenesis is poorly understood. Here, we investigated the contribution of Langerhans cells (LCs) and plasmacytoid DCs (pDCs) in psoriasis. In human psoriatic lesions and in a psoriasis mouse model (DKO* mice), LCs are severely reduced, whereas pDCs are increased. Depletion of pDCs in DKO* mice prior to psoriasis induction resulted in a milder phenotype, whereas depletion during active disease had no effect. In contrast, while depletion of Langerin-expressing APCs before disease onset had no effect, depletion from diseased mice aggravated psoriasis symptoms. Disease aggravation was due to the absence of LCs, but not other Langerin-expressing APCs. LCs derived from DKO* mice produced increased IL-10 levels, suggesting an immunosuppressive function. Moreover, IL-23 production was high in psoriatic mice and further increased in the absence of LCs. Conversely, pDC depletion resulted in reduced IL-23 production, and therapeutic inhibition of IL-23R signaling ameliorated disease symptoms. Therefore, LCs have an anti-inflammatory role during active psoriatic disease, while pDCs exert an instigatory function during disease initiation.


Subject(s)
Antigen-Presenting Cells/immunology , Dendritic Cells/immunology , Langerhans Cells/immunology , Psoriasis/immunology , Adjuvants, Immunologic/pharmacology , Aminoquinolines/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigen-Presenting Cells/metabolism , Bone Marrow Transplantation , Dendritic Cells/metabolism , Disease Progression , Flow Cytometry , Humans , Imiquimod , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-23/immunology , Interleukin-23/metabolism , Langerhans Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Proto-Oncogene Proteins c-jun/deficiency , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/immunology , Psoriasis/genetics , Psoriasis/prevention & control , Receptors, Interleukin/immunology , Receptors, Interleukin/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Skin/drug effects , Skin/immunology , Skin/pathology , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/immunology
15.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...