Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Publication year range
1.
Ann Rheum Dis ; 83(1): 72-87, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-37775153

ABSTRACT

OBJECTIVES: To investigate the effect of the L-arginine metabolism on arthritis and inflammation-mediated bone loss. METHODS: L-arginine was applied to three arthritis models (collagen-induced arthritis, serum-induced arthritis and human TNF transgenic mice). Inflammation was assessed clinically and histologically, while bone changes were quantified by µCT and histomorphometry. In vitro, effects of L-arginine on osteoclast differentiation were analysed by RNA-seq and mass spectrometry (MS). Seahorse, Single Cell ENergetIc metabolism by profilIng Translation inHibition and transmission electron microscopy were used for detecting metabolic changes in osteoclasts. Moreover, arginine-associated metabolites were measured in the serum of rheumatoid arthritis (RA) and pre-RA patients. RESULTS: L-arginine inhibited arthritis and bone loss in all three models and directly blocked TNFα-induced murine and human osteoclastogenesis. RNA-seq and MS analyses indicated that L-arginine switched glycolysis to oxidative phosphorylation in inflammatory osteoclasts leading to increased ATP production, purine metabolism and elevated inosine and hypoxanthine levels. Adenosine deaminase inhibitors blocking inosine and hypoxanthine production abolished the inhibition of L-arginine on osteoclastogenesis in vitro and in vivo. Altered arginine levels were also found in RA and pre-RA patients. CONCLUSION: Our study demonstrated that L-arginine ameliorates arthritis and bone erosion through metabolic reprogramming and perturbation of purine metabolism in osteoclasts.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Bone Resorption , Humans , Mice , Animals , Osteoclasts , Arthritis, Rheumatoid/pathology , Arthritis, Experimental/pathology , Inflammation/metabolism , Mice, Transgenic , Arginine/pharmacology , Inosine/metabolism , Inosine/pharmacology , Hypoxanthines/metabolism , Hypoxanthines/pharmacology , Purines/pharmacology
2.
J Immunol ; 205(2): 323-328, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32540999

ABSTRACT

TNF blockade is a successful treatment for human autoimmune disorders like rheumatoid arthritis and inflammatory bowel disease yet increases susceptibility to tuberculosis and other infections. The C-type lectin receptors (CLR) MINCLE, MCL, and DECTIN-2 are expressed on myeloid cells and sense mycobacterial cell wall glycolipids. In this study, we show that TNF is sufficient to upregulate MINCLE, MCL, and DECTIN-2 in macrophages. TNF signaling through TNFR1 p55 was required for upregulation of these CLR and for cytokine secretion in macrophages stimulated with the MINCLE ligand trehalose-6,6-dibehenate or infected with Mycobacterium bovis bacillus Calmette-Guérin. The Th17 response to immunization with the MINCLE-dependent adjuvant trehalose-6,6-dibehenate was specifically abrogated in TNF-deficient mice and strongly attenuated by TNF blockade with etanercept. Together, interference with production or signaling of TNF antagonized the expression of DECTIN-2 family CLR, thwarting vaccine responses and possibly increasing infection risk.


Subject(s)
Lectins, C-Type/metabolism , Mycobacterium bovis/physiology , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Th17 Cells/immunology , Tuberculosis/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , Etanercept/administration & dosage , Lectins, C-Type/genetics , Macrophage Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Immunologic/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Sugar Phosphates/metabolism , Trehalose/analogs & derivatives , Trehalose/metabolism , Tuberculosis/veterinary , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics
3.
J Immunol ; 205(6): 1580-1592, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32796022

ABSTRACT

Mycobacteria survive in macrophages despite triggering pattern recognition receptors and T cell-derived IFN-γ production. Mycobacterial cord factor trehalose-6,6-dimycolate (TDM) binds the C-type lectin receptor MINCLE and induces inflammatory gene expression. However, the impact of TDM on IFN-γ-induced macrophage activation is not known. In this study, we have investigated the cross-regulation of the mouse macrophage transcriptome by IFN-γ and by TDM or its synthetic analogue trehalose-6,6-dibehenate (TDB). As expected, IFN-γ induced genes involved in Ag presentation and antimicrobial defense. Transcriptional programs induced by TDM and TDB were highly similar but clearly distinct from the response to IFN-γ. The glycolipids enhanced expression of a subset of IFN-γ-induced genes associated with inflammation. In contrast, TDM/TDB exerted delayed inhibition of IFN-γ-induced genes, including pattern recognition receptors, MHC class II genes, and IFN-γ-induced GTPases, with antimicrobial function. TDM downregulated MHC class II cell surface expression and impaired T cell activation by peptide-pulsed macrophages. Inhibition of the IFN-γ-induced GTPase GBP1 occurred at the level of transcription by a partially MINCLE-dependent mechanism that may target IRF1 activity. Although activation of STAT1 was unaltered, deletion of Socs1 relieved inhibition of GBP1 expression by TDM. Nonnuclear Socs1 was sufficient for inhibition, suggesting a noncanonical, cytoplasmic mechanism. Taken together, unbiased analysis of transcriptional reprogramming revealed a significant degree of negative regulation of IFN-γ-induced Ag presentation and antimicrobial gene expression by the mycobacterial cord factor that may contribute to mycobacterial persistence.


Subject(s)
Cord Factors/metabolism , GTP-Binding Proteins/metabolism , Inflammation/microbiology , Lectins, C-Type/metabolism , Macrophages/physiology , Membrane Proteins/metabolism , Mycobacterium tuberculosis/physiology , Tuberculosis/microbiology , Animals , Antigen Presentation , Cells, Cultured , GTP-Binding Proteins/genetics , Gene Expression Profiling , Humans , Inflammation/immunology , Interferon-gamma/metabolism , Lectins, C-Type/genetics , Macrophage Activation , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Tuberculosis/immunology
4.
J Immunol ; 202(5): 1453-1464, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30665936

ABSTRACT

Arginase (Arg) 1 is expressed by hematopoietic (e.g., macrophages) and nonhematopoietic cells (e.g., endothelial cells) and converts l-arginine into ornithine and urea. The enzyme is implicated in tissue repair but also antagonizes the production of NO by type 2 NO synthase in myeloid cells and thereby impedes the control of intracellular parasites such as Leishmania major In this study, we tested whether Arg1 is required for spontaneous healing of acute cutaneous leishmaniasis in C57BL/6 mice and for lifelong parasite persistence in draining lymph nodes (dLNs) of healed mice. In vitro, bone marrow-derived macrophages and lymphoid endothelial cells readily expressed Arg1 in response to IL-4 and/or IL-13, whereas skin or dLN fibroblasts failed to do so, even during hypoxia. In vivo, Arg1 was found in skin lesions and, to a much lower extent, also in dLNs of acutely infected C57BL/6 mice but became undetectable at both sites after healing. Deletion of Arg1 in hematopoietic and endothelial cells using Tie2Cre+/-Arg1fl/fl C57BL/6 mice abolished the expression of Arg1 in skin lesions and dLNs but did not affect development and resolution of skin lesions, parasite burden, NO production, or host cell tropism of L. major during the acute or persistent phase of infection. Similar to wild-type controls, parasites persisting in Arg1-deficient mice favored NO synthase 2-negative areas and mainly resided in myeloid cells and fibroblasts. We conclude that Arg1 expression by hematopoietic and endothelial cells is completely dispensable for clinical resolution of cutaneous leishmaniasis and for long-term persistence of L. major.


Subject(s)
Arginase/metabolism , Leishmania major/metabolism , Leishmaniasis, Cutaneous/metabolism , Animals , Arginase/genetics , Cells, Cultured , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Nat Immunol ; 9(12): 1399-406, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18978793

ABSTRACT

Toll-like receptor (TLR) signaling in macrophages is required for antipathogen responses, including the biosynthesis of nitric oxide from arginine, and is essential for immunity to Mycobacterium tuberculosis, Toxoplasma gondii and other intracellular pathogens. Here we report a 'loophole' in the TLR pathway that is advantageous to these pathogens. Intracellular pathogens induced expression of the arginine hydrolytic enzyme arginase 1 (Arg1) in mouse macrophages through the TLR pathway. In contrast to diseases dominated by T helper type 2 responses in which Arg1 expression is greatly increased by interleukin 4 and 13 signaling through the transcription factor STAT6, TLR-mediated Arg1 induction was independent of the STAT6 pathway. Specific elimination of Arg1 in macrophages favored host survival during T. gondii infection and decreased lung bacterial load during tuberculosis infection.


Subject(s)
Arginase/immunology , Bacterial Infections/immunology , Macrophages/immunology , Macrophages/microbiology , Toll-Like Receptors/immunology , Animals , Arginase/metabolism , CCAAT-Enhancer-Binding Protein-beta/immunology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Immunoblotting , Immunohistochemistry , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , STAT6 Transcription Factor/immunology , STAT6 Transcription Factor/metabolism , Toll-Like Receptors/metabolism
6.
Infect Immun ; 85(8)2017 08.
Article in English | MEDLINE | ID: mdl-28507071

ABSTRACT

Similar to other intracellular pathogens, Leishmania parasites are known to evade the antimicrobial effector functions of host immune cells. To date, however, only a few virulence factors have been described for Leishmania major, one of the causative agents of cutaneous leishmaniasis. Here, we have characterized the expression and function of an L. major phosphatase, which we termed LmPRL-1. This enzyme shows a strong structural similarity to the human phosphatases of regenerating liver (PRL-1, -2, and -3) that regulate the proliferation, differentiation, and motility of cells. The biochemical characterization of the L. major phosphatase revealed that the enzyme is redox sensitive. When analyzing the subcellular localization of LmPRL-1 in promastigotes, amastigotes, and infected macrophages, we found that the phosphatase was predominantly expressed and secreted by promastigotes via the exosome route. Finally, we observed that ectopic expression of LmPRL-1 in L. major led to an increased number of parasites in macrophages. From these data, we conclude that the L. major phosphatase LmPRL-1 contributes to the intracellular survival of the parasites in macrophages.


Subject(s)
Exosomes/metabolism , Leishmania major/enzymology , Macrophages/parasitology , Protein Tyrosine Phosphatases/metabolism , Animals , Biological Transport , Cell Cycle Proteins/chemistry , Humans , Kinetics , Leishmania major/genetics , Membrane Proteins/chemistry , Mice , Neoplasm Proteins/chemistry , Oxidation-Reduction , Phylogeny , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics , Virulence , Virulence Factors
7.
Eur J Immunol ; 46(1): 114-21, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26449668

ABSTRACT

The genus leishmania comprises different protozoan parasites which are causative agents of muco-cutaneous and systemic, potentially lethal diseases. After infection with the species Leishmania major, resistant mice expand Th1 cells which stimulate macrophages for Leishmania destruction. In contrast, susceptible mice generate Th2 cells which deactivate macrophages, leading to systemic spread of the pathogens. Th-cell differentiation is determined within the first days, and Th2 cell differentiation requires IL-4, whereby the initial IL-4 source is often unknown. Mast cells are potential sources of IL-4, and hence their role in murine leishmaniasis has previously been studied in mast cell-deficient Kit mutant mice, although these mice display immunological phenotypes beyond mast cell deficiency. We therefore readdressed this question by infecting Kit-independent mast cell-deficient mice that are Th1 (C57BL/6 Cpa(Cre) ) or Th2 (BALB/c Cpa(Cre) ) prone with L. major. Using different parasite doses and intra- or subcutaneous infection routes, the results demonstrate no role of mast cells on lesion size development, parasite load, immune cell phenotypes expanding in draining lymph nodes, and cytokine production during murine cutaneous leishmaniasis. Thus, other cell types such as ILCs or T cells have to be considered as primary source of Th2-driving IL-4.


Subject(s)
Leishmaniasis, Cutaneous/immunology , Mast Cells/immunology , Th2 Cells/immunology , Animals , Cell Differentiation/immunology , Disease Models, Animal , Disease Susceptibility , Leishmania major , Leishmaniasis, Cutaneous/parasitology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Parasite Load
8.
Eur J Immunol ; 45(10): 2821-33, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26138432

ABSTRACT

DCs are professional APCs playing a crucial role in the initiation of T-cell responses to combat infection. However, systemic bacterial infection with various pathogens leads to DC-depletion in humans and mice. The mechanisms of pathogen-induced DC-depletion remain poorly understood. Previously, we showed that mice infected with Yersinia enterocolitica (Ye) had impaired de novo DC-development, one reason for DC-depletion. Here, we extend these studies to gain insight into the molecular mechanisms of DC-depletion and the impact of different bacteria on DC-development. We show that the number of bone marrow (BM) hematopoietic progenitors committed to the DC lineage is reduced following systemic infection with different Gram-positive and Gram-negative bacteria. This is associated with a TLR4- and IFN-γ-signaling dependent increase of committed monocyte progenitors in the BM and mature monocytes in the spleen upon Ye-infection. Adoptive transfer experiments revealed that infection-induced monopoiesis occurs at the expense of DC-development. Our data provide evidence for a general response of hematopoietic progenitors upon systemic bacterial infections to enhance monocyte production, thereby increasing the availability of innate immune cells for pathogen control, whereas impaired DC-development leads to DC-depletion, possibly driving transient immunosuppression in bacterial sepsis.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Immunity, Innate , Myelopoiesis/immunology , Yersinia Infections/immunology , Yersinia enterocolitica/immunology , Animals , Dendritic Cells/pathology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Interferon-gamma/immunology , Mice , Mice, Knockout , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Yersinia Infections/pathology
9.
BMC Infect Dis ; 14: 619, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25420793

ABSTRACT

BACKGROUND: A previously published proof of principle phase IIa trial with 113 patients from Kabul showed that bipolar high-frequency (HF) electro-cauterization (EC) of cutaneous leishmaniasis (CL) ulcers and subsequent moist wound treatment (MWT) closed 85% of all Leishmania (L.) tropica lesions within 60 days. METHODS: A three-armed phase IIb, randomized and controlled clinical trial was performed in Mazar-e-Sharif. L. tropica- or L. major-infected CL patients received intradermal sodium stibogluconate (SSG) (Group I); HF-EC followed by MWT with 0.045% DAC N-055 (Group II); or MWT with 0.045% DAC N-055 in basic crème alone (Group III). The primary outcome was complete epithelialisation before day 75 after treatment start. RESULTS: 87 patients enrolled in the trial were randomized into group I (n = 24), II (n = 32) and III (n = 31). The per-protocol analysis of 69 (79%) patients revealed complete epithelialisation before day 75 in 15 (of 23; 65%) patients of Group I, in 23 (of 23; 100%) patients of Group II, and in 20 (of 23; 87%) patients of Group III (p = 0.004, Fisher's Exact Test). In the per-protocol analysis, wound closure times were significantly different between all regimens in a pair-wise comparison (p = 0.000039, Log-Rank (Mantel-Cox) test). In the intention-to-treat analysis wound survival times in Group II were significantly different from those in Group I (p = 0.000040, Log-Rank (Mantel-Cox) test). Re-ulcerations occurred in four (17%), three (13%) and seven (30%) patients of Group I, II or III, respectively (p = 0.312, Pearson Chi-Square Test). CONCLUSIONS: Treatment of CL ulcers with bipolar HF-EC followed by MWT with 0.045% DAC N-055 or with DAC N-055 alone showed shorter wound closure times than with the standard SSG therapy. The results merit further exploration in larger trials in the light of our current knowledge of in vitro and in vivo activities of chlorite. Clinicaltrials.gov ID: NCT00996463. Registered: 15th October 2009.


Subject(s)
Antimony Sodium Gluconate/therapeutic use , Antiprotozoal Agents/therapeutic use , Chlorides/therapeutic use , Electrocoagulation/methods , Leishmaniasis, Cutaneous/therapy , Skin Ulcer/therapy , Adult , Afghanistan , Bandages , Female , Humans , Injections, Intralesional , Leishmaniasis, Cutaneous/complications , Male , Skin Ulcer/etiology , Time Factors , Treatment Outcome , Wound Healing , Young Adult
10.
J Exp Med ; 204(4): 893-906, 2007 Apr 16.
Article in English | MEDLINE | ID: mdl-17389237

ABSTRACT

Natural killer (NK) cells are sentinel components of the innate response to pathogens, but the cell types, pathogen recognition receptors, and cytokines required for their activation in vivo are poorly defined. Here, we investigated the role of plasmacytoid dendritic cells (pDCs), myeloid DCs (mDCs), Toll-like receptors (TLRs), and of NK cell stimulatory cytokines for the induction of an NK cell response to the protozoan parasite Leishmania infantum. In vitro, pDCs did not endocytose Leishmania promastigotes but nevertheless released interferon (IFN)-alpha/beta and interleukin (IL)-12 in a TLR9-dependent manner. mDCs rapidly internalized Leishmania and, in the presence of TLR9, produced IL-12, but not IFN-alpha/beta. Depletion of pDCs did not impair the activation of NK cells in L. infantum-infected mice. In contrast, L. infantum-induced NK cell cytotoxicity and IFN-gamma production were abolished in mDC-depleted mice. The same phenotype was observed in TLR9(-/-) mice, which lacked IL-12 expression by mDCs, and in IL-12(-/-) mice, whereas IFN-alpha/beta receptor(-/-) mice showed only a minor reduction of NK cell IFN-gamma expression. This study provides the first direct evidence that mDCs are essential for eliciting NK cell cytotoxicity and IFN-gamma release in vivo and demonstrates that TLR9, mDCs, and IL-12 are functionally linked to the activation of NK cells in visceral leishmaniasis.


Subject(s)
Dendritic Cells/immunology , Interleukin-12/immunology , Killer Cells, Natural/immunology , Leishmaniasis, Visceral/immunology , Lymphocyte Activation/immunology , Myeloid Cells/immunology , Toll-Like Receptor 9/immunology , Animals , CD11c Antigen/metabolism , Cell Differentiation , DNA, Protozoan/genetics , Dendritic Cells/cytology , Dendritic Cells/metabolism , Female , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Interleukin-12/deficiency , Interleukin-12/genetics , Killer Cells, Natural/cytology , Leishmania donovani/genetics , Leishmaniasis, Visceral/metabolism , Leishmaniasis, Visceral/parasitology , Leishmaniasis, Visceral/pathology , Mice , Mice, Knockout , Myeloid Cells/cytology , Phenotype , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism
11.
J Bone Miner Res ; 38(1): 86-102, 2023 01.
Article in English | MEDLINE | ID: mdl-36332102

ABSTRACT

Vector-borne infections of humans with the protozoan parasite Leishmania (L.) infantum can cause a systemic and potentially lethal disease termed visceral leishmaniasis. In the corresponding mouse model, an intravenous infection with L. infantum leads to the persistence of parasites in various organs, including bone marrow (BM). Considering the anatomical proximity between the BM and the cortical bone, we investigated whether a chronic infection with L. infantum affected bone homeostasis. Unexpectedly, chronic infection with L. infantum caused an increase in bone mass in mice. In vivo, an increased number of osteoblasts and osteocytes and a decreased maturation of osteoclasts characterized the phenotype. Confocal laser scanning fluorescence microscopy confirmed the infection of BM macrophages but also revealed the presence of parasites in osteoclasts. In vitro, mature osteoclasts took up L. infantum parasites. However, infection of osteoclast progenitors abolished their differentiation and function. In addition, secretory products of infected BM-derived macrophages inhibited the maturation of osteoclasts. Both in vitro and in vivo, infected macrophages and osteoclasts showed an enhanced expression of the anti-osteoclastogenic chemokine CCL5 (RANTES). Neutralization of CCL5 prevented the inhibition of osteoclast generation seen in the presence of culture supernatants from L. infantum-infected macrophages. Altogether, our study shows that chronic infection with Leishmania increases bone mass by inducing bone formation and impairing osteoclast differentiation and function. © 2022 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Leishmania infantum , Leishmaniasis, Visceral , Humans , Animals , Mice , Leishmania infantum/genetics , Persistent Infection , Leishmaniasis, Visceral/metabolism , Leishmaniasis, Visceral/parasitology , Leishmaniasis, Visceral/prevention & control , Macrophages/metabolism , Bone Marrow
12.
EMBO Mol Med ; 15(2): e15931, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36479617

ABSTRACT

Infection with the intracellular bacterium Coxiella (C.) burnetii can cause chronic Q fever with severe complications and limited treatment options. Here, we identify the enzyme cis-aconitate decarboxylase 1 (ACOD1 or IRG1) and its product itaconate as protective host immune pathway in Q fever. Infection of mice with C. burnetii induced expression of several anti-microbial candidate genes, including Acod1. In macrophages, Acod1 was essential for restricting C. burnetii replication, while other antimicrobial pathways were dispensable. Intratracheal or intraperitoneal infection of Acod1-/- mice caused increased C. burnetii burden, weight loss and stronger inflammatory gene expression. Exogenously added itaconate restored pathogen control in Acod1-/- mouse macrophages and blocked replication in human macrophages. In axenic cultures, itaconate directly inhibited growth of C. burnetii. Finally, treatment of infected Acod1-/- mice with itaconate efficiently reduced the tissue pathogen load. Thus, ACOD1-derived itaconate is a key factor in the macrophage-mediated defense against C. burnetii and may be exploited for novel therapeutic approaches in chronic Q fever.


Subject(s)
Coxiella burnetii , Q Fever , Animals , Humans , Mice , Coxiella burnetii/genetics , Macrophages , Q Fever/genetics , Q Fever/microbiology
13.
Pathogens ; 11(8)2022 Jul 25.
Article in English | MEDLINE | ID: mdl-35894051

ABSTRACT

Interleukin (IL)-4 plays a central role in the initiation of a type 2 T helper cell (Th2) response, which leads to non-healing and progressive infections with the protozoan parasite Leishmania (L.) major. Here, we tested the hypothesis that type 2 innate lymphoid cells (ILC2), which promote the development of Th2 cells, form an important source of IL-4 early after intradermal or subcutaneous L. major infection. Lineage-marker negative CD90.2+CD127+PD1- ILC2 were readily detectable in the ear or foot skin, but hardly in the draining lymph nodes of both naïve and L. major-infected self-healing C57BL/6 and non-healing BALB/c mice and made up approximately 20% to 30% of all CD45+SiglecF- cells. Dermal ILC2 of C57BL/6 mice expressed the inducible T cell-costimulator (ICOS, CD278), whereas BALB/C ILC2 were positive for the stem cell antigen (Sca)-1. Within the first 5 days of infection, the absolute numbers of ILC2 did not significantly change in the dermis, which is in line with the unaltered expression of cytokines activating (IL-18, IL-25, IL-33, TSLP) or inhibiting ILC2 (IL-27, IFN-γ). At day 5 to 6 post infection, we observed an upregulation of IL-4, but not of IL-5, IL-10 or IL-13 mRNA. Using IL-4-reporter (4get) mice, we found that the production of IL-4 by C57BL/6 or BALB/c mice was largely restricted to CD45+SiglecF+ cells of high granularity, i.e., eosinophils. From these data, we conclude that eosinophils, but not ILC2, are a major innate source of IL-4 at the skin site of L. major infection.

14.
Infect Immun ; 79(7): 2699-708, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21518784

ABSTRACT

Natural killer (NK) cells are important components of a protective immune response against intracellular pathogens such as Leishmania parasites, which reside within myeloid cells. Previous in vivo studies in murine cutaneous or visceral leishmaniasis showed that NK cells are activated by conventional dendritic cells in a Toll-like receptor 9-, interleukin-12 (IL-12)-, and IL-18-dependent manner during the early phase of infection and help to restrict the tissue parasite burden by unknown mechanisms. Here, we tested whether NK cells contribute to the control of Leishmania infections by lysing or by activating infected host cells. Coculture experiments revealed that activated NK cells from poly(I:C)-treated mice readily killed tumor target cells, whereas Leishmania infantum- or L. major-infected macrophages or dendritic cells remained viable. Infection with Leishmania did not significantly alter the expression of NK cell-activating molecules (retinoic acid early transcript alpha [Rae-1α], mouse UL16-binding protein-like transcript 1 [MULT-1], CD48) or inhibitory molecules (major histocompatibility complex [MHC] class I, nonclassical MHC class 1b molecule Qa-1) on the surface of myeloid cells, which offers an explanation for their protection from NK cell cytotoxicity. Consistent with these in vitro data, in vivo cytotoxicity assays revealed poor cytolytic activity of NK cells against adoptively transferred infected wild-type macrophages, whereas MHC class I-deficient macrophages were efficiently eliminated. NK cells activated by IL-12 and IL-18 stimulated macrophages to kill intracellular Leishmania in a cell contact-independent but gamma interferon-, tumor necrosis factor-, and inducible nitric oxide synthase-dependent manner. We conclude that Leishmania parasites, unlike viruses, do not render infected myeloid cells susceptible to the cytotoxicity of NK cells. Instead, soluble products of NK cells trigger the leishmanicidal activity of macrophages.


Subject(s)
Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Leishmania/immunology , Macrophages/parasitology , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , CD48 Antigen , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Coculture Techniques , Dendritic Cells/immunology , Flow Cytometry , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class I/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-12/biosynthesis , Interleukin-12/immunology , Interleukin-18/biosynthesis , Interleukin-18/immunology , Killer Cells, Natural/metabolism , Leishmaniasis/immunology , Lymphocyte Activation , Macrophages/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/immunology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Poly I-C/pharmacology , Tumor Necrosis Factors/immunology , Tumor Necrosis Factors/metabolism
15.
Eur J Immunol ; 40(6): 1708-17, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20213736

ABSTRACT

Activation of NK cells is a hallmark of infections with intracellular pathogens. We previously showed that the protozoan parasite Leishmania infantum triggered a rapid NK-cell response in mice that required TLR9-positive myeloid DC and IL-12, but no IFN-alpha/beta. Here, we investigated whether IL-15 or IL-18 mediate the activity of IL-12 or function as independent activators of NK cells. In contrast to earlier studies that described IL-15 as crucial for NK-cell priming in response to TLR ligands, the expression of IFN-gamma, FasL, perforin and granzyme B by NK cells in L. infantum-infected mice was completely preserved in the absence of IL-15, whereas the proliferative capacity of NK cells was lower than in WT mice. IFN-gamma secretion, cytotoxicity and FasL expression of NK cells from infected IL-18(-/-) mice were significantly reduced compared with controls, but, unlike IL-12, IL-18 was not essential for NK-cell effector functions. Part of the NK-cell-stimulatory effect of IL-12 was dependent on IL-18. We conclude that IL-15 is not functioning as a universal NK-cell priming signal and that IL-18 contributes to the NK-cell response in visceral leishmaniasis. The cytokine requirements for NK-cell activation appear to differ contingent upon the infectious pathogen.


Subject(s)
Interleukin-12/immunology , Interleukin-15/immunology , Interleukin-18/immunology , Killer Cells, Natural/immunology , Leishmaniasis, Visceral/immunology , Lymphocyte Activation/immunology , Animals , Cell Separation , Female , Flow Cytometry , Interferon-gamma/immunology , Leishmania infantum/immunology , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis
16.
Cells ; 10(7)2021 07 19.
Article in English | MEDLINE | ID: mdl-34359992

ABSTRACT

Arginase 1 (ARG1) is a cytosolic enzyme that cleaves L-arginine, the substrate of inducible nitric oxide synthase (iNOS), and thereby impairs the control of various intracellular pathogens. Herein, we investigated the role of ARG1 during infection with Salmonella enterica serovar Typhimurium (S.tm). To study the impact of ARG1 on Salmonella infections in vitro, bone marrow-derived macrophages (BMDM) from C57BL/6N wild-type, ARG1-deficient Tie2Cre+/-ARG1fl/fl and NRAMPG169 C57BL/6N mice were infected with S.tm. In wild-type BMDM, ARG1 was induced by S.tm and further upregulated by the addition of interleukin (IL)-4, whereas interferon-γ had an inhibitory effect. Deletion of ARG1 did not result in a reduction in bacterial numbers. In vivo, Arg1 mRNA was upregulated in the spleen, but not in the liver of C57BL/6N mice following intraperitoneal S.tm infection. The genetic deletion of ARG1 (Tie2Cre+/-ARG1fl/fl) or its pharmacological inhibition with CB-1158 neither affected the numbers of S.tm in spleen, liver and blood nor the expression of host response genes such as iNOS, IL-6 or tumour necrosis factor (TNF). Furthermore, ARG1 was dispensable for pathogen control irrespective of the presence or absence of the phagolysosomal natural resistance-associated macrophage protein 1 (NRAMP1). Thus, unlike the detrimental function of ARG1 seen during infections with other intraphagosomal microorganisms, ARG1 did not support bacterial survival in systemic salmonellosis, indicating differential roles of arginine metabolism for host immune response and microbe persistence depending on the type of pathogen.


Subject(s)
Arginase/metabolism , Cytokines/metabolism , Macrophages/metabolism , Macrophages/microbiology , Salmonella Infections, Animal/enzymology , Salmonella typhimurium/physiology , Animals , Bone Marrow Cells/microbiology , Cation Transport Proteins , Integrases/metabolism , Interleukin-4/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Mice, Transgenic , Pyrrolidines/pharmacology , Up-Regulation
17.
PLoS Pathog ; 4(11): e1000208, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19008951

ABSTRACT

The early systemic production of interferon (IFN)-alphabeta is an essential component of the antiviral host defense mechanisms, but is also thought to contribute to the toxic side effects accompanying gene therapy with adenoviral vectors. Here we investigated the IFN-alphabeta response to human adenoviruses (Ads) in mice. By comparing the responses of normal, myeloid (m)DC- and plasmacytoid (p)DC-depleted mice and by measuring IFN-alphabeta mRNA expression in different organs and cells types, we show that in vivo, Ads elicit strong and rapid IFN-alphabeta production, almost exclusively in splenic mDCs. Using knockout mice, various strains of Ads (wild type, mutant and UV-inactivated) and MAP kinase inhibitors, we demonstrate that the Ad-induced IFN-alphabeta response does not require Toll-like receptors (TLR), known cytosolic sensors of RNA (RIG-I/MDA-5) and DNA (DAI) recognition and interferon regulatory factor (IRF)-3, but is dependent on viral endosomal escape, signaling via the MAP kinase SAPK/JNK and IRF-7. Furthermore, we show that Ads induce IFN-alphabeta and IL-6 in vivo by distinct pathways and confirm that IFN-alphabeta positively regulates the IL-6 response. Finally, by measuring TNF-alpha responses to LPS in Ad-infected wild type and IFN-alphabetaR(-/-) mice, we show that IFN-alphabeta is the key mediator of Ad-induced hypersensitivity to LPS. These findings indicate that, like endosomal TLR signaling in pDCs, TLR-independent virus recognition in splenic mDCs can also produce a robust early IFN-alphabeta response, which is responsible for the bulk of IFN-alphabeta production induced by adenovirus in vivo. The signaling requirements are different from known TLR-dependent or cytosolic IFN-alphabeta induction mechanisms and suggest a novel cytosolic viral induction pathway. The hypersensitivity to components of the microbial flora and invading pathogens may in part explain the toxic side effects of adenoviral gene therapy and contribute to the pathogenesis of adenoviral disease.


Subject(s)
Adenoviridae/immunology , Dendritic Cells/immunology , Interferon Type I/immunology , Spleen/cytology , Animals , Endosomes , Humans , Interferon Type I/genetics , Interleukin-6/genetics , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred Strains , Mitogen-Activated Protein Kinases , RNA, Messenger/analysis , Signal Transduction , Spleen/immunology , Tissue Distribution , Up-Regulation
18.
Front Immunol ; 11: 209, 2020.
Article in English | MEDLINE | ID: mdl-32117319

ABSTRACT

Cryptococcus neoformans is an opportunistic fungal pathogen preferentially causing disease in immunocompromised individuals such as organ-transplant-recipients, patients receiving immunosuppressive medications or, in particular, individuals suffering from HIV infection. Numerous studies clearly indicated that the control of C. neoformans infections is strongly dependent on a prototypic type 1 immune response and classical macrophage activation, whereas type 2-biased immunity and alternative activation of macrophages has been rather implicated in disease progression and detrimental outcomes. However, little is known about regulatory pathways modulating and balancing immune responses during early phases of pulmonary cryptococcosis. Here, we analyzed the role of group 2 innate lymphoid cells (ILC2s) for the control of C. neoformans infection. Using an intranasal infection model with a highly virulent C. neoformans strain, we found that ILC2 numbers were strongly increased in C. neoformans-infected lungs along with induction of a type 2 response. Mice lacking ILC2s due to conditional deficiency of the transcription factor RAR-related orphan receptor alpha (Rora) displayed a massive downregulation of features of type 2 immunity as reflected by reduced levels of the type 2 signature cytokines IL-4, IL-5, and IL-13 at 14 days post-infection. Moreover, ILC2 deficiency was accompanied with increased type 1 immunity and classical macrophage activation, while the pulmonary numbers of eosinophils and alternatively activated macrophages were reduced in these mice. Importantly, this shift in pulmonary macrophage polarization in ILC2-deficient mice correlated with improved fungal control and prolonged survival of infected mice. Conversely, adoptive transfer of ILC2s was associated with a type 2 bias associated with less efficient anti-fungal immunity in lungs of recipient mice. Collectively, our date indicate a non-redundant role of ILC2 in orchestrating myeloid anti-cryptococcal immune responses toward a disease exacerbating phenotype.


Subject(s)
Cryptococcosis/immunology , Immunity, Innate , Lung Diseases, Fungal/immunology , Lymphocytes/physiology , Animals , Cytokines/biosynthesis , Macrophage Activation , Mice , Mice, Inbred C57BL , Myeloid Cells/physiology
19.
Virulence ; 11(1): 465-481, 2020 12.
Article in English | MEDLINE | ID: mdl-32408806

ABSTRACT

Mycobacterium avium subspecies paratuberculosis (MAP) causes Johne's disease (JD), an incurable chronic intestinal bowel disease in ruminants. JD occurs worldwide and causes enormous economic burden in dairy industry. Research on JD pathobiology is hampered by its complexity which cannot completely be mimicked by small animal models. As a model the mouse allows dissecting some pathogenicity features of MAP. However, for unknown reasons MAP exhibits reduced growth in granulomas of infected mice compared to other Mycobacterium avium subspecies. Here, we characterized immune reactions of MAP-infected C57BL/6 mice. After infection, mice appeared fully immunocompetent. A strong antigen-specific T cell response was elicited indicated by IFNγ production of splenic T cells re-stimulated with MAP antigens. Function of splenic dendritic cells and proliferation of adoptively transferred antigen-specific CD4+ T cells was unaltered. Isolated splenic myeloid cells from infected mice revealed that MAP resides in CD11b+ macrophages. Importantly, sorted CD11b+CD11c- cells expressed high level of type 2 nitric oxide synthase (NOS2) but only low levels of pro- and anti-inflammatory cytokines. Correspondingly, MAP-infected MAC2 expressing myeloid cells in spleen and liver granuloma displayed strong expression of NOS2. In livers of infected Nos2-/-mice higher bacterial loads, more granuloma and larger areas of tissue damage were observed 5 weeks post infection compared to wild type mice. In vitro, MAP was sensitive to NO released by a NO-donor. Thus, a strong T cell response and concomitant NOS2/NO activity appears to control MAP infection, but allows development of chronicity and pathogen persistence. A similar mechanism might explain persistence of MAP in ruminants.


Subject(s)
Cytokines/immunology , Nitric Oxide Synthase Type II/immunology , Paratuberculosis/immunology , Animals , Female , Immunity, Cellular , Liver/microbiology , Liver/pathology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium avium subsp. paratuberculosis , Nitric Oxide Synthase Type II/genetics , Spleen/microbiology , Spleen/pathology , T-Lymphocytes/immunology
20.
J Clin Invest ; 130(11): 5703-5720, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32721946

ABSTRACT

Arginase 1 (Arg1), which converts l-arginine into ornithine and urea, exerts pleiotropic immunoregulatory effects. However, the function of Arg1 in inflammatory bowel disease (IBD) remains poorly characterized. Here, we found that Arg1 expression correlated with the degree of inflammation in intestinal tissues from IBD patients. In mice, Arg1 was upregulated in an IL-4/IL-13- and intestinal microbiota-dependent manner. Tie2-Cre Arg1fl/fl mice lacking Arg1 in hematopoietic and endothelial cells recovered faster from colitis than Arg1-expressing (Arg1fl/fl) littermates. This correlated with decreased vessel density, compositional changes in intestinal microbiota, diminished infiltration by myeloid cells, and an accumulation of intraluminal polyamines that promote epithelial healing. The proresolving effect of Arg1 deletion was reduced by an l-arginine-free diet, but rescued by simultaneous deletion of other l-arginine-metabolizing enzymes, such as Arg2 or Nos2, demonstrating that protection from colitis requires l-arginine. Fecal microbiota transfers from Tie2-Cre Arg1fl/fl mice into WT recipients ameliorated intestinal inflammation, while transfers from WT littermates into Arg1-deficient mice prevented an advanced recovery from colitis. Thus, an increased availability of l-arginine as well as altered intestinal microbiota and metabolic products accounts for the accelerated resolution from colitis in the absence of Arg1. Consequently, l-arginine metabolism may serve as a target for clinical intervention in IBD patients.


Subject(s)
Arginase/metabolism , Gastrointestinal Microbiome , Hyperargininemia , Inflammatory Bowel Diseases , Metabolome , Animals , Arginase/genetics , Arginine/genetics , Arginine/metabolism , Endothelial Cells/enzymology , Endothelial Cells/pathology , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/pathology , Hyperargininemia/genetics , Hyperargininemia/metabolism , Hyperargininemia/microbiology , Hyperargininemia/pathology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL