Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters











Publication year range
1.
Sci Rep ; 10(1): 12317, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32704052

ABSTRACT

The contribution of the nucleotide-binding oligomerization domain protein NOD1 to obesity has been investigated in mice fed a high fat diet (HFD). Absence of NOD1 accelerates obesity as early as 2 weeks after feeding a HFD. The obesity was due to increases in abdominal and inguinal adipose tissues. Analysis of the resting energy expenditure showed an impaired function in NOD1-deficient animals, compatible with an alteration in thyroid hormone homeostasis. Interestingly, free thyroidal T4 increased in NOD1-deficient mice fed a HFD and the expression levels of UCP1 in brown adipose tissue were significantly lower in NOD1-deficient mice than in the wild type animals eating a HFD, thus contributing to the observed adiposity in NOD1-deficient mice. Feeding a HFD resulted in an alteration of the proinflammatory profile of these animals, with an increase in the infiltration of inflammatory cells in the liver and in the white adipose tissue, and an elevation of the circulating levels of TNF-α. In addition, alterations in the gut microbiota in NOD1-deficient mice correlate with increased vulnerability of their ecosystem to the HFD challenge and affect the immune-metabolic phenotype of obese mice. Together, the data are compatible with a protective function of NOD1 against low-grade inflammation and obesity under nutritional conditions enriched in saturated lipids. Moreover, one of the key players of this early obesity onset is a dysregulation in the metabolism and release of thyroid hormones leading to reduced energy expenditure, which represents a new role for these hormones in the metabolic actions controlled by NOD1.


Subject(s)
Diet, High-Fat , Feeding Behavior , Gastrointestinal Microbiome , Homeostasis , Nod1 Signaling Adaptor Protein/deficiency , Thyroid Hormones/metabolism , Adipocytes/metabolism , Adipose Tissue, White/pathology , Animals , Biodiversity , Body Weight , Fatty Liver/pathology , Glucose Tolerance Test , Inflammation/pathology , Intestines/pathology , Lipids/chemistry , Liver/pathology , Metabolomics , Mice, Inbred C57BL , Mice, Knockout , Nod1 Signaling Adaptor Protein/metabolism , Obesity/blood , Obesity/microbiology , Obesity/pathology , Thyroid Gland/pathology , Thyroid Gland/physiopathology , Thyroid Hormones/blood
2.
Basic Res Cardiol ; 115(4): 47, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32588196

ABSTRACT

Atherosclerosis is crucially fueled by inflammatory pathways including pattern recognition receptor (PRR)-related signaling of the innate immune system. Currently, the impact of the cytoplasmic PRRs nucleotide-binding oligomerization domain-containing protein (NOD) 1 and 2 is incompletely characterized. We, therefore, generated Nod1/Nod2 double knockout mice on a low-density lipoprotein receptor (Ldlr)-deficient background (= Ldlr-/-Nod1/2-/-) which were subsequently analyzed regarding experimental atherosclerosis, lipid metabolism, insulin resistance and gut microbiota composition. Compared to Ldlr-/- mice, Ldlr-/-Nod1/2-/- mice showed reduced plasma lipids and increased hepatic expression of the scavenger receptor LDL receptor-related protein 1 after feeding a high-fat diet for 12 weeks. Furthermore, intestinal cholesterol and its bacterial degradation product coprostanol were elevated in Ldlr-/-Nod1/2-/- mice, correlating with the increased abundance of Eubacterium coprostanoligenes as assessed by 3rd generation sequencing of the gut microbiota. Atherosclerotic plaques of Ldlr-/-Nod1/2-/- mice exhibited less lipid deposition and macrophage accumulation. Moreover, macrophages from Ldlr-/-Nod1/2-/- mice showed higher expression of the cholesterol efflux transporters Abca1 and Abcg1 and accordingly reduced foam cell formation. Deficiency of Nod1 and Nod2 led to reduced plaque lipid deposition and inflammatory cell infiltration in atherosclerotic plaques. This might be explained by diminished plasma lipid levels and foam cell formation due to altered expression of key regulators of the hepatic cholesterol pathway as well as differential intestinal cholesterol metabolism and microbiota composition.


Subject(s)
Atherosclerosis/metabolism , Gastrointestinal Microbiome/physiology , Lipid Metabolism/physiology , Nod1 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/deficiency , Animals , Hypercholesterolemia/complications , Mice , Mice, Knockout
3.
J Immunol ; 204(8): 2216-2231, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32169843

ABSTRACT

Nucleotide oligomerization domain-like receptors (NLRs) and RIG-I-like receptors (RLRs) detect diverse pathogen-associated molecular patterns to activate the innate immune response. The role of mammalian NLR NOD1 in sensing bacteria is well established. Although several studies suggest NOD1 also plays a role in sensing viruses, the mechanisms behind this are still largely unknown. In this study, we report on the synergism and antagonism between NOD1 and MDA5 isoforms in teleost. In zebrafish, the overexpression of NOD1 enhances the antiviral response and mRNA abundances of key antiviral genes involved in RLR-mediated signaling, whereas the loss of NOD1 has the opposite effect. Notably, spring viremia of carp virus-infected NOD1-/- zebrafish exhibit reduced survival compared with wild-type counterparts. Mechanistically, NOD1 targets MDA5 isoforms and TRAF3 to modulate the formation of MDA5-MAVS and TRAF3-MAVS complexes. The cumulative effects of NOD1 and MDA5a (MDA5 normal form) were observed for the binding with poly(I:C) and the formation of the MDA5a-MAVS complex, which led to increased transcription of type I IFNs and ISGs. However, the antagonism between NOD1 and MDA5b (MDA5 truncated form) was clearly observed during proteasomal degradation of NOD1 by MDA5b. In humans, the interactions between NOD1-MDA5 and NOD1-TRAF3 were confirmed. Furthermore, the roles that NOD1 plays in enhancing the binding of MDA5 to MAVS and poly(I:C) are also evolutionarily conserved across species. Taken together, our findings suggest that mutual regulation between NOD1 and MDA5 isoforms may play a crucial role in the innate immune response and that NOD1 acts as a positive regulator of MDA5/MAVS normal form-mediated immune signaling in vertebrates.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DEAD-box RNA Helicases/metabolism , Nod1 Signaling Adaptor Protein/metabolism , RNA, Viral/metabolism , Signal Transduction/immunology , Zebrafish Proteins/metabolism , Animals , Binding Sites , Cells, Cultured , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate , Nod1 Signaling Adaptor Protein/deficiency , Zebrafish , Zebrafish Proteins/deficiency
4.
J Biol Chem ; 294(22): 9007-9015, 2019 05 31.
Article in English | MEDLINE | ID: mdl-30996003

ABSTRACT

NOD1 and NOD2 are intracellular sensors of bacterial peptidoglycan that belong to the Nod-like receptor family of innate immune proteins. In addition to their role as direct bacterial sensors, it was proposed that the nucleotide-binding oligomerization domain (NOD) proteins could detect endoplasmic reticulum (ER) stress induced by thapsigargin, an inhibitor of the sarcoplasmic or endoplasmic reticulum calcium ATPase family that pumps Ca2+ into the ER, resulting in pro-inflammatory signaling. Here, we confirm that thapsigargin induces NOD-dependent pro-inflammatory signaling in epithelial cells. However, the effect was specific to thapsigargin, as tunicamycin and the subtilase cytotoxin SubAB from Shiga toxigenic Escherichia coli, which induce ER stress by other mechanisms, did not induce cytokine expression. The calcium ionophore A23187 also induced NOD-dependent signaling, and calcium chelators demonstrated a role for both intracellular and extracellular calcium in mediating thapsigargin-induced and NOD-dependent pro-inflammatory signaling, in part through the activation of plasma membrane-associated calcium release-activated channels. Moreover, our results demonstrate that both endocytosis and the addition of serum to the cell culture medium were required for thapsigargin-mediated NOD activation. Finally, we analyzed cell culture grade fetal calf serum as well as serum from laboratory mice using HPLC and MS identified the presence of various peptidoglycan fragments. We propose that cellular perturbations that affect intracellular Ca2+ can trigger internalization of peptidoglycan trace contaminants found in culture serum, thereby stimulating pro-inflammatory signaling. The presence of peptidoglycan in animal serum suggests that a homeostatic function of NOD signaling may have been previously overlooked.


Subject(s)
Cytokines/metabolism , Endoplasmic Reticulum Stress , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Peptidoglycan/blood , Calcimycin/chemistry , Calcimycin/pharmacology , Calcium/chemistry , Calcium/metabolism , Chemokine CXCL1/metabolism , Endoplasmic Reticulum Stress/drug effects , Gene Knockout Techniques , HCT116 Cells , Humans , Interleukin-8/metabolism , Nod1 Signaling Adaptor Protein/deficiency , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/genetics , Signal Transduction/drug effects , Thapsigargin/pharmacology
5.
BMC Nephrol ; 19(1): 78, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29609537

ABSTRACT

BACKGROUND: Chronic kidney disease (CKD) is characterized by sustained tissue damage and ongoing tubulo-interstitial inflammation and fibrosis. Pattern recognition receptors (PRRs) including Toll-like receptors (TLRs) and NOD-like receptors (NLRs) can sense endogenous ligands released upon tissue damage, leading to sterile inflammation and eventually irreversible kidney disease. It is known that NOD1 and NOD2 contribute to the pathogenesis of various inflammatory diseases, including acute kidney injury. However their role in chronic kidney disease is largely unknown. The aim of this study was therefore to investigate the contribution of NOD1 and NOD2 in renal interstitial fibrosis and obstructive nephropathy. METHODS: To do so, we performed unilateral ureteral obstruction (UUO) in wild type (WT) and NOD1/NOD2 double deficient (DKO) mice and analysed renal damage, fibrosis and inflammation. Data were analysed using the non-parametric Mann-Whitney U-test. RESULTS: Minor changes in inflammatory response were observed in NOD1/2 DKO mice, while no effects were observed on renal injury and the development of fibrosis. CONCLUSION: No difference in renal injury and fibrosis between WT and NOD1/NOD2 DKO mice following obstructive nephropathy induced by ureteral obstruction.


Subject(s)
Acute Kidney Injury/metabolism , Nod1 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/deficiency , Renal Insufficiency, Chronic/metabolism , Ureteral Obstruction/metabolism , Acute Kidney Injury/etiology , Acute Kidney Injury/genetics , Animals , Female , Fibrosis/etiology , Fibrosis/genetics , Fibrosis/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/genetics , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/genetics , Ureteral Obstruction/complications , Ureteral Obstruction/genetics
6.
PLoS Pathog ; 13(12): e1006725, 2017 12.
Article in English | MEDLINE | ID: mdl-29211798

ABSTRACT

Leptospirosis is a widespread zoonosis, potentially severe in humans, caused by spirochetal bacteria, Leptospira interrogans (L. interrogans). Host defense mechanisms involved in leptospirosis are poorly understood. Recognition of lipopolysaccharide (LPS) and lipoproteins by Toll-Like Receptors (TLR)4 and TLR2 is crucial for clearance of leptospires in mice, yet the role of Nucleotide Oligomerization Domain (NOD)-like receptors (NOD)1 and NOD2, recognizing peptidoglycan (PG) fragments has not previously been examined. Here, we show that pathogenic leptospires escape from NOD1 and NOD2 recognition both in vitro and in vivo, in mice. We found that leptospiral PG is resistant to digestion by certain hydrolases and that a conserved outer membrane lipoprotein of unknown function, LipL21, specific for pathogenic leptospires, is tightly bound to the PG. Leptospiral PG prepared from a mutant not expressing LipL21 (lipl21-) was more readily digested than the parental or complemented strains. Muropeptides released from the PG of the lipl21- mutant, or prepared using a procedure to eliminate the LipL21 protein from the PG of the parental strain, were recognized in vitro by the human NOD1 (hNOD1) and NOD2 (hNOD2) receptors, suggesting that LipL21 protects PG from degradation into muropeptides. LipL21 expressed in E. coli also resulted in impaired PG digestion and NOD signaling. We found that murine NOD1 (mNOD1) did not recognize PG of L. interrogans. This result was confirmed by mass spectrometry showing that leptospiral PG was primarily composed of MurTriDAP, the natural agonist of hNOD1, and contained only trace amounts of the tetra muropeptide, the mNOD1 agonist. Finally, in transgenic mice expressing human NOD1 and deficient for the murine NOD1, we showed enhanced clearance of a lipl21- mutant compared to the complemented strain, or to what was observed in NOD1KO mice, suggesting that LipL21 facilitates escape from immune surveillance in humans. These novel mechanisms allowing L. interrogans to escape recognition by the NOD receptors may be important in circumventing innate host responses.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Immune Evasion , Leptospira interrogans/immunology , Leptospira interrogans/pathogenicity , Lipoproteins/metabolism , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Peptidoglycan/metabolism , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Female , Humans , Immune Evasion/genetics , Immunity, Innate , Leptospira/immunology , Leptospira interrogans/genetics , Leptospirosis/genetics , Leptospirosis/immunology , Leptospirosis/microbiology , Lipoproteins/genetics , Lipoproteins/immunology , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , Nod1 Signaling Adaptor Protein/deficiency , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/genetics , Peptidoglycan/chemistry , Peptidoglycan/immunology , Protein Binding , Signal Transduction , Species Specificity , Virulence/genetics , Virulence/immunology
7.
J Immunol ; 199(3): 1196-1205, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28652394

ABSTRACT

Nucleotide-binding oligomerization domain (Nod)-containing proteins Nod1 and Nod2 play important roles in the innate immune response to pathogenic microbes, but mounting data suggest these pattern recognition receptors might also play key roles in adaptive immune responses. Targeting Nod1 and Nod2 signaling pathways in T cells is likely to provide a new strategy to modify inflammation in a variety of disease states, particularly those that depend on Ag-induced T cell activation. To better understand how Nod1 and Nod2 proteins contribute to adaptive immunity, this study investigated their role in alloantigen-induced T cell activation and asked whether their absence might impact in vivo alloresponses using a severe acute graft versus host disease model. The study provided several important observations. We found that the simultaneous absence of Nod1 and Nod2 primed T cells for activation-induced cell death. T cells from Nod1 × 2-/- mice rapidly underwent cell death upon exposure to alloantigen. The Nod1 × 2-/- T cells had sustained p53 expression that was associated with downregulation of its negative regulator MDM2. In vivo, mice transplanted with an inoculum containing Nod1 × 2-/- T cells were protected from severe graft versus host disease. The results show that the simultaneous absence of Nod1 and Nod2 is associated with accelerated T cell death upon alloantigen encounter, suggesting these proteins might provide new targets to ameliorate T cell responses in a variety of inflammatory states, including those associated with bone marrow or solid organ transplantation.


Subject(s)
Lymphocyte Activation , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Adaptive Immunity , Animals , Cell Death , Disease Models, Animal , Down-Regulation , Genes, p53/genetics , Genes, p53/immunology , Graft vs Host Disease/immunology , Immunity, Innate , Isoantigens/immunology , Mice , Nod1 Signaling Adaptor Protein/deficiency , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/immunology , Proto-Oncogene Proteins c-mdm2/metabolism , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , Signal Transduction
8.
Sci Rep ; 7(1): 2979, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28592872

ABSTRACT

Pattern recognition receptors (PRRs) are crucial for host defense and tissue homeostasis against infecting pathogens. PRRs are highly conserved cross species, suggesting their key roles in fundamental biological processes. Though much have been learned for NOD1 receptor in the innate and adaptive immune responses, the roles of NOD1 during embryonic and larval stages remain poorly understood. Here, we report that NOD1 is necessary for the modulation of PI3K-Akt pathway and larval survival in zebrafish. Transcriptome analysis revealed that the significantly enriched pathways in NOD1 -/- zebrafish larvae were mainly involved in metabolism and immune system processes. Biochemical analysis demonstrated that NOD1 was required for the expression of CD44a that, in turn, activated the PI3K-Akt pathway during larval development. Conversely, over-expression of CD44a in NOD1-deficient zebrafish restored the modulation of the PI3K-Akt pathway and improved larval survival. Collectively, our work indicates that NOD1 plays a previously undetected protective role in larval survival through CD44a-mediated activation of the PI3K-Akt signaling.


Subject(s)
Hyaluronan Receptors/metabolism , Nod1 Signaling Adaptor Protein/deficiency , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Animals, Genetically Modified , Base Sequence , CRISPR-Associated Protein 9/metabolism , Computational Biology/methods , Gene Expression , Gene Expression Profiling , Gene Knockout Techniques , Genes, MHC Class I , Histocompatibility Antigens Class II/genetics , Hyaluronan Receptors/genetics , Immune System/immunology , Immune System/metabolism , Larva , Models, Biological , RNA, Guide, Kinetoplastida/genetics , Zebrafish
9.
J Immunol ; 196(12): 5121-9, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27183588

ABSTRACT

Chronic intestinal inflammation is a major risk factor for the development of colorectal cancer. Nod1, a member of the Nod-like receptor (NLR) family of pattern recognition receptors, is a bacterial sensor that has been previously demonstrated to reduce susceptibility of mice to chemically induced colitis and subsequent tumorigenesis, but the mechanism by which it mediates its protection has not been elucidated. In this study, we show that Nod1 expression in the hematopoietic cell compartment is critical for limiting inflammation-induced intestinal tumorigenesis. Specifically, Nod1-deficient T cells exhibit impaired IFN-γ production during dextran sulfate sodium (DSS)-induced acute inflammation in vivo, and administration of the Nod1 ligand KF1B enhances IFN-γ responses by anti-CD3-activated T cells in vitro. Absence of IFN-γ signaling results in increased inflammation-associated tumors in mice, and adoptive transfer of Nod1(-/-) or IFNγ(-/-) T cells into T cell-deficient mice results in increased tumorigenesis as compared with T cell-deficient mice that were adoptively transferred with wild-type T cells. Collectively, these results suggest a previously unappreciated role for the innate immune receptor Nod1 in suppressing colitis-associated tumorigenesis through a T cell-mediated mechanism.


Subject(s)
Carcinogenesis , Colitis/complications , Gene Expression Regulation , Interferon-gamma/biosynthesis , Nod1 Signaling Adaptor Protein/metabolism , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Colitis/chemically induced , Colitis/immunology , Dextran Sulfate , Disease Models, Animal , Immunity, Innate , Inflammation/chemically induced , Inflammation/immunology , Interferon-gamma/immunology , Intestines/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod1 Signaling Adaptor Protein/deficiency , Nod1 Signaling Adaptor Protein/genetics
10.
Blood ; 127(20): 2460-71, 2016 05 19.
Article in English | MEDLINE | ID: mdl-26989200

ABSTRACT

Maintenance of myeloid cell homeostasis requires continuous turnover of phagocytes from the bloodstream, yet whether environmental signals influence phagocyte longevity in the absence of inflammation remains unknown. Here, we show that the gut microbiota regulates the steady-state cellular lifespan of neutrophils and inflammatory monocytes, the 2 most abundant circulating myeloid cells and key contributors to inflammatory responses. Treatment of mice with broad-spectrum antibiotics, or with the gut-restricted aminoglycoside neomycin alone, accelerated phagocyte turnover and increased the rates of their spontaneous apoptosis. Metagenomic analyses revealed that neomycin altered the abundance of intestinal bacteria bearing γ-d-glutamyl-meso-diaminopimelic acid, a ligand for the intracellular peptidoglycan sensor Nod1. Accordingly, signaling through Nod1 was both necessary and sufficient to mediate the stimulatory influence of the flora on myeloid cell longevity. Stimulation of Nod1 signaling increased the frequency of lymphocytes in the murine intestine producing the proinflammatory cytokine interleukin 17A (IL-17A), and liberation of IL-17A was required for transmission of Nod1-dependent signals to circulating phagocytes. Together, these results define a mechanism through which intestinal microbes govern a central component of myeloid homeostasis and suggest perturbations of commensal communities can influence steady-state regulation of cell fate.


Subject(s)
Gastrointestinal Microbiome/physiology , Homeostasis , Peptidoglycan/pharmacology , Phagocytes/cytology , Adoptive Transfer , Animals , Animals, Congenic , Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Cell Survival/physiology , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/pharmacology , Female , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Interleukin-17/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Neutrophils/cytology , Nod1 Signaling Adaptor Protein/deficiency , Nod1 Signaling Adaptor Protein/physiology , Nod2 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/physiology , Phagocytes/drug effects , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/physiology
11.
Mucosal Immunol ; 9(5): 1234-49, 2016 09.
Article in English | MEDLINE | ID: mdl-26813347

ABSTRACT

Nucleotide-binding oligomerization domain 1 (NOD1) fulfills important host-defense functions via its responses to a variety of gut pathogens. Recently, however, we showed that in acute pancreatitis caused by administration of cholecystokinin receptor (CCKR) agonist (cerulein) NOD1 also has a role in inflammation via its responses to gut commensal organisms. In the present study, we explored the long-term outcome of such NOD1 responsiveness in a new model of chronic pancreatitis induced by repeated administration of low doses of cerulein in combination with NOD1 ligand. We found that the development of chronic pancreatitis in this model requires intact NOD1 and type I IFN signaling and that such signaling mediates a macrophage-mediated inflammatory response that supports interleukin (IL)-33 production by acinar cells. The IL-33, in turn, has a necessary role in the induction of IL-13 and TGF-ß1, factors causing the fibrotic reaction characteristic of chronic pancreatitis. Interestingly, the Th2 effects of IL-33 were attenuated by the concomitant type I IFN response since the inflammation was marked by clear increases in IFN-γ and TNF-α production but only marginal increases in IL-4 production. These studies establish chronic pancreatitis as an IL-33-dependent inflammation resulting from synergistic interactions between the NOD1 and CCKR signaling pathways.


Subject(s)
Ceruletide/administration & dosage , Diaminopimelic Acid/analogs & derivatives , Interleukin-33/immunology , Nod1 Signaling Adaptor Protein/immunology , Pancreatitis, Chronic/immunology , Receptors, Cholecystokinin/immunology , Acinar Cells/drug effects , Acinar Cells/immunology , Acinar Cells/pathology , Animals , Diaminopimelic Acid/administration & dosage , Disease Models, Animal , Gene Expression Regulation , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-33/genetics , Interleukin-4/genetics , Interleukin-4/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod1 Signaling Adaptor Protein/deficiency , Nod1 Signaling Adaptor Protein/genetics , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/genetics , Pancreatitis, Chronic/pathology , Receptors, Cholecystokinin/genetics , Signal Transduction , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/pathology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
12.
Immunobiology ; 221(1): 70-5, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26255090

ABSTRACT

Tuberculosis due to Mycobacterium tuberculosis infection is a leading cause of death worldwide. Recognition of this pathogen is crucial for the activation of innate and adaptive immune responses. Nucleotide-binding oligomerization domain (Nod)1 and Nod2 are cytoplasmic receptors that can detect unique muropeptides of bacterial peptidoglycan. Nod2 is critical for the initiation of the host immune response against M. tuberculosis infection, however the role of Nod1 remains largely unknown. We investigated the role of Nod1 with respect to cytokine production by bone marrow-derived macrophages (BMDMs) in response to M. tuberculosis infection. Production of proinflammatory cytokines, such as IL-6, TNF-α, and IL-1ß were induced in BMDMs; cytokine levels were not affected by a deficiency in Nod1. Activation of NF-κB and MAPKs was also comparable between wild-type and Nod1-deficient BMDMs. Levels of IL-6 and IL-1ß were reduced in Nod1/Nod2 double-deficient BMDMs to a greater extent than in Nod2-deficient cells. Furthermore, when signaling of Toll-like receptors (TLRs) was inhibited by lipopolysaccharide pre-treatment, cytokine production was diminished in Nod1-deficient BMDMs. Our results indicate that Nod1 cooperates with Nod2 or TLRs to produce cytokines in macrophages in response to M. tuberculosis infection.


Subject(s)
Macrophages/immunology , Mycobacterium tuberculosis/immunology , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Toll-Like Receptors/immunology , Animals , Gene Expression Regulation , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Lipopolysaccharides/pharmacology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/immunology , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Nod1 Signaling Adaptor Protein/deficiency , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/genetics , Primary Cell Culture , Signal Transduction , Toll-Like Receptors/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
13.
Mol Oral Microbiol ; 31(3): 243-258, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26177212

ABSTRACT

Periodontitis is a polymicrobial inflammatory disease that results from the interaction between the oral microbiota and the host immunity. Although the innate immune response is important for disease initiation and progression, the innate immune receptors that recognize both classical and putative periodontal pathogens that elicit an immune response have not been elucidated. By using the Human Oral Microbe Identification Microarray (HOMIM), we identified multiple predominant oral bacterial species in human plaque biofilm that strongly associate with severe periodontitis. Ten of the identified species were evaluated in greater depth, six being classical pathogens and four putative novel pathogens. Using human peripheral blood monocytes (HPBM) and murine bone-marrow-derived macrophages (BMDM) from wild-type (WT) and Toll-like receptor (TLR)-specific and MyD88 knockouts (KOs), we demonstrated that heat-killed Campylobacter concisus, Campylobacter rectus, Selenomonas infelix, Porphyromonas endodontalis, Porphyromonas gingivalis, and Tannerella forsythia mediate high immunostimulatory activity. Campylobacter concisus, C. rectus, and S. infelix exhibited robust TLR4 stimulatory activity. Studies using mesothelial cells from WT and NOD1-specific KOs and NOD2-expressing human embryonic kidney cells demonstrated that Eubacterium saphenum, Eubacterium nodatum and Filifactor alocis exhibit robust NOD1 stimulatory activity, and that Porphyromonas endodontalis and Parvimonas micra have the highest NOD2 stimulatory activity. These studies allowed us to provide important evidence on newly identified putative pathogens in periodontal disease pathogenesis showing that these bacteria exhibit different immunostimulatory activity via TLR4, NOD1, and NOD2 (Clinicaltrials.gov NCT01154855).


Subject(s)
Dental Plaque/microbiology , Immunization , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Periodontal Diseases/immunology , Periodontal Diseases/microbiology , Toll-Like Receptor 4/immunology , Animals , Biofilms , Campylobacter rectus/immunology , Campylobacter rectus/isolation & purification , Campylobacter rectus/pathogenicity , Dental Plaque/immunology , Female , Humans , Macrophages/immunology , Male , Mice , Mice, Knockout , Monocytes , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/immunology , Nod1 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/deficiency , Periodontal Diseases/physiopathology , Porphyromonas/immunology , Porphyromonas/isolation & purification , Porphyromonas/pathogenicity , Porphyromonas endodontalis/immunology , Porphyromonas endodontalis/isolation & purification , Porphyromonas endodontalis/pathogenicity , Porphyromonas gingivalis/immunology , Porphyromonas gingivalis/isolation & purification , Tannerella forsythia/immunology , Tannerella forsythia/isolation & purification , Tannerella forsythia/pathogenicity
14.
Cardiovasc Res ; 106(3): 375-86, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25824149

ABSTRACT

AIMS: Inflammation is a significant contributor to cardiovascular disease and its complications; however, whether the myocardial inflammatory response is harmonized after cardiac injury remains to be determined. Some receptors of the innate immune system, including the nucleotide-binding oligomerization domain-like receptors (NLRs), play key roles in the host response after cardiac damage. Nucleotide-binding oligomerization domain containing 1 (NOD1), a member of the NLR family, is expressed in the heart, but its functional role has not been elucidated. We determine whether selective NOD1 activation modulates cardiac function and Ca(2+) signalling. METHODS AND RESULTS: Mice were treated for 3 days with the selective NOD1 agonist C12-iE-DAP (iE-DAP), and cardiac function and Ca(2+) cycling were assessed. We found that iE-DAP treatment resulted in cardiac dysfunction, measured as a decrease in ejection fraction and fractional shortening. Cardiomyocytes isolated from iE-DAP-treated mice displayed a decrease in the L-type Ca(2+) current, [Ca(2+)]i transients and Ca(2+) load, and decreased expression of phospho-phospholamban, sarcoplasmic reticulum-ATPase, and Na(+)-Ca(2+) exchanger. Furthermore, iE-DAP prompted 'diastolic Ca(2+) leak' in cardiomyocytes, resulting from increased Ca(2+) spark frequency and RyR2 over-phosphorylation. Importantly, these iE-DAP-induced changes in Ca(2+) cycling were lost in NOD1(-/-) mice, indicating that iE-DAP exerts its actions through NOD1. Co-treatment of mice with iE-DAP and a selective inhibitor of NF-κB (BAY11-7082) prevented cardiac dysfunction and Ca(2+) handling impairment induced by iE-DAP. CONCLUSION: Our data provide the first evidence that NOD1 activation induces cardiac dysfunction associated with excitation-contraction coupling impairment through NF-κB activation and uncover a new pro-inflammatory player in the regulation of cardiovascular function.


Subject(s)
Calcium/metabolism , Excitation Contraction Coupling , Inflammation Mediators/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Ventricular Dysfunction, Left/metabolism , Ventricular Function, Left , Animals , Anti-Inflammatory Agents/pharmacology , Calcium Channels, L-Type/metabolism , Calcium-Binding Proteins/metabolism , Cells, Cultured , Excitation Contraction Coupling/drug effects , Inflammation Mediators/agonists , Inflammation Mediators/antagonists & inhibitors , Male , Membrane Potentials , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Nod1 Signaling Adaptor Protein/deficiency , Nod1 Signaling Adaptor Protein/genetics , Phosphorylation , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium-Calcium Exchanger/metabolism , Stroke Volume , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left/drug effects
15.
PLoS One ; 9(5): e97675, 2014.
Article in English | MEDLINE | ID: mdl-24828250

ABSTRACT

Obesity is associated with inflammation that can drive metabolic defects such as hyperlipidemia and insulin resistance. Specific metabolites can contribute to inflammation, but nutrient intake and obesity are also associated with altered bacterial load in metabolic tissues (i.e. metabolic endotoxemia). These bacterial cues can contribute to obesity-induced inflammation. The specific bacterial components and host receptors that underpin altered metabolic responses are emerging. We previously showed that Nucleotide-binding oligomerization domain-containing protein 1 (NOD1) activation with bacterial peptidoglycan (PGN) caused insulin resistance in mice. We now show that PGN induces cell-autonomous lipolysis in adipocytes via NOD1. Specific bacterial PGN motifs stimulated lipolysis in white adipose tissue (WAT) explants from WT, but not NOD1⁻/⁻mice. NOD1-activating PGN stimulated mitogen activated protein kinases (MAPK),protein kinase A (PKA), and NF-κB in 3T3-L1 adipocytes. The NOD1-mediated lipolysis response was partially reduced by inhibition of ERK1/2 or PKA alone, but not c-Jun N-terminal kinase (JNK). NOD1-stimulated lipolysis was partially dependent on NF-κB and was completely suppressed by inhibiting ERK1/2 and PKA simultaneously or hormone sensitive lipase (HSL). Our results demonstrate that bacterial PGN stimulates lipolysis in adipocytes by engaging a stress kinase, PKA, NF-κB-dependent lipolytic program. Bacterial NOD1 activation is positioned as a component of metabolic endotoxemia that can contribute to hyperlipidemia, systemic inflammation and insulin resistance by acting directly on adipocytes.


Subject(s)
Adipocytes/drug effects , Adipose Tissue/drug effects , Lipolysis/drug effects , Nod1 Signaling Adaptor Protein/genetics , Peptidoglycan/pharmacology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Gonads/cytology , Gonads/drug effects , Gonads/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/deficiency , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Tissue Culture Techniques
16.
Gut Microbes ; 4(3): 222-31, 2013.
Article in English | MEDLINE | ID: mdl-23549220

ABSTRACT

Patients with inflammatory bowel diseases (IBD) harbour intestinal bacterial communities with altered composition compared with healthy counterparts; however, it is unknown whether changes in the microbiota are associated with genetic susceptibility of individuals for developing disease or instead reflect other changes in the intestinal environment related to the disease itself. Since deficiencies in the innate immune receptors Nod1 and Nod2 are linked to IBD, we tested the hypothesis that Nod-signaling alters intestinal immune profiles and subsequently alters bacterial community structure. We used qPCR to analyze expression patterns of selected immune mediators in the ileum and cecum of Nod-deficient mice compared with their Nod-sufficient littermates and assessed the relative abundance of major bacterial groups sampled from the ileum, cecum and colon. The Nod1-deficient ileum exhibited significantly lower expression of Nod2, Muc2, α- and ß-defensins and keratinocyte-derived chemokine (KC), suggesting a weakened epithelial barrier compared with WT littermates; however, there were no significant differences in the relative abundance of targeted bacterial groups, indicating that Nod1-associated immune differences alone do not promote dysbiosis. Furthermore, Nod2-deficient mice did not display any changes in the expression of immune markers or bacterial communities. Shifts in bacterial communities that were observed in this study correlated with housing conditions and were independent of genotype. These findings emphasize the importance of using F2 littermate controls to minimize environmental sources of variation in microbial analyses, to establish baseline conditions for host-microbe homeostasis in Nod-deficient mice and to strengthen models for testing factors contributing to microbial dysbiosis associated with IBD.


Subject(s)
Biota , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Homeostasis , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Signal Transduction , Animals , Biomarkers/analysis , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod1 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/deficiency , Real-Time Polymerase Chain Reaction
17.
J Immunol ; 188(10): 5116-22, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22491249

ABSTRACT

Intracellular pattern recognition receptors such as the nucleotide-binding oligomerization domain (NOD)-like receptors family members are key for innate immune recognition of microbial infection and may play important roles in the development of inflammatory diseases, including rheumatic diseases. In this study, we evaluated the role of NOD1 and NOD2 on development of experimental arthritis. Ag-induced arthritis was generated in wild-type, NOD1(-/-), NOD2(-/-), or receptor-interacting serine-threonine kinase 2(-/-) (RIPK2(-/-)) immunized mice challenged intra-articularly with methylated BSA. Nociception was determined by electronic Von Frey test. Neutrophil recruitment and histopathological analysis of proteoglycan lost was evaluated in inflamed joints. Joint levels of inflammatory cytokine/chemokine were measured by ELISA. Cytokine (IL-6 and IL-23) and NOD2 expressions were determined in mice synovial tissue by RT-PCR. The NOD2(-/-) and RIPK2(-/-), but not NOD1(-/-), mice are protected from Ag-induced arthritis, which was characterized by a reduction in neutrophil recruitment, nociception, and cartilage degradation. NOD2/RIPK2 signaling impairment was associated with a reduction in proinflammatory cytokines and chemokines (TNF, IL-1ß, and CXCL1/KC). IL-17 and IL-17 triggering cytokines (IL-6 and IL-23) were also reduced in the joint, but there is no difference in the percentage of CD4(+) IL-17(+) cells in the lymph node between arthritic wild-type and NOD2(-/-) mice. Altogether, these findings point to a pivotal role of the NOD2/RIPK2 signaling in the onset of experimental arthritis by triggering an IL-17-dependent joint immune response. Therefore, we could propose that NOD2 signaling is a target for the development of new therapies for the control of rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/immunology , Interleukin-17/metabolism , Knee Joint/immunology , Nod2 Signaling Adaptor Protein/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Signal Transduction/immunology , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Cattle , Cells, Cultured , Interleukin-17/physiology , Knee Joint/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod1 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/deficiency , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Serum Albumin, Bovine/immunology , Serum Albumin, Bovine/toxicity , Signal Transduction/genetics
18.
Microbes Infect ; 12(11): 819-27, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20685341

ABSTRACT

The intracellular bacterium Legionella pneumophila induces a severe form of pneumonia called Legionnaires diseases, which is characterized by a strong neutrophil (NE) infiltrate to the lungs of infected individuals. Although the participation of pattern recognition receptors, such as Toll-like receptors, was recently demonstrated, there is no information on the role of nod-like receptors (NLRs) for bacterial recognition in vivo and for NE recruitment to the lungs. Here, we employed a murine model of Legionnaires disease to evaluate host and bacterial factors involved in NE recruitment to the mice lungs. We found that L. pneumophila type four secretion system, known as Dot/Icm, was required for NE recruitment as dot/icm mutants fail to trigger NE recruitment in a process independent of bacterial multiplication. By using mice deficient for Nod1, Nod2, and Rip2, we found that these receptors accounted for NE recruitment to the lungs of infected mice. In addition, Rip2-dependent responses were important for cytokine production and bacterial clearance. Collectively, these studies show that Nod1, Nod2, and Rip2 account for generation of innate immune responses in vivo, which are important for NE recruitment and bacterial clearance in a murine model of Legionnaires diseases.


Subject(s)
Legionella pneumophila/immunology , Legionnaires' Disease/immunology , Lung/immunology , Neutrophil Infiltration , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Male , Mice , Mice, Knockout , Nod1 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/deficiency , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptors, Pattern Recognition/immunology , Virulence Factors/immunology
19.
J Immunol ; 184(3): 1148-52, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20042586

ABSTRACT

An effective innate immune recognition of the intracellular protozoan parasite Trypanosoma cruzi is critical for host resistance against Chagas disease, a severe and chronic illness that affects millions of people in Latin America. In this study, we evaluated the participation of nucleotide-binding oligomerization domain (Nod)-like receptor proteins in host response to T. cruzi infection and found that Nod1-dependent, but not Nod2-dependent, responses are required for host resistance against infection. Bone marrow-derived macrophages from Nod1(-/-) mice showed an impaired induction of NF-kappaB-dependent products in response to infection and failed to restrict T. cruzi infection in presence of IFN-gamma. Despite normal cytokine production in the sera, Nod1(-/-) mice were highly susceptible to T. cruzi infection, in a similar manner to MyD88(-/-) and NO synthase 2(-/-) mice. These studies indicate that Nod1-dependent responses account for host resistance against T. cruzi infection by mechanisms independent of cytokine production.


Subject(s)
Chagas Disease/immunology , Immunity, Innate , Nod1 Signaling Adaptor Protein/physiology , Trypanosoma cruzi/immunology , Animals , Chagas Disease/genetics , Chagas Disease/metabolism , Genetic Predisposition to Disease , Immunity, Innate/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/physiology , Nod1 Signaling Adaptor Protein/deficiency , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptors/physiology
20.
Infect Immun ; 77(10): 4529-37, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19620350

ABSTRACT

Toll-like receptors and Nod-like receptors (NLR) play an important role in sensing invading microorganisms for pathogen clearance and eliciting adaptive immunity for protection against rechallenge. Nod1 and Nod2, members of the NLR family, are capable of detecting bacterial peptidoglycan motifs in the host cytosol for triggering proinflammatory cytokine production. In the current study, we sought to determine if Nod1/Nod2 are involved in sensing Bacillus anthracis infection and eliciting protective immune responses. Using mice deficient in both Nod1 and Nod2 proteins, we showed that Nod1/Nod2 are involved in detecting B. anthracis for production of tumor necrosis factor alpha, interleukin-1 alpha (IL-1 alpha), IL-1 beta, CCL5, IL-6, and KC. Proinflammatory responses were higher when cells were exposed to viable spores than when they were exposed to irradiated spores, indicating that recognition of vegetative bacilli through Nod1/Nod2 is significant. We also identify a critical role for Nod1/Nod2 in priming responses after B. anthracis aerosol exposure, as mice deficient in Nod1/Nod2 were impaired in their ability to mount an anamnestic antibody response and were more susceptible to secondary lethal challenge than wild-type mice.


Subject(s)
Aerosols , Anthrax/immunology , Bacillus anthracis/immunology , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Animals , Antibodies, Bacterial/blood , Cytokines/metabolism , Mice , Mice, Knockout , Nod1 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/deficiency , Spores, Bacterial/immunology , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL