Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Int J Mol Sci ; 25(10)2024 May 13.
Article in English | MEDLINE | ID: mdl-38791357

ABSTRACT

The lung is prone to infections from respiratory viruses such as Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). A challenge in combating these infections is the difficulty in targeting antiviral activity directly at the lung mucosal tract. Boosting the capability of the respiratory mucosa to trigger a potent immune response at the onset of infection could serve as a potential strategy for managing respiratory infections. This study focused on screening immunomodulators to enhance innate immune response in lung epithelial and immune cell models. Through testing various subfamilies and pathways of pattern recognition receptors (PRRs), the nucleotide-binding and oligomerization domain (NOD)-like receptor (NLR) family was found to selectively activate innate immunity in lung epithelial cells. Activation of NOD1 and dual NOD1/2 by the agonists TriDAP and M-TriDAP, respectively, increased the number of IL-8+ cells by engaging the NF-κB and interferon response pathways. Lung epithelial cells showed a stronger response to NOD1 and dual NOD1/2 agonists compared to control. Interestingly, a less-pronounced response to NOD1 agonists was noted in PBMCs, indicating a tissue-specific effect of NOD1 in lung epithelial cells without inducing widespread systemic activation. The specificity of the NOD agonist pathway was confirmed through gene silencing of NOD1 (siRNA) and selective NOD1 and dual NOD1/2 inhibitors in lung epithelial cells. Ultimately, activation induced by NOD1 and dual NOD1/2 agonists created an antiviral environment that hindered SARS-CoV-2 replication in vitro in lung epithelial cells.


Subject(s)
COVID-19 , Epithelial Cells , Lung , Nod1 Signaling Adaptor Protein , SARS-CoV-2 , Humans , A549 Cells , Antiviral Agents/pharmacology , COVID-19/immunology , COVID-19/virology , COVID-19 Drug Treatment , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/pharmacology , Epithelial Cells/virology , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/immunology , Immunity, Innate/drug effects , Interleukin-8/metabolism , Lung/immunology , Lung/virology , Lung/metabolism , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Nod1 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/metabolism , SARS-CoV-2/physiology , SARS-CoV-2/immunology , Signal Transduction/drug effects
2.
J Immunol ; 206(9): 2206-2220, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33846227

ABSTRACT

Interactions between pattern-recognition receptors shape innate immune responses to pathogens. NOD1 and TLR4 are synergistically interacting receptors playing a pivotal role in the recognition of Gram-negative bacteria. However, mechanisms of their cooperation are poorly understood. It is unclear whether synergy is produced at the level of signaling pathways downstream of NOD1 and TLR4 or at more distal levels such as gene transcription. We analyzed sequential stages of human macrophage activation by a combination of NOD1 and TLR4 agonists (N-acetyl-d-muramyl-l-alanyl-d-isoglutamyl-meso-diaminopimelic acid [M-triDAP] and LPS, respectively). We show that events preceding or not requiring activation of transcription, such as activation of signaling kinases, rapid boost of glycolysis, and most importantly, nuclear translocation of NF-κB, are regulated nonsynergistically. However, at the output of the nucleus, the combination of M-triDAP and LPS synergistically induces expression of a subset of M-triDAP- and LPS-inducible genes, particularly those encoding proinflammatory cytokines (TNF, IL1B, IL6, IL12B, and IL23A). This synergistic response develops between 1 and 4 h of agonist treatment and requires continuous signaling through NOD1. The synergistically regulated genes have a lower basal expression and higher inducibility at 4 h than those regulated nonsynergistically. Both gene subsets include NF-κB-inducible genes. Therefore, activation of the NF-κB pathway does not explain synergistic gene induction, implying involvement of other transcription factors. Inhibition of IKKß or p38 MAPK lowers agonist-induced TNF mRNA expression but does not abolish synergy. Thus, nonsynergistic activation of NOD1- and TLR4-dependent signaling pathways results in the synergistic induction of a proinflammatory transcriptional program.


Subject(s)
Nod1 Signaling Adaptor Protein/immunology , Toll-Like Receptor 4/immunology , Acetylglucosamine/analogs & derivatives , Acetylglucosamine/pharmacology , Cytokines/genetics , Cytokines/immunology , Humans , Lipopolysaccharides/pharmacology , Macrophages , Nod1 Signaling Adaptor Protein/agonists , Signal Transduction/drug effects , Toll-Like Receptor 4/agonists
3.
Infect Immun ; 90(3): e0048521, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35225652

ABSTRACT

The human-restricted pathogen Neisseria meningitidis, which is best known for causing invasive meningococcal disease, has a nonpathogenic lifestyle as an asymptomatic colonizer of the human naso- and oropharyngeal space. N. meningitidis releases small peptidoglycan (PG) fragments during growth. It was demonstrated previously that N. meningitidis releases low levels of tripeptide PG monomer, which is an inflammatory molecule recognized by the human intracellular innate immune receptor NOD1. In the present study, we demonstrated that N. meningitidis released more PG-derived peptides than PG monomers. Using a reporter cell line overexpressing human NOD1, we showed that N. meningitidis activates NOD1 using PG-derived peptides. The generation of such peptides required the presence of the periplasmic N-acetylmuramyl-l-alanine amidase AmiC and the outer membrane lipoprotein NlpD. AmiC and NlpD were found to function in cell separation, and mutation of either amiC or nlpD resulted in large clumps of unseparated N. meningitidis cells instead of the characteristic diplococci. Using stochastic optical reconstruction microscopy, we demonstrated that FLAG epitope-tagged NlpD localized to the septum, while similarly tagged AmiC was found at the septum in some diplococci but was distributed around the cell in most cases. In a human whole-blood infection assay, an nlpD mutant was severely attenuated and showed particular sensitivity to complement. Thus, in N. meningitidis, the cell separation proteins AmiC and NlpD are necessary for NOD1 stimulation and survival during infection of human blood.


Subject(s)
Bacterial Proteins , Lipoproteins , Neisseria meningitidis , Nod1 Signaling Adaptor Protein , Peptidoglycan , Bacterial Proteins/metabolism , Cell Separation , Cell Wall/metabolism , Humans , Lipoproteins/metabolism , Meningococcal Infections/metabolism , Meningococcal Infections/microbiology , Neisseria meningitidis/metabolism , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/genetics , Nod1 Signaling Adaptor Protein/metabolism , Peptidoglycan/metabolism
4.
J Biol Chem ; 295(10): 3099-3114, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32005665

ABSTRACT

Upon activation with pathogen-associated molecular patterns, metabolism of macrophages and dendritic cells is shifted from oxidative phosphorylation to aerobic glycolysis, which is considered important for proinflammatory cytokine production. Fragments of bacterial peptidoglycan (muramyl peptides) activate innate immune cells through nucleotide-binding oligomerization domain (NOD) 1 and/or NOD2 receptors. Here, we show that NOD1 and NOD2 agonists induce early glycolytic reprogramming of human monocyte-derived macrophages (MDM), which is similar to that induced by the Toll-like receptor 4 (TLR4) agonist lipopolysaccharide. This glycolytic reprogramming depends on Akt kinases, independent of mTOR complex 1 and is efficiently inhibited by 2-deoxy-d-glucose (2-DG) or by glucose starvation. 2-DG inhibits proinflammatory cytokine production by MDM and monocyte-derived dendritic cells activated by NOD1 or TLR4 agonists, except for tumor necrosis factor production by MDM, which is inhibited initially, but augmented 4 h after addition of agonists and later. However, 2-DG exerts these effects by inducing unfolded protein response rather than by inhibiting glycolysis. By contrast, glucose starvation does not cause unfolded protein response and, in normoxic conditions, only marginally affects proinflammatory cytokine production triggered through NOD1 or TLR4. In hypoxia mimicked by treating MDM with oligomycin (a mitochondrial ATP synthase inhibitor), both 2-DG and glucose starvation strongly suppress tumor necrosis factor and interleukin-6 production and compromise cell viability. In summary, the requirement of glycolytic reprogramming for proinflammatory cytokine production in normoxia is not obvious, and effects of 2-DG on cytokine responses should be interpreted cautiously. In hypoxia, however, glycolysis becomes critical for cytokine production and cell survival.


Subject(s)
Cytokines/metabolism , Glycolysis/drug effects , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Nod1 Signaling Adaptor Protein/agonists , Toll-Like Receptor 4/agonists , Animals , Carboxy-Lyases/metabolism , Cell Hypoxia , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Deoxyglucose/pharmacology , Humans , Macrophages/drug effects , Macrophages/immunology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/metabolism , Oligomycins/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Toll-Like Receptor 4/metabolism , Unfolded Protein Response/drug effects
5.
J Immunol ; 200(9): 3170-3179, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29592964

ABSTRACT

Functional maturation of liver sinusoidal endothelial cells (LSECs) induced by a NOD1 ligand (diaminopimelic acid [DAP]) during viral infection has not been well defined. Thus, we investigated the role of DAP-stimulated LSEC maturation during hepatitis B virus (HBV) infection and its potential mechanism in a hydrodynamic injection (HI) mouse model. Primary LSECs were isolated from wild-type C57BL/6 mice and stimulated with DAP in vitro and in vivo and assessed for the expression of surface markers as well as for their ability to promote T cell responses via flow cytometry. The effects of LSEC maturation on HBV replication and expression and the role of LSECs in the regulation of other immune cells were also investigated. Pretreatment of LSECs with DAP induced T cell activation in vitro. HI-administered DAP induced LSEC maturation and subsequently enhanced T cell responses, which was accompanied by an increased production of intrahepatic cytokines, chemokines, and T cell markers in the liver. The HI of DAP significantly reduced the HBsAg and HBV DNA levels in the mice. Importantly, the DAP-induced anti-HBV effect was impaired in the LSEC-depleted mice, which indicated that LSEC activation and T cell recruitment into the liver were essential for the antiviral function mediated by DAP application. Taken together, the results showed that the Ag-presenting ability of LSECs was enhanced by DAP application, which resulted in enhanced T cell responses and inhibited HBV replication in a mouse model.


Subject(s)
Antigen Presentation/immunology , Endothelial Cells/immunology , Hepatitis B virus/physiology , Liver/immunology , Nod1 Signaling Adaptor Protein/agonists , Virus Replication/physiology , Animals , Capillaries/immunology , Diaminopimelic Acid/pharmacology , Hepatitis B/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Nod1 Signaling Adaptor Protein/immunology , T-Lymphocytes/immunology , Virus Replication/drug effects
6.
Med Sci Monit ; 26: e924748, 2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32855380

ABSTRACT

BACKGROUND Chronic hypertension changes the function and structure of the heart and blood vessels. This study aimed to explore the role of the NOD1/Rip2 (nucleotide-binding oligomerization domain 1/receptor-interacting protein 2) signaling pathway in myocardial remodeling in spontaneously hypertensive rats (SHRs). MATERIAL AND METHODS Blood pressure was measured using a tail cuff. The cardiac structure was observed using echocardiography. Slices of the myocardium were stained with hematoxylin and eosin. The expression of NOD1 and Rip2 was detected using real-time polymerase chain reaction, western blot, and immunohistochemistry. The content and distribution of collagen in the myocardium were observed using Van Gieson staining. Enzyme-linked immunosorbent assay was used to detect the interleukin-1 (IL-1) concentrations. SHRs were treated with the NOD1 agonist iE-DAP and NOD1 inhibitor ML130. RESULTS The NOD1 agonist increased blood pressure in SHRs, and the NOD1 inhibitor decreased blood pressure; the interventricular septum thickness (IVST) and left ventricular posterior wall thickness (LVPWT) of the agonist-treated group were thicker than those of the control group, and the antagonist exerted the opposite effects. The levels of the NOD1 and Rip2 mRNAs and proteins, serum IL-1 concentration, and myocardial collagen volume fraction (CVF%) increased in SHRs in the NOD1 agonist group, but the levels of NOD1 and Rip2, serum IL-1 concentration, and myocardial collagen volume fraction (CVF%) decreased in SHRs in the NOD1 inhibitor group. CONCLUSIONS NOD1/Rip2 expression increased during the progression of myocardial remodeling in SHRs. The NOD1 agonist increased NOD1 expression and promoted myocardial remodeling, while the NOD1 antagonist reduced NOD1/Rip2 expression and protected against myocardial remodeling.


Subject(s)
Nod1 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Signal Transduction , Ventricular Remodeling , Animals , Blood Pressure/drug effects , Male , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Real-Time Polymerase Chain Reaction
7.
J Hepatol ; 70(6): 1159-1169, 2019 06.
Article in English | MEDLINE | ID: mdl-30685324

ABSTRACT

BACKGROUND & AIMS: In liver transplantation, organ shortage leads to the use of marginal grafts that are more susceptible to ischemia-reperfusion (IR) injury. We identified nucleotide-binding oligomerization domain 1 (NOD1) as an important modulator of polymorphonuclear neutrophil (PMN)-induced liver injury, which occurs in IR. Herein, we aimed to elucidate the role of NOD1 in IR injury, particularly focusing on its effects on the endothelium and hepatocytes. METHOD: Nod1 WT and KO mice were treated with NOD1 agonists and subjected to liver IR. Expression of adhesion molecules was analyzed in total liver, isolated hepatocytes and endothelial cells. Interactions between PMNs and hepatocytes were studied in an ex vivo co-culture model using electron microscopy and lactate dehydrogenase levels. We generated NOD1 antagonist-loaded nanoparticles (np ALINO). RESULTS: NOD1 agonist treatment increased liver injury, PMN tissue infiltration and upregulated ICAM-1 and VCAM-1 expression 20 hours after reperfusion. NOD1 agonist treatment without IR increased expression of adhesion molecules (ICAM-1, VCAM-1) in total liver and more particularly in WT hepatocytes, but not in Nod1 KO hepatocytes. This induction is dependent of p38 and ERK signaling pathways. Compared to untreated hepatocytes, a NOD1 agonist markedly increased hepatocyte lysis in co-culture with PMNs as shown by the increase of lactate dehydrogenase in supernatants. Interaction between hepatocytes and PMNs was confirmed by electron microscopy. In a mouse model of liver IR, treatment with np ALINO significantly reduced the area of necrosis, aminotransferase levels and ICAM-1 expression. CONCLUSION: NOD1 regulates liver IR injury through induction of adhesion molecules and modulation of hepatocyte-PMN interactions. NOD1 antagonist-loaded nanoparticles reduced liver IR injury and provide a potential approach to prevent IR, especially in the context of liver transplantation. LAY SUMMARY: Nucleotide-binding oligomerization domain 1 (NOD1) is as an important modulator of polymorphonuclear neutrophil (PMN)-induced liver injury, which occurs in ischemia-reperfusion. Here, we show that the NOD1 pathway targets liver adhesion molecule expression on the endothelium and on hepatocytes through p38 and ERK signaling pathways. The early increase of adhesion molecule expression after reperfusion emphasizes the importance of adhesion molecules in liver injury. In this study we generated nanoparticles loaded with NOD1 antagonist. These nanoparticles reduced liver necrosis by reducing PMN liver infiltration and adhesion molecule expression.


Subject(s)
Intercellular Adhesion Molecule-1/physiology , Liver/blood supply , Nod1 Signaling Adaptor Protein/physiology , Reperfusion Injury/prevention & control , Vascular Cell Adhesion Molecule-1/physiology , Animals , Humans , Mice , Mice, Inbred C57BL , Neutrophils/physiology , Nod1 Signaling Adaptor Protein/agonists , Signal Transduction/physiology
8.
Int J Mol Sci ; 20(17)2019 Aug 30.
Article in English | MEDLINE | ID: mdl-31480368

ABSTRACT

THP-1 cells express high levels of native functional nucleotide-binding oligomerization domain 1 (NOD1), NOD2, and Toll-like receptor 4 (TLR4) receptors, and have often been used for investigating the immunomodulatory effects of small molecules. We postulated that they would represent an ideal cell-based model for our study, the aim of which was to develop a new in vitro tool for functional characterization of NOD antagonists. NOD antagonists were initially screened for their effect on NOD agonist-induced interleukin-8 (IL-8) release. Next, we examined the extent to which the selected NOD antagonists block the NOD-TLR4 synergistic crosstalk by measuring the effect of NOD antagonism on tumor necrosis factor-α (TNF-α) secretion from doubly activated THP-1 cells. Overall, the results obtained indicate that pro-inflammatory cytokine secretion from THP-1 provides a valuable, simple and reproducible in vitro tool for functional characterization of NOD antagonists.


Subject(s)
Cytokines/biosynthesis , Inflammation Mediators/metabolism , Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Cell Survival , Humans , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/metabolism , THP-1 Cells , Toll-Like Receptor 4/metabolism
9.
Immunity ; 30(6): 789-801, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19464198

ABSTRACT

Cellular inhibitor of apoptosis proteins (cIAPs) block apoptosis, but their physiological functions are still under investigation. Here, we report that cIAP1 and cIAP2 are E3 ubiquitin ligases that are required for receptor-interacting protein 2 (RIP2) ubiquitination and for nucleotide-binding and oligomerization (NOD) signaling. Macrophages derived from Birc2(-/-) or Birc3(-/-) mice, or colonocytes depleted of cIAP1 or cIAP2 by RNAi, were defective in NOD signaling and displayed sharp attenuation of cytokine and chemokine production. This blunted response was observed in vivo when Birc2(-/-) and Birc3(-/-) mice were challenged with NOD agonists. Defects in NOD2 signaling are associated with Crohn's disease, and muramyl dipeptide (MDP) activation of NOD2 signaling protects mice from experimental colitis. Here, we show that administration of MDP protected wild-type but not Ripk2(-/-) or Birc3(-/-) mice from colitis, confirming the role of the cIAPs in NOD2 signaling in vivo. This discovery provides therapeutic opportunities in the treatment of NOD-dependent immunologic and inflammatory diseases.


Subject(s)
Immunity, Innate , Inhibitor of Apoptosis Proteins/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Animals , Apoptosis/immunology , Baculoviral IAP Repeat-Containing 3 Protein , Colitis/enzymology , Colitis/immunology , Colitis/pathology , Cytokines/immunology , Cytokines/metabolism , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/immunology , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Receptors, Pattern Recognition/agonists , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Ubiquitin-Protein Ligases , Ubiquitination/immunology
10.
J Immunol ; 196(6): 2779-87, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26880761

ABSTRACT

Intrauterine fetal growth restriction (IUGR) and death (IUFD) are both serious problems in the perinatal medicine. Fetal vasculopathy is currently considered to account for a pathogenic mechanism of IUGR and IUFD. We previously demonstrated that an innate immune receptor, the nucleotide-binding oligomerization domain-1 (Nod1), contributed to the development of vascular inflammations in mice at postnatal stages. However, little is known about the deleterious effects of activated Nod1 signaling on embryonic growth and development. We report that administration of FK565, one of the Nod1 ligands, to pregnant C57BL/6 mice induced IUGR and IUFD. Mass spectrometry analysis revealed that maternally injected FK565 was distributed to the fetal tissues across placenta. In addition, maternal injection of FK565 induced robust increases in the amounts of CCL2, IL-6, and TNF proteins as well as NO in maternal, placental and fetal tissues. Nod1 was highly expressed in fetal vascular tissues, where significantly higher levels of CCL2 and IL-6 mRNAs were induced with maternal injection of FK565 than those in other tissues. Using Nod1-knockout mice, we verified that both maternal and fetal tissues were involved in the development of IUGR and IUFD. Furthermore, FK565 induced upregulation of genes associated with immune response, inflammation, and apoptosis in fetal vascular tissues. Our data thus provided new evidence for the pathogenic role of Nod1 in the development of IUGR and IUFD at the maternal-fetal interface.


Subject(s)
Fetal Death/prevention & control , Fetal Growth Retardation/immunology , Nod1 Signaling Adaptor Protein/metabolism , Oligopeptides/administration & dosage , Vasculitis/immunology , Animals , Chemokine CCL2/metabolism , Female , Fetal Death/etiology , Fetal Growth Retardation/chemically induced , Humans , Interleukin-6/metabolism , Ligands , Maternal Exposure/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/genetics , Pregnancy , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Vasculitis/chemically induced
11.
Biochem J ; 474(3): 399-410, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27803247

ABSTRACT

Cardiac fibrosis and chronic inflammation are common complications in type 2 diabetes mellitus (T2D). Since nucleotide oligomerization-binding domain 1 (NOD1), an innate immune receptor, is involved in the pathogenesis of insulin resistance and diabetes outcomes, we sought to investigate its involvement in cardiac fibrosis. Here, we show that selective staining of cardiac fibroblasts from T2D (db/db;db) mice exhibits up-regulation and activation of the NOD1 pathway, resulting in enhanced NF-κB and TGF-ß signalling. Activation of the TGF-ß pathway in cardiac fibroblasts from db mice was prevented after inhibition of NF-κB with BAY-11-7082 (BAY). Moreover, fibrosis progression in db mice was also prevented by BAY treatment. Enhanced TGF-ß signalling and cardiac fibrosis of db mice was dependent, at least in part, on the sequential activation of NOD1 and NF-κB since treatment of db mice with a selective NOD1 agonist induced activation of the TGF-ß pathway, but co-administration of a NOD1 agonist plus BAY, or a NOD1 inhibitor prevented the NOD1-induced fibrosis. Therefore, NOD1 is involved in cardiac fibrosis associated with diabetes, and establishes a new mechanism for the development of heart fibrosis linked to T2D.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Endomyocardial Fibrosis/metabolism , Myocardium/metabolism , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/pharmacology , Endomyocardial Fibrosis/genetics , Endomyocardial Fibrosis/pathology , Endomyocardial Fibrosis/prevention & control , Gene Expression Regulation , Humans , Insulin/blood , Insulin Resistance , Mice , Mice, Transgenic , Myocardium/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NIH 3T3 Cells , Nitriles/pharmacology , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/genetics , Signal Transduction , Sulfones/pharmacology , Transforming Growth Factor beta/agonists , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
12.
Infect Immun ; 84(1): 275-85, 2016 01.
Article in English | MEDLINE | ID: mdl-26527214

ABSTRACT

Mucins secreted by intestinal goblet cells are considered an important component of innate defense in a number of enteric infections, including many parasitic infections, but also likely provide protection against the gut microbiota. Nod proteins are intracellular receptors that play key roles in innate immune response and inflammation. Here, we investigated the role of Nod proteins in regulation of intestinal goblet cell response in naive mice and mice infected with the enteric parasite Trichuris muris. We observed significantly fewer periodic acid-Schiff (PAS)-stained intestinal goblet cells and less mucin (Muc2) in Nod1 and Nod2 double-knockout (Nod DKO) mice after T. muris infection than in wild-type (WT) mice. Expulsion of parasites from the intestine was significantly delayed in Nod DKO mice. Treatment of naive WT mice with Nod1 and Nod2 agonists simultaneously increased numbers of PAS-stained goblet cells and Muc2-expressing cells, whereas treatment with Nod1 or Nod2 separately had no significant effect. Stimulation of mucin-secreting LS174T cells with Nod1 and Nod2 agonists upregulated core 3 ß1,3-N-acetylglucosaminyltransferase (C3GnT; an important enzyme in mucin synthesis) and MUC2. We also observed lower numbers of PAS-stained goblet cells and less Muc2 in germfree mice. Treatment with Nod1 and Nod2 agonists enhanced the production of PAS-stained goblet cells and Muc2 in germfree mice. These data provide novel information on the role of Nod proteins in goblet cell response and Muc2 production in relation to intestinal innate defense.


Subject(s)
Goblet Cells/immunology , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Trichuriasis/immunology , Trichuris/immunology , Animals , Cell Line , Chitin Synthase/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-2/metabolism , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/genetics , Trichuriasis/parasitology
13.
J Oral Pathol Med ; 45(4): 262-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26332444

ABSTRACT

OBJECTIVES: Microbial Pattern-recognition receptors (PRRs), such as nucleotide-binding oligomerization domains (NODs), are essential for mammalian innate immune response. This study was designed to determine the effect of NOD1 and NOD2 agonist on innate immune responses and antitumor activity in oral squamous cell carcinoma (OSCC) cells. MATERIALS AND METHODS: NODs expression was examined by RT-PCR, and IL-8 production by NODs agonist was examined by ELISA. Western blot analysis was performed to determine the MAPK activation in response to their agonist. Cell proliferation was determined by MTT assay. Flow cytometry and Western blot analysis were performed to determine the MDP-induced cell death. RESULTS: The levels of NODs were apparently expressed in OSCC cells. NODs agonist, Tri-DAP and MDP, led to the production of IL-8 and MAPK activation. NOD2 agonist, MDP, inhibited the proliferation of YD-10B cells in a dose-dependent manner. Also, the ratio of Annexin V-positive cells and cleaved PARP was increased by MDP treatment in YD-10B cells, suggesting that MDP-induced cell death in YD-10B cells may be owing to apoptosis. CONCLUSIONS: Our results indicate that NODs are functionally expressed in OSCC cells and can trigger innate immune responses. In addition, NOD2 agonist inhibited cell proliferation and induced apoptosis. These findings provide the potential value of MDP as novel candidates for antitumor agents of OSCC.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Diaminopimelic Acid/analogs & derivatives , Head and Neck Neoplasms/drug therapy , Mouth Neoplasms/drug therapy , Nod2 Signaling Adaptor Protein/agonists , Oligopeptides/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Blotting, Western , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Diaminopimelic Acid/pharmacology , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Immunity, Innate/drug effects , Interleukin-8/biosynthesis , Interleukin-8/metabolism , Mitogen-Activated Protein Kinases/biosynthesis , Mitogen-Activated Protein Kinases/metabolism , Mouth Neoplasms/immunology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/biosynthesis , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/biosynthesis , Nod2 Signaling Adaptor Protein/genetics , RNA, Messenger/biosynthesis , Squamous Cell Carcinoma of Head and Neck
14.
Brain Behav Immun ; 44: 106-20, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25218901

ABSTRACT

Toll-like receptors (TLRs) and nuclear-binding domain (NOD)-like receptors (NLRs) are sensors of bacterial cell wall components to trigger an immune response. The TLR4 agonist lipopolysaccharide (LPS) is a strong immune activator leading to sickness and depressed mood. NOD agonists are less active but can prime immune cells to augment LPS-induced cytokine production. Since the impact of NOD and TLR co-activation in vivo has been little studied, the effects of the NOD1 agonist FK565 and the NOD2 agonist muramyl dipeptide (MDP), alone and in combination with LPS, on immune activation, brain function and sickness behavior were investigated in male C57BL/6N mice. Intraperitoneal injection of FK565 (0.001 or 0.003mg/kg) or MDP (1 or 3mg/kg) 4h before LPS (0.1 or 0.83mg/kg) significantly aggravated and prolonged the LPS-evoked sickness behavior as deduced from a decrease in locomotion, exploration, food intake and temperature. When given alone, FK565 and MDP had only minor effects. The exacerbation of sickness behavior induced by FK565 or MDP in combination with LPS was paralleled by enhanced plasma protein and cerebral mRNA levels of proinflammatory cytokines (IFN-γ, IL-1ß, IL-6, TNF-α) as well as enhanced plasma levels of kynurenine. Immunohistochemical visualization of c-Fos in the brain revealed that NOD2 synergism with TLR4 resulted in increased activation of cerebral nuclei relevant to sickness. These data show that NOD1 or NOD2 synergizes with TLR4 in exacerbating the immune, sickness and brain responses to peripheral immune stimulation. Our findings demonstrate that the known interactions of NLRs and TLRs at the immune cell level extend to interactions affecting brain function and behavior.


Subject(s)
Brain/immunology , Illness Behavior/physiology , Nod1 Signaling Adaptor Protein/physiology , Nod2 Signaling Adaptor Protein/physiology , Toll-Like Receptor 4/physiology , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Adjuvants, Immunologic/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Corticosterone/blood , Cytokines/blood , Cytokines/metabolism , Eating/drug effects , Illness Behavior/drug effects , Kynurenine/blood , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Nod1 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/agonists , Oligopeptides/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/metabolism , Toll-Like Receptor 4/agonists , Tryptophan/blood
15.
Am J Respir Crit Care Med ; 189(8): 899-908, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24661094

ABSTRACT

RATIONALE: Pattern recognition receptors are attractive targets for vaccine adjuvants, and polymorphisms of the innate receptor NOD1 have been associated with allergic asthma. OBJECTIVES: To elucidate whether NOD1 agonist may favor allergic asthma in humans through activation of dendritic cells, and to evaluate the mechanisms involved using an in vivo model. METHODS: NOD1-primed dendritic cells from allergic and nonallergic donors were characterized in vitro on their phenotype, cytokine secretion, and Th2 polarizing ability. The in vivo relevance was examined in experimental allergic asthma, and the mechanisms were assessed using transfer of NOD1-conditioned dendritic cells from wild-type or CCL17-deficient mice. MEASUREMENTS AND MAIN RESULTS: NOD1 priming of human dendritic cells promoted a Th2 polarization profile that involved the production of CCL17 and CCL22 in nonallergic subjects but only CCL17 in allergic patients, without requiring allergen costimulation. Moreover, NOD1-primed dendritic cells from allergic donors exhibited enhanced maturation that led to abnormal CCL22 and IL-10 secretion compared with nonallergic donors. In mice, systemic NOD1 ligation exacerbated allergen-induced experimental asthma by amplifying CCL17-mediated Th2 responses in the lung. NOD1-mediated sensitization of purified murine dendritic cells enhanced production of CCL17 and CCL22, but not of thymic stromal lymphopoietin and IL-33, in vitro. Consistently, adoptive transfer of NOD1-conditioned dendritic cells exacerbated the Th2 pulmonary response in a CCL17-dependent manner in vivo. CONCLUSIONS: Data from this study unveil a deleterious role of NOD1 in allergic asthma through direct induction of CCL17 by dendritic cells, arguing for a need to address vaccine formulation safety issues related to allergy.


Subject(s)
Asthma/immunology , Chemokine CCL17/immunology , Chemokine CCL22/immunology , Dendritic Cells/immunology , Nod1 Signaling Adaptor Protein/immunology , Allergens/immunology , Animals , Asthma/genetics , Asthma/prevention & control , Disease Models, Animal , Female , Humans , In Vitro Techniques , Interleukin-10/immunology , Mice , Mice, Inbred C57BL , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/genetics , Phenotype , Polymorphism, Genetic , Th2 Cells/immunology , Up-Regulation/immunology
16.
Immunology ; 143(2): 269-76, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24766550

ABSTRACT

Nod-like receptors are a family of innate immune receptors that link cytosolic sensing of microbial and danger stimuli to the activation of immune responses. Two Nod-like receptor family members, Nod1 and Nod2, recognize bacterial peptidoglycan and activate immune responses via nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK). The function of Nod1 and Nod2 has been largely studied in macrophages, but the role of these receptors in other innate immune cells remains unclear. In this study, we examined the function of Nod1 and Nod2 in innate immune responses of neutrophils. Mice were injected intraperitoneally with thioglycollate, and then peritoneal neutrophils were isolated 4 hr after injection. Tri-DAP and muramyl-dipeptide (MDP) were used as Nod1 and Nod2 agonists, respectively. The level of cytokines [interleukin-6 (IL-6) and tumour necrosis factor-α (TNF-α)] and chemokines (CXCL1 and CCL2) was increased by MDP, but not Tri-DAP in wild-type (WT) neutrophils. Increased production of cytokines and chemokines with MDP was abolished in Nod2- and Rip2-deficient neutrophils. MDP also induced the activation of NF-κB and MAPK in WT neutrophils, but not in Nod2- and Rip2-deficient cells. Flow cytometry analysis showed that L-selectin shedding was induced by MDP in WT neutrophils, but not in Nod2- and Rip2-deficient cells. MDP and Toll-like receptor (TLR) agonists (Pam3 CSK4 and lipopolysaccharide) exerted synergistic effects on the production of IL-6 and CXCL1 in neutrophils. Moreover, Nod2 and TLR4 cooperated to produce IL-6, TNF-α, CXCL1 and CCL2 in neutrophils in response to Gram-negative bacteria. Our findings suggest that the Nod2-Rip2 axis may contribute to the innate immune response of neutrophils against bacterial infection.


Subject(s)
Immunity, Innate , Neutrophil Activation , Neutrophils/immunology , Nod2 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Animals , Cells, Cultured , Chemokine CCL2/metabolism , Chemokine CXCL1/metabolism , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/pharmacology , Immunity, Innate/drug effects , Interleukin-6/metabolism , L-Selectin/metabolism , Lipopolysaccharides/pharmacology , Listeria monocytogenes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Neutrophil Activation/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/microbiology , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/genetics , Oligopeptides/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism , Yersinia pseudotuberculosis/immunology
17.
Molecules ; 18(1): 1162-73, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23325104

ABSTRACT

Synthesis of beneficial protected meso-DAP 9 by cross metathesis of the Garner aldehyde-derived vinyl glycine 1b with protected allyl glycine 2 in the presence of Grubbs second-generation catalyst was performed. Preparation of lipophilic N-acyl iE-DAP as potent agonists of NOD 1-mediated immune response from 9 is described.


Subject(s)
Diaminopimelic Acid/analogs & derivatives , Catalysis , Diaminopimelic Acid/chemical synthesis , Esterification , Hydrophobic and Hydrophilic Interactions , Nod1 Signaling Adaptor Protein/agonists , Oxidation-Reduction , Peptidoglycan/chemistry
18.
Arterioscler Thromb Vasc Biol ; 31(5): 1093-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21330608

ABSTRACT

OBJECTIVE: The goal of this study was to investigate the effects of stimulants for a nucleotide-binding domain, leucine-rich repeat-containing (NLR) protein family on human artery endothelial cells and murine arteries. METHODS AND RESULTS: Human coronary artery endothelial cells were challenged in vitro with microbial components that stimulate NLRs or Toll-like receptors. We found stimulatory effects of NLR and Toll-like receptor ligands on the adhesion molecule expression and cytokine secretion by human coronary artery endothelial cells. On the basis of these results, we examined the in vivo effects of these ligands in mice. Among them, FK565, 1 of the nucleotide-binding oligomerization domain (Nod)-1 ligands induced strong site-specific inflammation in the aortic root. Furthermore, coronary arteritis/valvulitis developed after direct oral administration or ad libitum drinking of FK565. The degree of the respective vascular inflammation was associated with persistent high expression of proinflammatory chemokine/cytokine and matrix metallopeptidase (Mmp) genes in each tissue in vivo by microarray analysis. CONCLUSIONS: This is the first coronary arteritis animal model induced by oral administration of a pure synthetic Nod1 ligand. The present study has demonstrated an unexpected role of Nod1 in the development of site-specific vascular inflammation, especially coronary arteritis. These findings might lead to the clarification of the pathogenesis and pathophysiology of coronary artery disease in humans.


Subject(s)
Arteritis/immunology , Coronary Vessels/immunology , Endothelial Cells/immunology , Immunity, Innate , Nod1 Signaling Adaptor Protein/metabolism , Animals , Arteritis/chemically induced , Arteritis/genetics , Arteritis/metabolism , Arteritis/pathology , Cells, Cultured , Chemokines/genetics , Chemokines/metabolism , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gene Expression Profiling/methods , Humans , Immunity, Innate/drug effects , Inflammation Mediators/metabolism , Intercellular Adhesion Molecule-1/metabolism , Ligands , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred Strains , Nod1 Signaling Adaptor Protein/agonists , Oligonucleotide Array Sequence Analysis , Oligopeptides , Organ Culture Techniques , Toll-Like Receptors/metabolism
19.
Article in Russian | MEDLINE | ID: mdl-22937699

ABSTRACT

AIM: Evaluate influence of mutation of Listeria monocytogenes genes coding murein-tetrapeptide L,D-carboxypeptidase Lmo0028 and Lmo1638 on dynamics of infectious process and interaction of purified muropeptides with NOD1 receptor. MATERIALS AND METHODS: Wild type EGDe strain and recombinant strains GIMins1638 H GIMins0028 obtained on its basis by site-specific mutagenesis were used. Infectious process dynamics was studied on the model of intravenous infection of BALB/c mice. Ligand-receptor interaction activity of muropeptides isolated from recombinant and parent strains were assayed on HEK293-hNOD1 cell line expressing NOD1 receptor and containing in their genome beta-galactosidase reporter gene under the control of NF-kappaB dependent promoter expression. RESULTS: Lack of Lmo0028 decelerates reproduction of listerias in animal liver starting from 24 hours and at later terms after the infection whereas lack of Lmo1638 leads to increase of microbial load 6 and 24 hours after the infection with no influence on further infection. Differences in activation of NOD1 receptor by muropeptides isolated from recombinant and parent strains were not detected. CONCLUSION: Despite high homology murein-tetrapeptide L,D-carboxypeptidase Lmo0028 and Lmo1638 make a different contribution to the development of infectious process caused by L. monocytogenes in BALB/c line mice. Lack of differences in NOD1 receptor activation may be associated with compensation of enzymatic functions in strains with mutation in each of the genes owing to the presence of homologous protein.


Subject(s)
Bacterial Proteins/genetics , Carboxypeptidases/genetics , Listeria monocytogenes/enzymology , Listeria monocytogenes/pathogenicity , Nod1 Signaling Adaptor Protein/agonists , Animals , Bacterial Load , Bacterial Proteins/metabolism , Carboxypeptidases/metabolism , Genes, Reporter , HEK293 Cells , Humans , Injections, Intravenous , Isoenzymes/genetics , Isoenzymes/metabolism , Listeria monocytogenes/genetics , Listeriosis/microbiology , Mice , Mice, Inbred BALB C , Mutagenesis, Site-Directed , NF-kappa B/genetics , Nod1 Signaling Adaptor Protein/genetics , Nod1 Signaling Adaptor Protein/metabolism , Peptides/genetics , Peptides/pharmacology , Peptidoglycan/metabolism , Promoter Regions, Genetic , Transfection , Virulence , beta-Galactosidase/analysis
20.
Immunology ; 134(3): 314-25, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21978001

ABSTRACT

NOD-like receptors (NLRs) and RIG-I-like receptors (RLRs) are newly discovered pattern-recognition receptors. They detect substructures of bacterial peptidoglycan and viral RNA, respectively, thereby initiating an immune response. However, their role in eosinophil activation remains to be explored. The aim of this study was to characterize the expression of a range of NLRs and RLRs in purified human eosinophils and assess their functional importance. Expression of NOD1, NOD2, NLRP3, RIG-I and MDA-5 was investigated using real-time reverse transcription PCR, flow cytometry and immunohistochemistry. The effects of the corresponding agonists iE-DAP (NOD1), MDP (NOD2), alum (NLRP3) and poly(I:C)/LyoVec (RIG-I/MDA-5) were studied in terms of cytokine secretion, degranulation, survival, expression of adhesion molecules and activation markers, and chemotactic migration. Eosinophils expressed NOD1 and NOD2 mRNA and protein. Low levels of RIG-I and MDA-5 were found, whereas expression of NLRP3 was completely absent. In accordance, stimulation with iE-DAP and MDP was found to induce secretion of interleukin-8, up-regulate expression of CD11b, conversely down-regulate CD62 ligand, increase expression of CD69 and induce migration. The MDP also promoted release of eosinophil-derived neurotoxin, whereas iE-DAP failed to do so. No effects were seen upon stimulation with alum or poly(I:C)/LyoVec. Moreover, the NOD1-induced and NOD2-induced activation was mediated via the nuclear factor-κB signalling pathway and augmented by interleukin-5 and granulocyte-macrophage colony-stimulating factor, but not interferon-γ. Taken together, the NLR system represents a novel pathway for eosinophil activation. The responses are enhanced in the presence of cytokines that regulate T helper type 2 immunity, suggesting that the NLRs constitute a link between respiratory infections and exacerbations of allergic disease.


Subject(s)
Carrier Proteins/metabolism , DEAD-box RNA Helicases/metabolism , Eosinophils/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , CD11b Antigen/metabolism , Carrier Proteins/genetics , Cell Movement/drug effects , Cells, Cultured , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/pharmacology , Eosinophil-Derived Neurotoxin/metabolism , Eosinophils/cytology , Flow Cytometry , Gene Expression/drug effects , Humans , Immunohistochemistry , Interferon-Induced Helicase, IFIH1 , Interleukin-8/metabolism , L-Selectin/metabolism , Lectins, C-Type/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/genetics , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Receptors, Immunologic , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL