Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Sci Adv ; 10(27): eado2365, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38959302

ABSTRACT

Pityriasis rubra pilaris (PRP) is a rare inflammatory skin disease with a poorly understood pathogenesis. Through a molecularly driven precision medicine approach and an extensive mechanistic pathway analysis in PRP skin samples, compared to psoriasis, atopic dermatitis, healed PRP, and healthy controls, we identified IL-1ß as a key mediator, orchestrating an NF-κB-mediated IL-1ß-CCL20 axis, including activation of CARD14 and NOD2. Treatment of three patients with the IL-1 antagonists anakinra and canakinumab resulted in rapid clinical improvement and reversal of the PRP-associated molecular signature with a 50% improvement in skin lesions after 2 to 3 weeks. This transcriptional signature was consistent with in vitro stimulation of keratinocytes with IL-1ß. With the central role of IL-1ß underscoring its potential as a therapeutic target, our findings propose a redefinition of PRP as an autoinflammatory keratinization disorder. Further clinical trials are needed to validate the efficacy of IL-1ß antagonists in PRP.


Subject(s)
Antibodies, Monoclonal, Humanized , Interleukin 1 Receptor Antagonist Protein , Interleukin-1beta , Keratinocytes , Pityriasis Rubra Pilaris , Humans , Pityriasis Rubra Pilaris/drug therapy , Pityriasis Rubra Pilaris/pathology , Pityriasis Rubra Pilaris/genetics , Interleukin-1beta/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Keratinocytes/metabolism , Keratinocytes/drug effects , Keratinocytes/pathology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Male , NF-kappa B/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Female , CARD Signaling Adaptor Proteins/metabolism , CARD Signaling Adaptor Proteins/genetics , Skin/pathology , Skin/metabolism , Skin/drug effects , Interleukin-1/antagonists & inhibitors , Interleukin-1/metabolism , Interleukin-1/genetics , Middle Aged , Guanylate Cyclase/metabolism , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/genetics , Adult , Signal Transduction/drug effects , Membrane Proteins
2.
Eur J Med Chem ; 215: 113252, 2021 Apr 05.
Article in English | MEDLINE | ID: mdl-33601309

ABSTRACT

Receptor interacting protein kinase-2 (RIPK2) is an enzyme involved in the transduction of pro-inflammatory nucleotide-binding oligomerization domain (NOD) cell signaling, a pathway implicated in numerous chronic inflammatory conditions. Herein, a pyrido[2,3-d]pyrimidin-7-one based class of RIPK2 kinase and NOD2 cell signaling inhibitors is described. For example, 33 (e.g. UH15-15) inhibited RIPK2 kinase (IC50 = 8 ± 4 nM) and displayed > 300-fold selectivity versus structurally related activin receptor-like kinase 2 (ALK2). This molecule blocked NOD2-dependent HEKBlue NF-κB activation (IC50 = 20 ± 5 nM) and CXCL8 production (at concentrations > 10 nM). Molecular docking suggests that engagement of Ser25 in the glycine-rich loop may provide increased selectivity versus ALK2 and optimal occupancy of the region between the gatekeeper and the αC-helix may contribute to potent NOD2 cell signaling inhibition. Finally, this compound also demonstrated favorable in vitro ADME and pharmacokinetic properties (e.g. Cmax = 5.7 µM, Tmax = 15 min, t1/2 = 3.4 h and Cl = 45 mL/min/kg following single 10 mg/kg intraperitoneal administration) further supporting the use of pyrido[2,3-d]pyrimidin-7-ones as a new structure class of RIPK2 kinase and NOD cell signaling inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrimidinones/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Cell Line, Tumor , Drug Design , Humans , Molecular Docking Simulation , Nod2 Signaling Adaptor Protein/chemistry , Nod2 Signaling Adaptor Protein/metabolism , Protein Binding , Protein Domains , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Pyridines/chemical synthesis , Pyridines/metabolism , Pyrimidinones/chemical synthesis , Pyrimidinones/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/chemistry , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Signal Transduction/drug effects
3.
Biochem Pharmacol ; 183: 114315, 2021 01.
Article in English | MEDLINE | ID: mdl-33152345

ABSTRACT

BACKGROUND AND PURPOSE: Previously, we discovered that the activation of nucleotide-binding oligomerization domain 2 (NOD2) enhances platelet activation. We here investigated the antiplatelet and antithrombotic potential of GSK669, a NOD2 antagonist. EXPERIMENTAL APPROACH: Effects of GSK669 on platelet functions, reactive oxygen species (ROS) and proinflammatory cytokine generation were detected. NOD2-/- platelets were used to confirm GSK669 target. The interaction between GSK669 and glycoprotein VI (GPVI) was detected using surface plasmon resonance (SPR) spectroscopy. GPVI downstream signaling was examined by Western blot. The antithrombotic and antioxidative effects were investigated using mouse mesenteric arteriole thrombosis model and pulmonary embolism model. KEY RESULTS: GSK669 significantly inhibits platelet proinflammatory cytokine release induced by muramyl dipeptide, platelet aggregation, ATP release, and ROS generation induced by collagen and collagen related peptide (CRP). Platelet spreading and clot retraction are also inhibited. GSK669 also decreases collagen-induced phosphorylation of Src, Syk, PLCγ2, and Akt. The antiplatelet effect of GSK669 is NOD2-independent and mediated by GPVI antagonism. Consistent with its antiplatelet activity as a GPVI antagonist, GSK669 inhibits platelet adhesion on collagen in flow condition. Notably, GSK669 inhibits mouse mesenteric arteriole thrombosis similarly to aspirin without bleeding. The antithrombotic effect of GSK669 is further confirmed in the pulmonary embolism model; decreased malonaldehyde (MDA) and increased superoxide dismutase (SOD) levels in mouse plasma reveal a significant antioxidant effect of GSK669. CONCLUSION AND IMPLICATIONS: Beyond its anti-inflammatory effect as a NOD2 antagonist, GSK669 is also an efficient and safe antiplatelet agent combined with antioxidant effect by targeting GPVI. An antiplatelet agent bearing antioxidative and anti-inflammatory activities without bleeding risk may have therapeutic advantage over current antiplatelet drugs for atherothrombosis.


Subject(s)
Blood Platelets/drug effects , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Oxidative Stress/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Platelet Membrane Glycoproteins/antagonists & inhibitors , Thrombosis/drug therapy , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/metabolism , Blood Platelets/metabolism , Drug Delivery Systems/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod2 Signaling Adaptor Protein/metabolism , Oxidative Stress/physiology , Platelet Aggregation Inhibitors/metabolism , Platelet Membrane Glycoproteins/metabolism , Thrombosis/metabolism
4.
Eur J Med Chem ; 207: 112723, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-32920426

ABSTRACT

Nucleotide-binding oligomerization domain-containing protein 1 and 2 (NOD1/2) receptors are potential immune checkpoints. In this article, a quinazolinone derivative (36b) as a NOD1/2 dual antagonist was identified that significantly sensitizes B16 tumor-bearing mice to paclitaxel treatment by inhibiting both nuclear factor κB (NF-κB) and mitogen-activated protein kinase inflammatory signaling that mediated by NOD1/2.


Subject(s)
Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Quinazolinones/chemistry , Quinazolinones/pharmacology , Animals , Cell Line , Drug Discovery , Humans , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Nucleotides/metabolism
5.
Eur J Med Chem ; 204: 112575, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32731185

ABSTRACT

Nucleotide-binding oligomerization domain-containing proteins 1 and 2 play important roles in immune system activation. Recently, a shift has occurred due to the emerging knowledge that preventing nucleotide-binding oligomerization domains (NODs) signaling could facilitate the treatment of some cancers, which warrants the search for dual antagonists of NOD1 and NOD2. Herein, we undertook the synthesis and identification of a new class of derivatives of dual NOD1/NOD2 antagonists with novel benzofused five-membered sultams. Compound 14k was finally demonstrated to be the most potent molecule that inhibits both NOD1-and NOD2-stimulated NF-κB and MAPK signaling in vitro and in vivo.


Subject(s)
Naphthalenesulfonates/chemistry , Naphthalenesulfonates/pharmacology , Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Animals , Drug Design , HEK293 Cells , Humans , Male , Mice , Nod1 Signaling Adaptor Protein/chemistry , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/chemistry , Nod2 Signaling Adaptor Protein/metabolism , Protein Domains , Signal Transduction/drug effects
6.
Brain Res Bull ; 158: 20-30, 2020 05.
Article in English | MEDLINE | ID: mdl-32109527

ABSTRACT

Streptococcus pneumoniae is responsible for pneumococcal meningitis, with significant mortality and morbidity worldwide. Microglial inflammation plays a vital role in meningitis. The peptidoglycan sensor NOD2 (nucleotide-binding oligomerization domain 2) has been identified to promote microglia activation, but the role in autophagy following pneumococcal meningitis remains unclear. In the present study, we investigated the role of NOD2 in microglial inflammation and autophagy, as well as related signaling pathways, during S. pneumonia infection. NOD2 expression was knocked down by the injection of lentivirus-mediated short-hairpin RNA (shRNA). Our results revealed that NOD2 promotes microglial inflammation by increasing inflammatory mediators. We also showed that the TAK1-NF-κB pathway is involved in this process. In addition, NOD2 increased the expression of autophagy-related proteins and induced autophagosome formation. Rapamycin and 3-MA were utilized to assess the role of autophagy in microglial inflammation induced by S. pneumonia. We demonstrated that autophagy serves as a cellular defense mechanism to reduce inflammatory mediators. Similar to the in vitro results, NOD2 induced inflammation and autophagy in the brain in a mouse meningitis model. Moreover, NOD2 silencing significantly reduced brain edema and improved the neurological function of pneumococcal meningitis mice. Taken together, these data demonstrate that NOD2 promotes microglial inflammation and autophagy in murine pneumococcal meningitis, and the TAK1-NF-κB pathway is involved in microglial activation.


Subject(s)
Autophagy/physiology , MAP Kinase Kinase Kinases/metabolism , Meningitis, Pneumococcal/metabolism , Microglia/metabolism , NF-kappa B/metabolism , Nod2 Signaling Adaptor Protein/deficiency , Animals , Cell Line , Inflammation/metabolism , Inflammation/pathology , Male , Meningitis, Pneumococcal/pathology , Mice , Mice, Inbred C57BL , Microglia/pathology , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/biosynthesis , Signal Transduction/physiology
7.
Eur J Med Chem ; 190: 112089, 2020 Mar 15.
Article in English | MEDLINE | ID: mdl-32014680

ABSTRACT

NOD1 and NOD2 are pattern recognition receptors that have important roles in innate immune responses. Although their overactivation has been linked to a number of diseases, NOD2 in particular remains a virtually unexploited target in this respect, with only one structural class of antagonist reported. To gain insight into the structure-activity relationships of NOD2 antagonists, a series of novel analogs was designed and synthesized, and then screened for antagonist activity versus NOD2, and counter-screened versus NOD1. Compounds 32 and 38 were identified as potent and moderately selective NOD2 antagonists, and 33 and 42 as dual NOD1/NOD2 antagonists, with balanced activities against both targets in the low micromolar range. These data enable in-depth exploration of their structure-activity relationships and provide deeper understanding of the structural features required for NOD2 antagonism.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Benzimidazoles/pharmacology , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/toxicity , Benzimidazoles/chemical synthesis , Benzimidazoles/toxicity , Drug Design , HEK293 Cells , Humans , Molecular Structure , Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Structure-Activity Relationship
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.
Mol Cell ; 69(4): 551-565.e7, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29452636

ABSTRACT

Inflammatory responses mediated by NOD2 rely on RIP2 kinase and ubiquitin ligase XIAP for the activation of nuclear factor κB (NF-κB), mitogen-activated protein kinases (MAPKs), and cytokine production. Herein, we demonstrate that selective XIAP antagonism blocks NOD2-mediated inflammatory signaling and cytokine production by interfering with XIAP-RIP2 binding, which removes XIAP from its ubiquitination substrate RIP2. We also establish that the kinase activity of RIP2 is dispensable for NOD2 signaling. Rather, the conformation of the RIP2 kinase domain functions to regulate binding to the XIAP-BIR2 domain. Effective RIP2 kinase inhibitors block NOD2 signaling by disrupting RIP2-XIAP interaction. Finally, we identify NOD2 signaling and XIAP-dependent ubiquitination sites on RIP2 and show that mutating these lysine residues adversely affects NOD2 pathway signaling. Overall, these results reveal a critical role for the XIAP-RIP2 interaction in NOD2 inflammatory signaling and provide a molecular basis for the design of innovative therapeutic strategies based on XIAP antagonists and RIP2 kinase inhibitors.


Subject(s)
Aminoquinolines/pharmacology , Inflammation/prevention & control , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Protein Interaction Domains and Motifs/drug effects , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Sulfones/pharmacology , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Cells, Cultured , Humans , Inflammation/metabolism , Inflammation/pathology , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Signal Transduction , Ubiquitin/metabolism , Ubiquitination , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors
10.
Life Sci ; 191: 180-185, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29055802

ABSTRACT

AIMS: The epidermal growth factor receptor (EGFR) and nucleotide-binding and oligomerization-domain containing 2 (NOD2) are important in cancer and in microbial recognition, respectively. These molecules trigger intracellular signaling pathways inducing the expression of inflammatory genes by NF-kB translocation. Gefitinib (GBTC) and pyrrolidine dithiocarbamate (PDTC) are capable of inhibiting EGFR/NOD2 and NF-kB, respectively. In earlier stages of dengue virus (DENV) infection, monocytes are capable of sustaining viral replication and increasing cytokine production, suggesting that monocyte/macrophages play an important role in early DENV replication. GBTC and PDTC have not been used to modify the pathogenesis of DENV in infected cells. This study was aimed to determine the effect of GBTC and PDTC on viral replication and cytokine production in DENV serotype 2 (DENV2)-infected human monocyte cultures. MAIN METHODS: GBTC and PDTC were used to inhibit EGFR/NOD2 and NF-kB, respectively. Cytokine production was measured by ELISA and viral replication by plaque forming unit assay. KEY FINDINGS: Increased DENV2 replication and anti-viral cytokine production (IFN-α/ß, TNF-α, IL-12 and IL-18) in infected cultures were found. These parameters were decreased after EGFR/NOD2 or NF-kB inhibitions. SIGNIFICANCE: The inhibitory effects of GBTC and PDTC on viral replication and cytokine production can be beneficial in the treatment of patients infected by dengue and suggest a possible role of EGFR/NOD2 receptors and NF-kB in dengue pathogenesis.


Subject(s)
Antiviral Agents/pharmacology , Cytokines/immunology , Dengue Virus/drug effects , Dengue/drug therapy , Monocytes/drug effects , Pyrrolidines/pharmacology , Quinazolines/pharmacology , Thiocarbamates/pharmacology , Adult , Animals , Cell Line , Cells, Cultured , Culicidae , Cytokines/antagonists & inhibitors , Dengue/immunology , Dengue/virology , Dengue Virus/immunology , Dengue Virus/physiology , ErbB Receptors/antagonists & inhibitors , Gefitinib , Humans , Male , Monocytes/immunology , Monocytes/virology , NF-kappa B/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Virus Replication/drug effects
11.
J Med Chem ; 60(12): 5162-5192, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28541685

ABSTRACT

Nucleotide-binding oligomerization domain-like receptors (NLRs) are intracellular sensors of pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). Previously, we reported nucleotide-binding oligomerization domain-containing protein 1 (NOD1) antagonists (11, 12) and a NOD2 antagonist (9) that sensitized docetaxel (DTX) or paclitaxel (PTX) treatment for breast or lung cancer. In this article, we describe for the first time a 1,4-benzodiazepine-2,5-dione (BZD) derivative (26bh) that acts as a dual NOD1/NOD2 antagonist and inhibits both nuclear factor κB (NF-κB) and mitogen-activated protein kinase (MAPK) inflammatory signaling, thereby sensitizing PTX to suppress Lewis lung carcinoma (LLC) growth. After investigation of the compound's cytotoxicity, a systematic structure-activity relationship (SAR) was completed and revealed several key factors that were necessary to maintain antagonistic ability. This study establishes the possibility for using adjuvant treatment to combat cancer by antagonizing both NOD1 and NOD2 signaling.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzodiazepines/chemistry , Carcinoma, Lewis Lung/drug therapy , Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Animals , Benzodiazepines/pharmacology , Cytokines/metabolism , Drug Screening Assays, Antitumor/methods , Humans , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Molecular Targeted Therapy , Paclitaxel/administration & dosage , Protein Domains , Structure-Activity Relationship
12.
Bioorg Med Chem ; 24(21): 5221-5234, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27601373

ABSTRACT

NOD1 and NOD2 are important members of the pattern recognition receptor family and play a crucial role within the context of innate immunity. However, overactivation of NODs, especially of NOD1, has also been implicated in a number of diseases. Surprisingly, NOD1 remains a virtually unexploited target in this respect. To gain additional insight into the structure-activity relationships of NOD1 inhibitors, a series of novel analogs has been designed and synthesized and then screened for their NOD1-inhibitory activity. Selected compounds were also investigated for their NOD2-inhibitory activity. Two compounds 4 and 15, were identified as potent mixed inhibitors of NOD1 and NOD2, displaying a balanced inhibitory activity on both targets in the low micromolar range. The results obtained have enabled a deeper understanding of the structural requirements for NOD1 and NOD2 inhibition.


Subject(s)
Indoles/pharmacology , Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Design , Humans , Indoles/chemical synthesis , Indoles/chemistry , Molecular Structure , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/genetics , Structure-Activity Relationship
13.
Clin Exp Immunol ; 185(1): 98-106, 2016 07.
Article in English | MEDLINE | ID: mdl-26990648

ABSTRACT

Salmonellosis or Salmonella, one of the most common food-borne diseases, remains a major public health problem worldwide. Intestinal epithelial cells (IECs) play an essential role in the mucosal innate immunity of the host to defend against the invasion of Salmonella by interleukin (IL)-8 and human ß-defensin-2 (hBD-2). Accumulated research has unravelled important roles of vitamin D in the regulation of innate immunity. Therefore, we investigated the effects of 1,25-dihydroxyvitamin D3 (1,25D3) on Salmonella-induced innate immunity in IECs. We demonstrate that pretreatment of 1,25D3 results in suppression of Salmonella-induced IL-8 but enhancement of hBD-2, either protein secretion and mRNA expression, in IECs. Furthermore, 1,25D3 enhanced Salmonella-induced membranous recruitment of nucleotide oligomerization domain (NOD2) and its mRNA expression and activation of protein kinase B (Akt), a downstream effector of phosphoinositide 3-kinase (PI3K). Inhibition of the PI3K/Akt signal counteracted the suppressive effect of 1,25D3 on Salmonella-induced IL-8 expression, while knock-down of NOD2 by siRNA diminished the enhanced hBD-2 expression. These data suggest differential regulation of 1,25D3 on Salmonella-induced IL-8 and hBD-2 expression in IECs via PI3K/Akt signal and NOD2 protein expression, respectively. Active vitamin D-enhanced anti-microbial peptide in Salmonella-infected IECs protected the host against infection, while modulation of proinflammatory responses by active vitamin D prevented the host from the detrimental effects of overwhelming inflammation. Thus, active vitamin D-induced innate immunity in IECs enhances the host's protective mechanism, which may provide an alternative therapy for invasive Salmonella infection.


Subject(s)
Calcitriol/pharmacology , Epithelial Cells/drug effects , Host-Pathogen Interactions/drug effects , Interleukin-8/genetics , beta-Defensins/genetics , Cell Line, Tumor , Epithelial Cells/immunology , Epithelial Cells/microbiology , Gene Expression Regulation , Humans , Interleukin-8/antagonists & inhibitors , Interleukin-8/immunology , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/immunology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Salmonella typhimurium/growth & development , Salmonella typhimurium/immunology , Signal Transduction , beta-Defensins/agonists , beta-Defensins/immunology
14.
Org Biomol Chem ; 14(3): 1013-23, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26631868

ABSTRACT

Peptidoglycan (PGN) is a major component of bacterial cell wall and is recognized as a potent immunostimulant. The PGN in the cell envelope of Mycobacterium Tuberculosis has been shown to possess several unique characteristics including the presence of N-glycolyl groups (in addition to N-acetyl groups) in the muramic acid residues, and amidation of the free carboxylic acid of d-Glu or of meso-DAP in the peptide chains. Using a newly developed, highly stereoselective, chemoenzymatic approach for the synthesis of meso-DAP in peptide stems, we successfully synthesized for the first time, a series of Mycobacterium PGN fragments that include both mono- and disaccharides of MurNGlyc or 1,6-anhydro-MurNGlyc, as well as peptide-amidated variants. The ability of these PGN fragments to stimulate the immune system through activation of human Nod1 and Nod2 was examined. The PGN fragments were found to modulate immune stimulation, specifically, amidation at the d-Glu and meso-DAP in the peptide stem strongly reduced hNod1 activation. This effect was dependent on modification position. Additionally, N-glycolyl (instead of acetyl) of muramic acid was associated with slightly reduced human Nod1 and Nod2 stimulatory capabilities.


Subject(s)
Diaminopimelic Acid/pharmacology , Immunity, Innate/drug effects , Immunity, Innate/immunology , Mycobacterium tuberculosis/chemistry , Peptidoglycan/immunology , Diaminopimelic Acid/chemistry , Diaminopimelic Acid/metabolism , HEK293 Cells , Humans , Molecular Conformation , Mycobacterium tuberculosis/immunology , Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/immunology , Peptidoglycan/biosynthesis , Peptidoglycan/chemistry , Structure-Activity Relationship
15.
Chem Biol ; 22(9): 1174-84, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26320862

ABSTRACT

RIPK2 mediates pro-inflammatory signaling from the bacterial sensors NOD1 and NOD2, and is an emerging therapeutic target in autoimmune and inflammatory diseases. We observed that cellular RIPK2 can be potently inhibited by type II inhibitors that displace the kinase activation segment, whereas ATP-competitive type I inhibition was only poorly effective. The most potent RIPK2 inhibitors were the US Food and Drug Administration-approved drugs ponatinib and regorafenib. Their mechanism of action was independent of NOD2 interaction and involved loss of downstream kinase activation as evidenced by lack of RIPK2 autophosphorylation. Notably, these molecules also blocked RIPK2 ubiquitination and, consequently, inflammatory nuclear factor κB signaling. In monocytes, the inhibitors selectively blocked NOD-dependent tumor necrosis factor production without affecting lipopolysaccharide-dependent pathways. We also determined the first crystal structure of RIPK2 bound to ponatinib, and identified an allosteric site for inhibitor development. These results highlight the potential for type II inhibitors to treat indications of RIPK2 activation as well as inflammation-associated cancers.


Subject(s)
Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Amino Acid Sequence , Animals , Cells, Cultured , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Inflammation/metabolism , Models, Molecular , Molecular Sequence Data , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Protein Binding , Protein Kinase Inhibitors/chemistry , Pyridazines/chemistry , Pyridazines/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Sf9 Cells , Signal Transduction/drug effects , Ubiquitination/drug effects
16.
Sci Rep ; 5: 12018, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-26153766

ABSTRACT

Although Toll-like receptor 4 (TLR4)- and nucleotide-binding oligomerization domain 2 (NOD2)-mediated signaling mechanisms have been extensively studied individually, the crosstalk between them in the regulation of intestinal mucosal defense and tissue homeostasis has been underappreciated. Here, we uncover some novel activities of NOD2 by gene expression profiling revealing the global nature of the cross-regulation between TLR4- and NOD2-mediated signaling. Specifically, NOD2 is able to sense the intensity of TLR4-mediated signaling, resulting in either synergistic stimulation of Interluekin-12 (IL-12) production when the TLR signaling intensity is low; or in the inhibition of IL-12 synthesis and maintenance of intestinal mucosal homeostasis when the TLR signaling intensifies. This balancing act is mediated through receptor-interacting serine/threonine kinase 2, and the transcriptional regulator CCAAT/enhancer-binding protein α (C/EBPα) via its serine 248 phosphorylation by Protein Kinase C. Mice deficient in C/EBPα in the hematopoietic compartment are highly susceptible to chemically induced experimental colitis in an IL-12-dependent manner. Additionally, in contrast to the dogma, we find that the major Crohn's disease-associated NOD2 mutations could cause a primarily immunodeficient phenotype by selectively impairing TLR4-mediated IL-12 production and host defense. To restore the impaired homeostasis would be a way forward to developing novel therapeutic strategies for inflammatory bowel diseases.


Subject(s)
Enterocolitis/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Animals , Cell Line , Cluster Analysis , Colitis/genetics , Colitis/metabolism , Colitis/pathology , Cyclic AMP Response Element-Binding Protein/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Enterocolitis/genetics , Enterocolitis/immunology , Enterocolitis/pathology , Gene Expression Profiling , Gene Expression Regulation , Humans , Interleukin-12 Subunit p35/genetics , Interleukin-12 Subunit p35/metabolism , Lipopolysaccharides/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , NF-kappa B/metabolism , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/genetics , Promoter Regions, Genetic , Protein Binding , Protein Kinase C/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Transcription, Genetic
17.
Mol Immunol ; 64(2): 235-43, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25549945

ABSTRACT

Innate immunity is considered to be critical in the pathogenesis of fungal keratitis. Pattern recognition receptors (PRRs) recognize conserved microbial structures called pathogen-associated molecular patterns (PAMPS), thereby initiating the innate immunity. Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain (NOD)-like receptors (leucine-rich repeat-containing receptors, NLRs) are two major PRR families. The crosstalk between TLR2 and NOD2 is not completely understood, and their interrelationship in Aspergillus fumigates keratitis is still unclear. To our surprise, we found herein that NOD2 and TLR2 were increased by A. fumigatus conidia in immortalized human corneal epithelial cells (HCECs). In addition, NOD2 expression was up-regulated by its agonist muramyl dipeptide (MDP), along with receptor interacting protein 2 (RIP2), nuclear factor κB (NFκB)-p65, inhibitor of NFκB (IκB)-α, and multiple inflammatory cytokines, including interleukin-6 (IL-6), IL-8 and tumor necrosis factor α (TNF-α). Interestingly, zymosan, a TLR2 agonist, promoted the expression of NOD2 and RIP2 in a TLR2-dependent manner. Furthermore, we demonstrated that the increased expression of NOD2 and RIP2 caused by A. fumigatus conidia occurred in part through a TLR2-dependent pathway. However, zymosan pretreatment decreased NOD2 and RIP2 expression along with the MDP induced secretion of inflammatory cytokines in HCECs. In agreement, NOD2 knockdown by small interfering RNA (siRNA) reduced the release of IL-6, IL-8 and TNF-α induced by A. fumigatus conidia. These findings suggest the existence of complex interactions between TLR2 and NOD2 in HCECs inflammatory response against A. fumigatus infection.


Subject(s)
Aspergillus fumigatus/immunology , Epithelial Cells/metabolism , Nod2 Signaling Adaptor Protein/immunology , Receptor Cross-Talk , Spores, Fungal/immunology , Toll-Like Receptor 2/immunology , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Cell Line, Transformed , Cornea/immunology , Cornea/metabolism , Cornea/microbiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Gene Expression Regulation , Host-Pathogen Interactions , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-8/genetics , Interleukin-8/immunology , NF-KappaB Inhibitor alpha , Nod2 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/immunology , Signal Transduction , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/genetics , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Zymosan/pharmacology
18.
J Biol Chem ; 289(41): 28213-24, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25170077

ABSTRACT

NOD2 encodes an intracellular multidomain pattern recognition receptor that is the strongest known genetic risk factor in the pathogenesis of Crohn disease (CD), a chronic relapsing inflammatory disorder of the intestinal tract. NOD2 functions as a sensor for bacterial cell wall components and activates proinflammatory and antimicrobial signaling pathways. Here, using a genome-wide small interfering RNA (siRNA) screen, we identify numerous genes that regulate secretion of the proinflammatory cytokine IL-8 in response to NOD2 activation. Moreover, many of the identified IL-8 regulators are linked by protein-protein interactions, revealing subnetworks of highly connected IL-8 regulators implicated in processes such as vesicle formation, mRNA stability, and protein ubiquitination and trafficking. A TNFα counterscreen to induce IL-8 secretion in an NOD2-independent manner reveals that the majority of the identified regulators affect IL-8 secretion irrespective of the initiating stimuli. Using immortalized macrophages, we validate the ubiquitin protease, USP8, and the endosomal sorting protein, VPS28, as negative regulators of NOD2-induced cytokine secretion. Interestingly, several genes that affect NOD2-induced IL-8 secretion are present in loci associated with CD risk by genome-wide association studies, supporting a role for the NOD2/IL-8 pathway, and not just NOD2, in the pathogenesis of CD. Overall, this screen provides a valuable resource in the advancement of our understanding of the genes that regulate the secretion of IL-8.


Subject(s)
Crohn Disease/genetics , Endopeptidases/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Interleukin-8/genetics , Macrophages/metabolism , Nod2 Signaling Adaptor Protein/genetics , RNA, Small Interfering/genetics , Ubiquitin Thiolesterase/genetics , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Line, Transformed , Crohn Disease/metabolism , Crohn Disease/pathology , Endopeptidases/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Genetic Loci , Genome-Wide Association Study , HEK293 Cells , High-Throughput Screening Assays , Humans , Interleukin-8/agonists , Interleukin-8/antagonists & inhibitors , Interleukin-8/metabolism , Macrophages/pathology , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/metabolism , Protein Interaction Mapping , Protein Transport , RNA Stability , RNA, Small Interfering/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitination
19.
J Med Chem ; 57(16): 6897-918, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-24707857

ABSTRACT

Dysregulation of nucleotide-binding oligomerization domains 1 and 2 (NOD1 and NOD2) has been implicated in the pathology of various inflammatory disorders, rendering them and their downstream signaling proteins potential therapeutic targets. Selective inhibition of NOD1 and NOD2 signaling could be advantageous in treating many acute and chronic diseases; therefore, harnessing the full potential of NOD inhibitors is a key topic in medicinal chemistry. Although they are among the best studied NOD-like receptors (NLRs), the therapeutic potential of pharmacological modulation of NOD1 and NOD2 is largely unexplored. This review is focused on the scientific progress in the field of NOD inhibitors over the past decade, including the recently reported selective inhibitors of NOD1 and NOD2. In addition, the potential approaches to inhibition of NOD signaling as well as the advantages and disadvantages linked with inhibition of NOD signaling are discussed. Finally, the potential directions for drug discovery are also discussed.


Subject(s)
Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/metabolism , Signal Transduction , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Asthma/drug therapy , Asthma/etiology , Crohn Disease/drug therapy , Crohn Disease/metabolism , Drug Discovery/methods , Humans , Immunity, Innate/drug effects , Molecular Targeted Therapy/methods , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/genetics , Polymorphism, Genetic
20.
Cell Physiol Biochem ; 32(6): 1857-66, 2013.
Article in English | MEDLINE | ID: mdl-24356239

ABSTRACT

BACKGROUND/AIMS: Although the pathogenesis of myocardial infarction (MI) is multifactorial, activation of innate immune system to induce inflammation has emerged as a key pathophysiological process in MI. NOD2, one member of the NOD-like receptor (NLR) family, plays an important role in the innate immune response. This study was to examine the role of NOD2 during MI. METHODS: MI was induced by permanent ligation of the left coronary artery in wild type and NOD2(-/-) mice and cardiac fibroblasts were isolated. RESULTS: NOD2 expression was significantly increased in myocardium in post-MI mice. NOD2 deficiency improved cardiac dysfunction and remodeling after MI as evidenced by echocardiographic analysis, reduced the levels of cytokines, inflammatory cell infiltration and matrix metalloproteinase-9 (MMP-9) activity. In vitro, we further found that NOD2 activation induced the activation of MAPK signaling pathways, production of proinflammatory mediators and MMP-9 activity in cardiac fibroblasts. CONCLUSIONS: Our studies demonstrate that NOD2 is a critical component of a signal transduction pathway that links cardiac injury by exacerbation of inflammation and MMP-9 activity. Pharmacological targeting of NOD2-mediated signaling pathways may provide a novel approach to treatment of cardiovascular diseases.


Subject(s)
Nod2 Signaling Adaptor Protein/metabolism , Ventricular Remodeling , Animals , Cell Hypoxia , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Echocardiography , Fibroblasts/cytology , Fibroblasts/metabolism , MAP Kinase Signaling System , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Nod2 Signaling Adaptor Protein/genetics , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL