Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters











Publication year range
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.
Fish Shellfish Immunol ; 149: 109566, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636735

ABSTRACT

Fish rely on innate immune system for immunity, and nucleotide-binding oligomerization domain-like receptors (NLRs) are a vital group of receptor for recognition. In the present study, NOD1 gene was cloned and characterized from golden pompano Trachinotus ovatus, a commercially important aquaculture fish species. The ORF of T. ovatus NOD1 was 2820 bp long, encoding 939 amino acid residues with a highly conserved domains containing CARD-NACHT-LRRs. Phylogenetic analysis revealed that the T. ovatus NOD1 clustered with those of fish and separated from those of birds and mammals. T. ovatus NOD1 has wide tissue distribution with the highest expression in gills. Bacterial challenges (Streptococcus agalactiae and Vibrio alginolyticus) significantly up-regulated the expression of NOD1 with different response time. The results of T. ovatus NOD1 ligand recognition and signaling pathway analysis revealed that T. ovatus NOD1 could recognize iE-DAP at the concentration of ≧ 100 ng/mL and able to activate NF-κB signaling pathway. This study confirmed that NOD1 play a crucial role in the innate immunity of T. ovatus. The findings of this study improve our understanding on the immune function of NOD1 in teleost, especially T. ovatus.


Subject(s)
Amino Acid Sequence , Fish Diseases , Fish Proteins , Immunity, Innate , Nod1 Signaling Adaptor Protein , Phylogeny , Sequence Alignment , Vibrio alginolyticus , Animals , Nod1 Signaling Adaptor Protein/genetics , Nod1 Signaling Adaptor Protein/immunology , Nod1 Signaling Adaptor Protein/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/chemistry , Immunity, Innate/genetics , Fish Diseases/immunology , Sequence Alignment/veterinary , Vibrio alginolyticus/physiology , Streptococcal Infections/immunology , Streptococcal Infections/veterinary , Streptococcus agalactiae/physiology , Gene Expression Regulation/immunology , Gene Expression Profiling/veterinary , Vibrio Infections/immunology , Vibrio Infections/veterinary , Diaminopimelic Acid/chemistry , Diaminopimelic Acid/analogs & derivatives , Perciformes/immunology , Perciformes/genetics , Fishes/immunology , Fishes/genetics
3.
J Oral Biosci ; 66(1): 105-111, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38182046

ABSTRACT

OBJECTIVES: During innate immune defense, host pattern recognition receptors, including toll-like receptors and nucleotide-binding oligomerization domain-like receptors (NLRs), can activate downstream pathways by recognizing pathogen-associated molecular patterns produced by microorganisms, triggering immune responses. NOD1, an important cell membrane protein in the NLR-like receptor protein family, exerts anti-infective effects through γ-D-glutamyl-meso-diaminopimelic acid (iE-DAP) recognition. Oral epithelial cells resist bacterial invasion through iE-DAP-induced interleukin (IL)-8 production, recruiting neutrophils to sites of inflammation in response to bacterial threats to periodontal tissues. To date, the regulatory mechanisms of iE-DAP in gingival epithelial cells (GECs) are poorly understood. This study was conducted to investigate the role of the NOD1 pathway in the development of periodontitis by examining the effect of iE-DAP on IL-8 production in Ca9-22 cells. METHODS: IL-8 production by iE-DAP-stimulated-Ca9-22 cells was assessed using an enzyme-linked immunosorbent assay. Phosphorylation levels of intracellular signaling molecules were evaluated using western blot analyses. RESULTS: iE-DAP induced NOD1 receptor expression in Ca9-22 cells. Additionally, iE-DAP induced expression of pro-IL-1ß protein without extracellular secretion. Our results suggest that iE-DAP regulates IL-8 production by activating p38 mitogen-activated protein kinase (MAPK) and ERK1/2 signaling pathways. iE-DAP also promoted nuclear factor kappa-B p65 phosphorylation, facilitating its nuclear translocation. Notably, p38 MAPK and ERK1/2 inhibitors suppressed iE-DAP-stimulated IL-8 production, suggesting that JNK is not involved in this mechanism. CONCLUSIONS: Our results indicate that p38 MAPK and ERK1/2, but not JNK, are involved in innate immune responses in GECs.


Subject(s)
Diaminopimelic Acid/analogs & derivatives , Interleukin-8 , Periodontitis , Humans , Nucleotides , p38 Mitogen-Activated Protein Kinases
4.
J Biomol Struct Dyn ; 40(16): 7483-7495, 2022 10.
Article in English | MEDLINE | ID: mdl-33710949

ABSTRACT

Nucleotide-binding and oligomerization domain (NOD)-like receptors (NLRs) are cytosolic receptors implicated in recognition of intracellular pathogen associated molecular patterns (PAMPs) and danger associated molecular patterns (DAMPs). Depending upon their effector binding domain (EBD) at the C-terminal, the NLRs are categorized into NLRA, NLRB, NLRC, NLRP and NLRX. NOD1 is a pivotal player in immune responses against bacterial and viral invasions and interacts with pathogens via C-terminal leucine rich repeat (LRR) domain. This study aims at characterizing NOD1 in an economically important teleost of the Indian subcontinent, spotted snakehead Channa punctata. The understanding of pathogen-receptor interaction in teleosts is still obscure. In light of this, combinatorial approach involving protein modeling, docking, MD simulation and binding free energy calculation were employed to identify key motifs involved in binding iE-DAP. In silico analysis revealed that NOD1 consists of 943 amino acids comprising of one caspase recruitment domain (CARD) at N-terminal, one central NACHT domain and nine leucine rich repeat (LRR) regions at C-terminal. Structural dynamics study showed that the C-terminal ß-sheet LRR4-7 region is involved in iE-DAP binding. NOD1 was ubiquitously and constitutively expressed in all tissues studied. Differential expression profile of NOD1 induced by Aeromonas hydrophila infection was also investigated. Lymphoid organs and phagocytes of infected spotted snakehead showed significant downregulation of NOD1 expression. The current study thus gives an insight into structural and functional dynamics of NOD1 which might have future prospect for structure-based drug designing in teleosts.Communicated by Ramaswamy H. Sarma.


Subject(s)
Computational Biology , Nod1 Signaling Adaptor Protein , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/chemistry , Diaminopimelic Acid/metabolism , Leucine/chemistry , Nod1 Signaling Adaptor Protein/chemistry , Nod1 Signaling Adaptor Protein/metabolism
5.
Drug Deliv ; 28(1): 1020-1030, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34060399

ABSTRACT

NOD1 is an intracellular receptor that, when activated, induces gene expression of pro-inflammatory factors promoting macrophages and neutrophils recruitment at the infection site. However, iE-DAP, the dipeptide agonist that promotes this receptor's activation, cannot permeate cell membranes. To develop a nanocarrier capable of achieving a high and prolonged activation over time, iE-DAP was encapsulated in nanoparticles (NPs) made of poly (3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV). The physicochemical properties, colloidal stability, encapsulation efficiency, and cellular uptake of iE-DAP-loaded PHVB NPs were analyzed. Results evidenced that physicochemical properties of iE-DAP-loaded NPs remained stable over time, and NPs were efficiently internalized into cells, a process that depends on time and concentration. Moreover, our results showed that NPs elicited a controlled cargo release in vitro, and the encapsulated agonist response was higher than its free form, suggesting the possibility of activating intracellular receptors triggering an immune response through the release of NOD1 agonist.


Subject(s)
Diaminopimelic Acid/analogs & derivatives , Nanoparticles/chemistry , Nod1 Signaling Adaptor Protein/drug effects , Animals , Cell Survival/drug effects , Chemistry, Pharmaceutical , Diaminopimelic Acid/administration & dosage , Diaminopimelic Acid/pharmacology , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Liberation , Drug Stability , Mice , Polyesters/chemistry , RAW 264.7 Cells
6.
J Sci Food Agric ; 101(3): 1218-1227, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32789879

ABSTRACT

BACKGROUND: Long-term high-concentrate (HC) diet feeding increased bacterial endotoxins, which translocated into the mammary glands of dairy goats and induced inflammatory response. γ-d-Glutamyl-meso-diaminopimelic acid (iE-DAP), bacterial peptidoglycan component, triggered inflammatory response through activating nucleotide oligomerization domain protein 1 (NOD1) signaling pathway. While dietary supplemented with sodium butyrate (SB) relieved inflammatory response and improved animal health and production. To investigate the effects and the mechanisms of action of SB on the inflammatory response in the mammary glands of dairy goats fed HC diet, 12 Saanen dairy goats were randomly assigned into HC group and SB regulated (BHC) group. RESULTS: The results showed that SB supplementation attenuated ruminal pH decrease caused by HC diet in dairy goats resulting in a decrease of proinflammatory cytokines and iE-DAP plasma concentration and the mRNA expression of NOD1 and other inflammation-related genes. The protein levels of NOD1, NF-κB p65 and NF-κB pp65 were decreased by the SB supplementation. The expression of histone deacetylase 3 (HDAC3) was also inhibited by the SB supplementation. Meanwhile, the chromatin compaction ratios and DNA methylation levels of NOD1 and receptor-interacting protein 2 (RIP2) of BHC group were upregulated. CONCLUSION: Collectively, the SB supplementation mitigated the inflammatory response in the mammary glands of dairy goats during HC-induced subacute ruminal acidosis (SARA) by inhibiting the activation of the NOD1/NF-κB signaling pathway through the decrease of the iE-DAP concentration in the rumen fluid and plasma and HDAC3 expression. DNA methylation and chromatin remodeling also contributed to the anti-inflammatory effect of SB. © 2020 Society of Chemical Industry.


Subject(s)
Butyric Acid/administration & dosage , Diaminopimelic Acid/analogs & derivatives , Goat Diseases/drug therapy , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/immunology , Acidosis/drug therapy , Acidosis/immunology , Acidosis/veterinary , Animal Feed/adverse effects , Animal Feed/analysis , Animals , Diaminopimelic Acid/adverse effects , Diaminopimelic Acid/analysis , Diet/adverse effects , Diet/veterinary , Dietary Supplements/analysis , Female , Goat Diseases/immunology , Goats/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Nod1 Signaling Adaptor Protein/immunology
7.
J Dairy Sci ; 104(1): 776-785, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33189269

ABSTRACT

Nucleotide-binding oligomerization domain (NOD)-like receptor 1 (NOD1) is a cytosolic pattern recognition receptor with a crucial role in the innate immune response of cells triggered by the presence of compounds such as gamma-d-glutamyl-meso-diaminopimelic acid (iE-DAP) present in the peptidoglycan of all gram-negative and certain gram-positive bacteria. Methionine (Met) and arginine (Arg) are functional AA with immunomodulatory properties. In the present study, we aimed to assess the effect of increased Met and Arg supply on mRNA abundance of genes associated with innate immune response, antioxidant function, and AA metabolism during iE-DAP challenge in bovine mammary epithelial cells (BMEC). Primary BMEC (n = 4 per treatment) were precultured in modified medium for 12 h with the following AA formulations: ideal profile of AA (control), increased Met supply (incMet), increased Arg supply (incArg), or increased supply of Met plus Arg (incMetArg). Subsequently, cells were challenged with or without iE-DAP (10 µg/mL) for 6 h. Data were analyzed as a 2 × 2 × 2 factorial using the MIXED procedure of SAS 9.4. Greater mRNA abundance of NOD1, the antioxidant enzyme SOD1, and AA transporters (SLC7A1 and SLC3A2) was observed in the incMet cells after iE-DAP stimulation. Although increased Met alone had no effect, incMetArg led to greater abundance of the inflammatory cytokine IL-6, and the antioxidant enzyme GPX1 after iE-DAP stimulation. The increased Arg alone downregulated NOD1 after iE-DAP stimulation, coupled with a downregulation in the AA transporters mRNA abundance (SLC7A1, SLC7A5, SLC3A2, and SLC38A9), and upregulation in GSS and KEAP1 mRNA abundance. Overall, the data indicated that increased supply of both Met and Arg in the culture medium were more effective in modulating the innate immune response and antioxidant capacity of BMEC during in vitro iE-DAP stimulation.


Subject(s)
Arginine/administration & dosage , Cattle , Diaminopimelic Acid/analogs & derivatives , Immunity, Innate/genetics , Mammary Glands, Animal/drug effects , Methionine/administration & dosage , Amino Acid Transport Systems/genetics , Animals , Antioxidants/metabolism , Cells, Cultured , Diaminopimelic Acid/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Gene Regulatory Networks/drug effects , Immunity, Innate/drug effects , Nod1 Signaling Adaptor Protein/genetics , RNA, Messenger/analysis , Superoxide Dismutase-1/genetics
8.
J Cell Physiol ; 236(7): 5212-5234, 2021 07.
Article in English | MEDLINE | ID: mdl-33368240

ABSTRACT

Autophagy is a crucial cellular homeostatic process and an important part of the host defense system. Dysfunction in autophagy enhances tissue susceptibility to infection and multiple diseases. However, the role of nucleotide oligomerization domain 1 (NOD1) in autophagy in bovine hepatocytes is not well known. Therefore, our aim was to study the contribution of NOD1 to autophagy during inflammation in response to a specific ligand γ-d-glutamyl-meso-diaminopimelic acid (iE-DAP). To achieve this aim, hepatocytes separated from cows at ∼160 days in milk (DIM) were divided into six groups: the nontreated control (CON) group, the rapamycin-treated (RAP) group as a positive control, the iE-DAP-treated (DAP) group, the 3-MA-treated (MA) group, the rapamycin with 3-MA (RM) group, and the iE-DAP with 3-MA (DM) group. iE-DAP administration significantly increased the mRNA expression of NOD1, ATG16L1, RIPK2, ULK1, AMBRA1, DFCP1, WIPI1, ATG5, ATG7, ATG10, ATG4A, IκBα, NF-κB, CXCL1, IL-8, and STAT6 and significantly decreased PIK3C3. The protein expression of NOD1, p-IκBα, p-NF-κB/p-p65, LC3-II, ATG5, and beclin 1 were significantly upregulated and that of SQSTM1/p62, p-mTOR, and FOXA2 were significantly downregulated in response to iE-DAP. iE-DAP also induced the formation of LC3-GFP autophagic puncta in bovine hepatocytes. We also knocked down the NOD1 with siRNA. NOD1 silencing suppressed the autophagy and inflammation-related genes and proteins. The application of the autophagy inhibitor increased the expression of inflammatory molecules and alleviated autophagy-associated molecules. Taken together, these findings suggest that NOD1 is a key player for regulating both ATG16L1 and RIPK2-ULK1 directed autophagy during inflammation in response to iE-DAP in bovine hepatocytes.


Subject(s)
Autophagy/drug effects , Diaminopimelic Acid/analogs & derivatives , Hepatocytes/metabolism , Inflammation/pathology , Nod1 Signaling Adaptor Protein/metabolism , Animals , Autophagosomes/metabolism , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Cattle , Cell Survival/physiology , Cells, Cultured , Diaminopimelic Acid/pharmacology , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism
9.
J Dairy Sci ; 104(2): 2123-2139, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33358155

ABSTRACT

Glutamine (GLN) has many types of biological activity in rats, including anti-inflammatory, antioxidative stress, and anti-apoptosis effects. However, little is known about the effects of GLN on bovine mammary epithelial cells (BMEC). γ-d-Glutamyl-meso-diaminopimelic acid (iE-DAP) is a cell wall peptidoglycan component of gram-negative bacteria that can be recognized by the intracellular receptor nucleotide-binding oligomerization domain-containing protein 1 (NOD1) and can cause bovine mastitis. The goal of the present study was to investigate whether GLN protects BMEC from iE-DAP-induced inflammation, oxidative stress, and apoptosis. We cultured BMEC in a GLN-free medium for 24 h and then separated them into 4 groups: cells treated with 1× PBS for 26 or 32 h (control); cells stimulated by 10 µg/mL iE-DAP for 2 or 8 h (2- or 8-h iE-DAP); cells pretreated with 8 or 4 mM GLN for 24 h followed by 2 or 8 h of 1× PBS treatment (8 or 4 mM GLN); and cells pretreated with 8 or 4 mM GLN for 24 h followed by 2 or 8 h of iE-DAP treatment (DG). In the 2-h iE-DAP group, when levels of inflammation peaked, iE-DAP treatment increased both the mRNA and protein expression of NOD1, inhibitor of nuclear factor-κB (NFKBIA, IκB), and nuclear factor-κB subunit p65 (RELA, NF-κB p65), as well as the mRNA expression of IL6 and IL8 and levels of IL-6 and tumor necrosis factor-α in cell culture supernatants. In contrast, 8 mM GLN pretreatment inhibited the mRNA and protein expression of inflammatory-related factors by suppressing the NOD1/NF-κB pathway. In the 8-h iE-DAP group, iE-DAP treatment decreased the mRNA and protein expression of extracellular regulated kinase (Erk, ERK) and nuclear factor erythroid 2-associated factor2 (NFE2L2, Nrf2), as well as the mRNA expression of superoxide dismutase 1 (SOD1), catalase (CAT), coenzyme II oxidoreductase 1 (NQO1), and heme oxygenase 1 (HMOX1, HO1). In addition, iE-DAP treatment increased the expression of malondialdehyde in BMEC when oxidative stress levels peaked. Interestingly, 4 mM GLN pretreatment induced the mRNA and protein expression of antioxidative stress-related factors and inhibited the expression of reactive oxygen species in BMEC by promoting the ERK/Nrf2 pathway. Moreover, GLN reduced apoptosis caused by inflammation and oxidative stress in BMEC. This is the first report showing that GLN protects against iE-DAP-induced inflammation and oxidative stress via the NOD1/NF-κB and ERK/Nrf2 pathways in BMEC.


Subject(s)
Cattle Diseases/prevention & control , Diaminopimelic Acid/analogs & derivatives , Glutamine/therapeutic use , Inflammation/veterinary , Mammary Glands, Animal/drug effects , Animals , Apoptosis/drug effects , Cattle , Cells, Cultured , Diaminopimelic Acid/antagonists & inhibitors , Epithelial Cells/metabolism , Female , Heme Oxygenase-1/metabolism , Inflammation/chemically induced , Inflammation/prevention & control , Mammary Glands, Animal/cytology , NF-kappa B/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Transcription Factor RelA/metabolism , Transcription Factor RelA/pharmacology
10.
Molecules ; 25(22)2020 Nov 10.
Article in English | MEDLINE | ID: mdl-33182604

ABSTRACT

The dipeptide d-Glu-meso-DAP (iE-DAP) is the minimal structural fragment capable of activating the innate immune receptor nucleotide-binding oligomerization domain protein (NOD1). The meso-diaminopimelic acid (meso-DAP) moiety is known to be very stringent in terms of the allowed structural modifications which still retain the NOD1 activity. The aim of our study was to further explore the chemical space around the meso-DAP portion and provide a deeper understanding of the structural features required for NOD1 agonism. In order to achieve the rigidization of the terminal amine functionality of meso-DAP, isoxazoline and pyridine heterocycles were introduced into its side-chain. Further, we incorporated the obtained meso-DAP mimetics into the structure of iE-DAP. Collectively, nine innovative iE-DAP derivatives additionally equipped with lauroyl or didodecyl moieties at the α-amino group of d-Glu have been prepared and examined for their NOD1 activating capacity. Overall, the results obtained indicate that constraining the terminal amino group of meso-DAP abrogates the compounds' ability to activate NOD1, since only compound 6b retained noteworthy NOD1 agonistic activity, and underpin the stringent nature of this amino acid with regard to the allowed structural modifications.


Subject(s)
Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/chemical synthesis , Immunity, Innate , NF-kappa B p50 Subunit/chemistry , Nod1 Signaling Adaptor Protein/chemistry , Cell Proliferation , Chemistry Techniques, Synthetic , Esters/chemistry , Humans , Isoxazoles/chemistry , Molecular Conformation , Protein Conformation , Pyridines/chemistry
11.
ACS Chem Biol ; 15(11): 2966-2975, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33078931

ABSTRACT

Bacterial cell walls contain peptidoglycan (PG), a scaffold that provides proper rigidity to resist lysis from internal osmotic pressure and a barrier to protect cells against external stressors. It consists of repeating sugar units with a linkage to a stem peptide that becomes cross-linked by cell wall transpeptidases (TP). While synthetic PG fragments containing l-lysine in the third position on the stem peptide are easier to access, those with meso-diaminopimelic acid (m-DAP) pose a severe synthetic challenge. Herein, we describe a solid phase synthetic scheme based on widely available building blocks to assemble meso-cystine (m-CYT), which mimics key structural features of m-DAP. To demonstrate proper mimicry of m-DAP, cell wall probes were synthesized with m-CYT in place of m-DAP and evaluated for their metabolic processing in live bacterial cells. We found that m-CYT-based cell wall probes were properly processed by TPs in various bacterial species that endogenously contain m-DAP in their PG. Additionally, we have used hybrid quantum mechanical/molecular mechanical (QM/MM) and molecular dynamics (MD) simulations to explore the influence of m-DAP analogs on the PG cross-linking. The results showed that the cross-linking mechanism of transpeptidases occurred through a concerted process. We anticipate that this strategy, which is based on the use of inexpensive and commercially available building blocks, can be widely adopted to provide greater accessibility of PG mimics for m-DAP containing organisms.


Subject(s)
Bacteria/metabolism , Cell Wall/metabolism , Cystine/metabolism , Diaminopimelic Acid/metabolism , Bacteria/chemistry , Cell Wall/chemistry , Cystine/analogs & derivatives , Cystine/chemical synthesis , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/chemical synthesis , Mycobacterium smegmatis/metabolism , Peptidoglycan
12.
Int Immunopharmacol ; 83: 106392, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32182568

ABSTRACT

The anti-inflammatory effects of sodium valproate (VPA) in vivo and in vitro have been demonstrated in recent studies. The aim of this study was to evaluate whether VPA can suppress inflammation in bovine mammary epithelial cells (BMECs) stimulated by γ-D-glutamyl-meso-diaminopimelic acid (iE-DAP). First, the concentration and treatment points of iE-DAP and VPA were optimized. Then, BMECs were cultured in complete media and separated into four groups: untreated control cells (CON group), cells stimulated by 10 µg/mL iE-DAP for 6 h (DAP group), cells stimulated by 0.5 mmol/L VPA for 6 h (VPA group), and cells pretreated with VPA (0.5 mmol/L) for 6 h followed by 10 µg/mL of iE-DAP for 6 h (VD group). The results showed that the level of  interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in the culture medium increased in the iE-DAP-treated cells and that pretreatment with VPA reversed this increase. iE-DAP increased both mRNA and protein expression levels of nucleotide-binding oligomerization domain-containing protein 1 (NOD1) and receptor-interacting protein kinas (RIPK2) and activated inhibitor of NF-κB (IκB) and nuclear factor-kappa B p65 (NF-κB p65) through phosphorylation. Upon activation of the NF-κB pathway, the expression of the pro-inflammatory cytokines IL-6, interleukin-8 (IL-8) and interleukin-1ß (IL-1ß), the acute phase protein serum amyloid A 3 (SAA3) and the lingual antimicrobial peptide (LAP) but not  haptoglobi (HP) or bovine neutrophil beta defensing 5 (BNBD5) were increased in the DAP group. The VPA pretreatment induced the acetylation of signal transducers and activators of transcription(STAT1) and histone 3 (H3) by inhibiting histone deacetylase (HDAC) and then suppressed the NF-κB pathway. Moreover, VPA induced autophagy and reduced apoptosis in BMECs in the VD group. These results suggested that VPA treatment can attenuate the inflammatory response induced by iE-DAP.


Subject(s)
Epithelial Cells/physiology , Histones/metabolism , Inflammation/drug therapy , Mastitis, Bovine/drug therapy , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Valproic Acid/pharmacology , Acetylation , Animals , Apoptosis , Autophagy , Cattle , Cells, Cultured , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/metabolism , Female , Protein Processing, Post-Translational , STAT1 Transcription Factor/metabolism , Signal Transduction
13.
Org Lett ; 22(6): 2313-2317, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32133861

ABSTRACT

The syntheses of peptidoglycan (PG)-derived peptides containing meso-diaminopimelic acid (meso-Dap) are typically quite lengthy due to the need to prepare orthogonally protected meso-Dap. In this work, the preparation of the PG pentapeptide containing the isosteric analog meso-oxa-Dap is described. The synthesis relies on the ring opening of a peptide embedded aziridine via the attack of a serine residue. The pentapeptide was attached to a GlcNAc-anhydro-MurNAc disaccharide, to produce a putative substrate for the AmpG pore protein.


Subject(s)
Acetylglucosamine/chemistry , Diaminopimelic Acid/analogs & derivatives , Disaccharides/chemistry , Muramic Acids/chemistry , Oligopeptides/chemical synthesis , Peptidoglycan/chemistry , Diaminopimelic Acid/chemical synthesis , Diaminopimelic Acid/chemistry , Oligopeptides/chemistry , Oxazoles/chemical synthesis , Oxazoles/chemistry
14.
J Mol Neurosci ; 70(4): 600-609, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31907866

ABSTRACT

The purpose of the study was studying the influence of different NOD agonists on the morphological phenotype of primary murine microglia and to examine their influence on characteristic cytokines. Primary CD11b-positive cells were isolated from the brain of neonatal mice. The microglial phenotype of the cells was examined by ionized calcium-binding adapter molecule (Iba)1 staining. After14 days in culture, these cells were stimulated by iE-DAP, L18-MDP, or M-TriDAP as NOD1, NOD2, and NOD1/2 agonists, respectively. The cellular morphology was recorded and compared to the phenotype of cells cultured in medium alone or after LPS stimulation. The cells developed a specific phenotype only after treatment with the NOD2 agonist L18-MDP. These cells were characterized by straight extensions carrying tiny spikes and had a high ramification index. This was in sharp contrast to all other treatments, which always resulted in an amoeboid phenotype typically shown by activated microglia in vivo and by cultured microglia in vitro. The staining intensity of IL-6 and TNF-α did not reveal any clear difference independent of the NOD agonist treatment. In contrast, an increased staining intensity was observed for IL-10 after L18-MDP treatment. The NOD2 agonist L18-MDP induced a morphologically distinct phenotype characterized by microspike-decorated dendritiform extensions and a high degree of ramification in primary murine microglia. Increased ramification index and elevated staining intensity of anti-inflammatory IL-10 as hallmarks suggest that a M2-like phenotype of microglia was induced.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Adjuvants, Immunologic/pharmacology , Diaminopimelic Acid/analogs & derivatives , Microglia/drug effects , Nod1 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/agonists , Phenotype , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Shape , Cell Surface Extensions/drug effects , Cells, Cultured , Diaminopimelic Acid/pharmacology , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microglia/cytology , Microglia/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
15.
Cell Biochem Funct ; 37(7): 464-473, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31396989

ABSTRACT

We sought to explore the functions and modulated factors of NOD1 in normal decidual stromal cells (DSCs) derived from the first trimester pregnancy and whether existed different expression of NOD1 between normal and unexplained recurrent pregnancy loss (URPL) in DSCs. Twenty-six patients with normal pregnancies that required abortion and 12 URPL patients at first trimester were enrolled for the study. As a result, we found lower levels of NOD1 in the DSCs derived from URPL compared with those from normal early trimester pregnancy. Furthermore, increased NOD1 expression in the normal DSCs induced apoptosis and increased monocyte chemotactic protein-1 (MCP-1) and IL-1ß (interleukin 1 beta) secretion but decreased their invasion capacity. In addition, several cytokines such as IL-1ß, tumour necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), and interleukin-17 (IL-17) were present at the maternal-fetal interface in RPL and were found to regulate NOD1 expression in primary DSCs. Our study indicates that RPL may be associated with NOD1 aberrant expression in DSCs, which plays a significant role in maintaining pregnancy via infection control and regulation of immune responses that might affect the pregnancy outcome. We expect that our results will bring more comprehensively understanding about the connection between NOD1 and RPL for researchers.


Subject(s)
Decidua/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Pregnancy Trimester, First/metabolism , Stromal Cells/metabolism , Adult , Apoptosis/drug effects , Cells, Cultured , Cytokines/analysis , Cytokines/metabolism , Decidua/cytology , Decidua/drug effects , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/pharmacology , Female , Humans , Nod1 Signaling Adaptor Protein/genetics , Pregnancy , Pregnancy Trimester, First/drug effects , Stromal Cells/drug effects
16.
Life Sci ; 229: 173-179, 2019 Jul 15.
Article in English | MEDLINE | ID: mdl-31103606

ABSTRACT

AIMS: The innate immune response induced by bacterial peptidoglycan peptides, such as γ-d-glutamyl-meso-diaminopimelic acid (iE-DAP), is an important host defense system. However, little is known about the innate immune response in the lung alveolar region. In this study, we examined induction of the innate immune response by iE-DAP in human alveolar epithelial cell lines, NCI-H441 (H441) and A549. MAIN METHODS: Induction of the innate immune response was evaluated by measuring the mRNA expression of cytokines and their release into the culture medium. KEY FINDINGS: iE-DAP treatment increased the mRNA expression of interleukin (IL)-6 and IL-8, and increased release of these pro-inflammatory cytokines into the culture medium in H441 cells, but not in A549 cells. Lack of release of these cytokines in A549 cells may have been due to lack of peptide transporter 2 (PEPT2) function. Intracellular nucleotide-binding oligomerization domain 1 (NOD1) recognizes iE-DAP and activates downstream signaling pathways to initiate the immune response. Therefore, the role of mitogen-activated protein kinase (MAPK) signaling pathways was examined in H441 cells. As a result of inhibition studies, receptor-interacting serine/threonine-protein kinase 2 and MAPK signaling pathways, such as p38 MAPK and extracellular signal-regulated kinase, but not c-Jun N-terminal kinase, were determined to be involved in the innate immune response in H441 cells. In addition, the nuclear factor κB pathway also played a role in the innate immune response. SIGNIFICANCE: These findings indicated that the innate immune response induced by bacterial peptides could occur in a PEPT2- and NOD1-dependent manner in alveolar epithelial cells.


Subject(s)
Alveolar Epithelial Cells/immunology , Diaminopimelic Acid/analogs & derivatives , Immunity, Innate/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Symporters/metabolism , A549 Cells , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Cytokines/metabolism , Diaminopimelic Acid/pharmacology , Humans , Immunity, Innate/drug effects , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Signal Transduction , Symporters/genetics
17.
J Cell Physiol ; 234(11): 19602-19620, 2019 11.
Article in English | MEDLINE | ID: mdl-30941762

ABSTRACT

Nucleotide oligomerization domain protein-1 (NOD1), a cytosolic pattern recognition receptor for the γ-D-glutamyl-meso-diaminopimelic acid (iE-DAP) is associated with the inflammatory diseases. Very little is known how bovine hepatocytes respond to specific ligands of NOD1 and sodium butyrate (SB). Therefore, the aim of our study was to investigate the role of bovine hepatocytes in NOD1-mediated inflammation during iE-DAP or LPS treatment or SB pretreatment. To achieve this aim, hepatocytes separated from cows at ∼160 days in milk (DIM) were divided into six groups: The nontreated control group (CON), the iE-DAP-treated group (DAP), the lipopolysaccharide-treated group (LPS), iE-DAP with SB group (DSB), LPS with SB group (LSB), and the SB group. Both iE-DAP and LPS highly increased the expression of both NOD1 and RIPK2, the two key factors for the immune response in hepatocytes. IκBα, NF-κB/p65, and MAP kinases (ERK, JNK, and p38) were activated through phosphorylation. The activation of NF-κB and MAPK pathway consequently increased the proinflammatory cytokines, IL-6, TNF-α, IL-8, and IFN-γ and the chemokines CCL5, CCL20, and CXCL-10. Both treatments improved iNOS/NOS2 expression. However, iE-DAP was failed to express acute phase protein SAA3, but HP and LPS HP but SAA3. These ligands also increased LRRK2, TAK1, TAB1, and ß-defensins expression. The SB pretreatment at lower dose restored the function of hepatocytes by suppressing these increased molecules, as HDAC3 was inhibited. The activated NOD1 negatively regulated the expression of FOXA2. Altogether these data suggest an important role of bovine hepatocytes to promote immune responses via NOD1 expression during infection in the liver and a key role of SB to attenuate inflammation.


Subject(s)
Butyric Acid/pharmacology , Hepatocytes/drug effects , Inflammation/drug therapy , Nod1 Signaling Adaptor Protein/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Animals , Cattle , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/pharmacology , Hepatocytes/pathology , Inflammation/chemically induced , Inflammation/genetics , Ligands , Lipopolysaccharides/toxicity , Liver/drug effects , Liver/metabolism , NF-kappa B , Phosphorylation/drug effects
18.
ACS Chem Biol ; 14(3): 405-414, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30735346

ABSTRACT

The peptidoglycan fragments γ-d-glutamyl- meso-diaminopimelic acid (iE-DAP) and muramyl-dipeptide (MDP) are microbial-specific metabolites that activate intracellular pattern recognition receptors and stimulate immune signaling pathways. While extensive structure-activity studies have demonstrated that these bacterial cell wall metabolites trigger NOD1- and NOD2-dependent signaling, their direct binding to these innate immune receptors or other proteins in mammalian cells has not been established. To characterize these fundamental microbial metabolite-host interactions, we synthesized a series of peptidoglycan metabolite photoaffinity reporters and evaluated their cross-linking to NOD1 and NOD2 in mammalian cells. We show that active iE-DAP and MDP photoaffinity reporters selectively cross-linked NOD1 and NOD2, respectively, and not their inactive mutants. We also discovered MDP reporter cross-linking to Arf GTPases, which interacted most prominently with GTP-bound Arf6 and coimmunoprecipitated with NOD2 upon MDP stimulation. Notably, MDP binding to NOD2 and Arf6 was abrogated with loss-of-function NOD2 mutants associated with Crohn's disease. Our studies demonstrate peptidoglycan metabolite photoaffinity reporters can capture their cognate immune receptors in cells and reveal unpredicted ligand-induced interactions with other cellular cofactors. These photoaffinity reporters should afford useful tools to discover and characterize other peptidoglycan metabolite-interacting proteins.


Subject(s)
ADP-Ribosylation Factors/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/metabolism , Diaminopimelic Acid/analogs & derivatives , Peptidoglycan/metabolism , Receptors, Pattern Recognition/metabolism , Cell Wall/metabolism , Cytokines/metabolism , Diaminopimelic Acid/metabolism , HEK293 Cells , Humans , Ligands , Mutant Proteins/metabolism , Mutation , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Protein Binding , Signal Transduction , Structure-Activity Relationship
19.
Chembiochem ; 20(12): 1591-1598, 2019 06 14.
Article in English | MEDLINE | ID: mdl-30746833

ABSTRACT

The enzymes Csd6 and Pgp2 are peptidoglycan (PG) proteases found in the pathogenic bacteria Helicobacter pylori and Campylobacter jejuni, respectively. These enzymes are involved in the trimming of non-crosslinked PG sidechains and catalyze the cleavage of the bond between meso-diaminopimelic acid (meso-Dap) and d-alanine, thus converting a PG tetrapeptide into a PG tripeptide. They are known to be cell-shape-determining enzymes, because deletion of the corresponding genes results in mutant strains that have lost the normal helical phenotype and instead possess a straight-rod morphology. In this work, we report two approaches directed towards the synthesis of the tripeptide substrate Ac-iso-d-Glu-meso-oxa-Dap-d-Ala, which serves as a mimic of the terminus of an non-crosslinked PG tetrapeptide substrate. The isosteric analogue meso-oxa-Dap was utilized in place of meso-Dap to simplify the synthetic procedure. The more efficient synthesis involved ring opening of a peptide-embedded aziridine by a serine-based nucleophile. A branched tetrapeptide was also prepared as a mimic of the terminus of a crosslinked PG tetrapeptide. We used MS analysis to demonstrate that the tripeptide serves as a substrate for both Csd6 and Pgp2 and that the branched tetrapeptide serves as a substrate for Pgp2, albeit at a significantly slower rate.


Subject(s)
Alanine/analogs & derivatives , Aziridines , Diaminopimelic Acid/analogs & derivatives , Peptide Hydrolases/chemistry , Aziridines/chemical synthesis , Aziridines/chemistry , Campylobacter jejuni/enzymology , Helicobacter pylori/enzymology , Peptidoglycan/metabolism , Substrate Specificity
20.
Microb Pathog ; 128: 390-395, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30703473

ABSTRACT

Long term high-concentrate (HC) diet feeding induces subacute ruminal acidosis (SARA), which is reported to trigger a pro-inflammatory response. This study aimed to investigate the role of nucleotide-binding oligomerization domain protein 1 (NOD1) in initiating the pro-inflammatory response triggered by grain-induced SARA in the mammary gland of mid-lactating dairy cows. Twelve multiparous mid-lactating Holstein cows (455 ±â€¯28 kg) were randomly assigned into two groups to conduct the experiment for 18 weeks as follows: one group was fed a low-concentrate (LC) diet as a control (40% grain), and the other was fed an HC diet as a treatment (60% grain). Overall, the results showed that a decreased rumen pH and elevated γ-D-glutamyl-meso-diaminopimelic acid (iE-DAP) concentrations in the HC group compared with LC group. The concentration of pro-inflammatory cytokines, including interleukin (IL)-1ß, IL-6 and tumour necrosis factor-alpha (TNF-α), significantly increased in the lacteal vein of the HC group than LC group. The mRNA expression levels of NOD1, receptor-interacting protein2 (RIP2), NF-κBp65 (p65), IL-1ß, IL-6, IL-8 and TNF-α, which involved in inflammatory response, were up-regulated in the HC-induced mammary gland. The changes of the target proteins, including NOD1, p65 and pp65 presented the same tendency as those of the target genes. Collectively, long-term high concentrate feeding-induced SARA increased the rumen iE-DAP concentration which activated NOD1-NF-κB signalling pathway-dependent inflammation in the mammary gland of mid-lactating cows.


Subject(s)
Acidosis/veterinary , Animal Feed/adverse effects , Diet/adverse effects , Lactation/drug effects , Mammary Glands, Animal/drug effects , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Signal Transduction/drug effects , Acidosis/metabolism , Animals , Cattle , Cattle Diseases/metabolism , Cytokines/blood , Cytokines/genetics , Cytokines/metabolism , Diaminopimelic Acid/analogs & derivatives , Diaminopimelic Acid/metabolism , Diet/methods , Diet/veterinary , Female , Gastrointestinal Tract/chemistry , Gene Expression Regulation/drug effects , Humans , Hydrogen-Ion Concentration , Inflammation/chemically induced , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Mammary Glands, Animal/metabolism , Rumen/chemistry , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL